1101
|
Preventive Effects of Kaempferol on High-Fat Diet-Induced Obesity Complications in C57BL/6 Mice. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4532482. [PMID: 32337249 PMCID: PMC7166264 DOI: 10.1155/2020/4532482] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 03/16/2020] [Indexed: 02/08/2023]
Abstract
Kaempferol is a dietary flavanol that regulates cellular lipid and glucose metabolism. Its mechanism of action in preventing hepatic steatosis and obesity-related disorders has yet to be clarified. The purpose of this research was to examine kaempferol's antiobesity effects in high-fat diet- (HFD-) fed mice and to investigate its impact on their gut microbiota. Using a completely randomized design, 30 mice were equally assigned to a control group, receiving a low-fat diet, an HFD group, receiving a high-fat diet, and an HFD+kaempferol group, receiving a high-fat diet and kaempferol doses of 200 mg/kg in the diet. After eight weeks, the HFD mice displayed substantial body and liver weight gain and high blood glucose and serum cholesterol levels. However, treatment with kaempferol moderated body and liver weight gain and elevation of blood glucose and serum cholesterol and triglyceride levels. Examination of 16S ribosomal RNA showed that HFD mice exhibited decreased microbial diversity, but kaempferol treatment maintained it to nearly the same levels as those in the control group. In conclusion, kaempferol can protect against obesity and insulin resistance in mice on a high-fat diet, partly through regulating their gut microbiota and moderating the decrease in insulin resistance.
Collapse
|
1102
|
Wada N, Yamada H, Motoyama S, Saburi M, Sugimoto T, Kubota H, Miyawaki D, Wakana N, Kami D, Ogata T, Matoba S. Maternal high-fat diet exaggerates diet-induced insulin resistance in adult offspring by enhancing inflammasome activation through noncanonical pathway of caspase-11. Mol Metab 2020; 37:100988. [PMID: 32272237 PMCID: PMC7210595 DOI: 10.1016/j.molmet.2020.100988] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/22/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023] Open
Abstract
Objective Maternal high-fat diet (HFD) has been shown to promote the development of insulin resistance (IR) in adult offspring; however, the underlying mechanisms remain unclear. Methods Eight-week-old female wild-type mice (C57BL/6) were fed either an HFD or a normal diet (ND), one week prior to mating, and the diet was continued throughout gestation and lactation. Eight-week-old male offspring of both groups were fed an HFD for 8 weeks. Results Offspring of HFD-fed dams (O-HFD) exhibited significantly impaired insulin sensitivity compared with the offspring of ND-fed dams (O-ND). The adipocyte size of the eWAT increased significantly in O-HFD and was accompanied by abundant crown-like structures (CLSs), as well as a higher concentration of interleukin 1β (IL-1β) in the eWAT. Treatment with an inflammasome inhibitor, MCC950, completely abrogated the enhanced IR in O-HFD. However, ex vivo caspase-1 activity in eWAT revealed no difference between the two groups. In contrast, noncanonical inflammasome activation of caspase-11 was significantly augmented in O-HFD compared with O-ND, suggesting that membrane pore formation, but not cleavage of pro-IL-1β by caspase-1, is augmented in O-HFD. To examine the membrane pore formation, we performed metabolic activation of bone marrow-derived macrophages (BMDMs). The percentage of pore formation assessed by ethidium bromide staining was significantly higher in BMDMs of O-HFD, accompanied by an enhanced active caspase-11 expression. Consistently, the concentration of IL-1β in culture supernatants was significantly higher in the BMDMs from O-HFD than those from O-ND. Conclusions These findings demonstrate that maternal HFD exaggerates diet-induced IR in adult offspring by enhancing noncanonical caspase-11-mediated inflammasome activation. Maternal HFD increases the vulnerability to HFD-induced IR in adult offspring. Maternal HFD augments inflammasome activation in eWAT in adult offspring. Treatment with an inflammasome inhibitor abrogates IR in offspring of HFD-fed dam. Maternal HFD augments the noncanonical inflammasome activation pathway of caspase-11. Maternal HFD augments IL-1β release from BMDMs by enhancing membrane pore formation.
Collapse
Affiliation(s)
- Naotoshi Wada
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Yamada
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Shinichiro Motoyama
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Makoto Saburi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Sugimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Kubota
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Miyawaki
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Noriyuki Wakana
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
1103
|
Wahwah N, Kras KA, Roust LR, Katsanos CS. Subpopulation-specific differences in skeletal muscle mitochondria in humans with obesity: insights from studies employing acute nutritional and exercise stimuli. Am J Physiol Endocrinol Metab 2020; 318:E538-E553. [PMID: 31990577 DOI: 10.1152/ajpendo.00463.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mitochondria from skeletal muscle of humans with obesity often display alterations with respect to their morphology, proteome, biogenesis, and function. These changes in muscle mitochondria are considered to contribute to metabolic abnormalities observed in humans with obesity. Most of the evidence describing alterations in muscle mitochondria in humans with obesity, however, lacks reference to a specific subcellular location. This is despite data over the years showing differences in the morphology and function of subsarcolemmal (found near the plasma membrane) and intermyofibrillar (nested between the myofibrils) mitochondria in skeletal muscle. Recent studies reveal that impairments in mitochondrial function in obesity with respect to the subcellular location of the mitochondria in muscle are more readily evident following exposure of the skeletal muscle to physiological stimuli. In this review, we highlight the need to understand skeletal muscle mitochondria metabolism in obesity in a subpopulation-specific manner and in the presence of physiological stimuli that modify mitochondrial function in vivo. Experimental approaches employed under these conditions will allow for more precise characterization of impairments in skeletal muscle mitochondria and their implications in inducing metabolic dysfunction in human obesity.
Collapse
Affiliation(s)
- Nisreen Wahwah
- Center for Metabolic and Vascular Biology and School of Life Sciences, Arizona State University, Scottsdale, Arizona
| | - Katon A Kras
- Center for Metabolic and Vascular Biology and School of Life Sciences, Arizona State University, Scottsdale, Arizona
| | - Lori R Roust
- College of Medicine, Mayo Clinic in Arizona, Scottsdale, Arizona
| | - Christos S Katsanos
- Center for Metabolic and Vascular Biology and School of Life Sciences, Arizona State University, Scottsdale, Arizona
| |
Collapse
|
1104
|
Muddapu VR, Dharshini SAP, Chakravarthy VS, Gromiha MM. Neurodegenerative Diseases - Is Metabolic Deficiency the Root Cause? Front Neurosci 2020; 14:213. [PMID: 32296300 PMCID: PMC7137637 DOI: 10.3389/fnins.2020.00213] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/26/2020] [Indexed: 01/31/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer, Parkinson, Huntington, and amyotrophic lateral sclerosis, are a prominent class of neurological diseases currently without a cure. They are characterized by an inexorable loss of a specific type of neurons. The selective vulnerability of specific neuronal clusters (typically a subcortical cluster) in the early stages, followed by the spread of the disease to higher cortical areas, is a typical pattern of disease progression. Neurodegenerative diseases share a range of molecular and cellular pathologies, including protein aggregation, mitochondrial dysfunction, glutamate toxicity, calcium load, proteolytic stress, oxidative stress, neuroinflammation, and aging, which contribute to neuronal death. Efforts to treat these diseases are often limited by the fact that they tend to address any one of the above pathological changes while ignoring others. Lack of clarity regarding a possible root cause that underlies all the above pathologies poses a significant challenge. In search of an integrative theory for neurodegenerative pathology, we hypothesize that metabolic deficiency in certain vulnerable neuronal clusters is the common underlying thread that links many dimensions of the disease. The current review aims to present an outline of such an integrative theory. We present a new perspective of neurodegenerative diseases as metabolic disorders at molecular, cellular, and systems levels. This helps to understand a common underlying mechanism of the many facets of the disease and may lead to more promising disease-modifying therapeutic interventions. Here, we briefly discuss the selective metabolic vulnerability of specific neuronal clusters and also the involvement of glia and vascular dysfunctions. Any failure in satisfaction of the metabolic demand by the neurons triggers a chain of events that precipitate various manifestations of neurodegenerative pathology.
Collapse
Affiliation(s)
- Vignayanandam Ravindernath Muddapu
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - S. Akila Parvathy Dharshini
- Protein Bioinformatics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - V. Srinivasa Chakravarthy
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - M. Michael Gromiha
- Protein Bioinformatics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
1105
|
Hjelmgren O, Gummesson A, Bergström G, Schmidt C. Beta-Cell Function, Self-rated Health, and Lifestyle Habits in 64-Year-Old Swedish Women with Metabolically Healthy Obesity Phenotype. J Obes Metab Syndr 2020; 29:39-46. [PMID: 32200605 PMCID: PMC7117998 DOI: 10.7570/jomes19078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/08/2020] [Accepted: 02/29/2020] [Indexed: 01/11/2023] Open
Abstract
Background A subset of obese individuals do not present metabolic abnormalities that commonly define the metabolic syndrome (MetS). This is referred to as a metabolically healthy obese (MHO) phenotype. The aim of the present study was to evaluate the prevalence of the MHO phenotype and its relationship with beta cell dysfunction by measuring C-peptide and proinsulin, anthropometric-, metabolic- and lipid appearance, as well as lifestyle behaviors and self-rated health in a cohort of 64-year-old Swedish women. Methods The National Cholesterol Education Program definition was used to assess MetS. We defined normal weight as body mass index (BMI) 18.5–24.9 kg/m2 and obesity as BMI ≥30 kg/m2 to categorize participants as metabolically healthy normal weight, MHO, and metabolically unhealthy obese. Results The MHO phenotype represented 36.3% of obese participants and 16.3% of total participants. The MHO group were at greater risk of having proinsulin levels >11 pmol/L, indicating impaired beta cell function. Further, homeostatic model assessment for insulin resistance, fasting plasma levels of insulin, and C-peptide showed significant trends, with the MHO phenotype group having intermediate levels among three groups. Health behaviors such as leisure time physical activity and alcohol intake were also intermediate in individuals with the MHO phenotype. Conclusion In this study, we demonstrate that over a third of the obese women in our sample were MHO. Further, women with the MHO phenotype showed intermediate profiles considering beta cell function and insulin resistance, as well as metabolic variables, and tended to rate their general health as worse than otherwise similar individuals of normal weight.
Collapse
Affiliation(s)
- Ola Hjelmgren
- Department of Clinical Physiology, Institution of Neuroscience and Physiology, Sahlgrenska Academy at Gothenburg University, Göteborg, Sweden
| | - Anders Gummesson
- Department of Molecular and Clinical Medicine, Institution of Medicine, Sahlgrenska Academy at Gothenburg University, Göteborg, Sweden
| | - Göran Bergström
- Department of Clinical Physiology, Institution of Neuroscience and Physiology, Sahlgrenska Academy at Gothenburg University, Göteborg, Sweden.,Department of Molecular and Clinical Medicine, Institution of Medicine, Sahlgrenska Academy at Gothenburg University, Göteborg, Sweden
| | - Caroline Schmidt
- Department of Molecular and Clinical Medicine, Institution of Medicine, Sahlgrenska Academy at Gothenburg University, Göteborg, Sweden
| |
Collapse
|
1106
|
Guglielmo P, Ekström S, Strand R, Visvanathar R, Malmberg F, Johansson E, Pereira MJ, Skrtic S, Carlsson BCL, Eriksson JW, Ahlström H, Kullberg J. Validation of automated whole-body analysis of metabolic and morphological parameters from an integrated FDG-PET/MRI acquisition. Sci Rep 2020; 10:5331. [PMID: 32210327 PMCID: PMC7093440 DOI: 10.1038/s41598-020-62353-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/11/2020] [Indexed: 11/09/2022] Open
Abstract
Automated quantification of tissue morphology and tracer uptake in PET/MR images could streamline the analysis compared to traditional manual methods. To validate a single atlas image segmentation approach for automated assessment of tissue volume, fat content (FF) and glucose uptake (GU) from whole-body [18F]FDG-PET/MR images. Twelve subjects underwent whole-body [18F]FDG-PET/MRI during hyperinsulinemic-euglycemic clamp. Automated analysis of tissue volumes, FF and GU were achieved using image registration to a single atlas image with reference segmentations of 18 volume of interests (VOIs). Manual segmentations by an experienced radiologist were used as reference. Quantification accuracy was assessed with Dice scores, group comparisons and correlations. VOI Dice scores ranged from 0.93 to 0.32. Muscles, brain, VAT and liver showed the highest scores. Pancreas, large and small intestines demonstrated lower segmentation accuracy and poor correlations. Estimated tissue volumes differed significantly in 8 cases. Tissue FFs were often slightly but significantly overestimated. Satisfactory agreements were observed in most tissue GUs. Automated tissue identification and characterization using a single atlas segmentation performs well compared to manual segmentation in most tissues and will be valuable in future studies. In certain tissues, alternative quantification methods or improvements to the current approach is needed.
Collapse
Affiliation(s)
- P Guglielmo
- Section of Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
- University of Milan Bicocca, Milan, Italy.
| | - S Ekström
- Section of Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - R Strand
- Section of Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - R Visvanathar
- Section of Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - F Malmberg
- Section of Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - E Johansson
- Section of Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- GE Healthcare, Chicago, USA
| | - M J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - S Skrtic
- Pharmaceutical Technology & Development, AstraZeneca AB, Gothenburg, Sweden
- Department of Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - B C L Carlsson
- Early Clinical Development, Cardiovascular, Renal & Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - J W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - H Ahlström
- Section of Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Antaros Medical, Mölndal, Sweden
| | - J Kullberg
- Section of Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Antaros Medical, Mölndal, Sweden
| |
Collapse
|
1107
|
Adiponectin and Cognitive Decline. Int J Mol Sci 2020; 21:ijms21062010. [PMID: 32188008 PMCID: PMC7139651 DOI: 10.3390/ijms21062010] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/10/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022] Open
Abstract
Adiponectin (ADPN) is a plasma protein secreted by adipose tissue showing pleiotropic effects with anti-diabetic, anti-atherogenic, and anti-inflammatory properties. Initially, it was thought that the main role was only the metabolism control. Later, ADPN receptors were also found in the central nervous system (CNS). In fact, the receptors AdipoR1 and AdipoR2 are expressed in various areas of the brain, including the hypothalamus, hippocampus, and cortex. While AdipoR1 regulates insulin sensitivity through the activation of the AMP-activated protein kinase (AMPK) pathway, AdipoR2 stimulates the neural plasticity through the activation of the peroxisome proliferator-activated receptor alpha (PPARα) pathway that inhibits inflammation and oxidative stress. Overall, based on its central and peripheral actions, ADPN appears to have neuroprotective effects by reducing inflammatory markers, such as C-reactive protein (PCR), interleukin 6 (IL6), and Tumor Necrosis Factor a (TNFa). Conversely, high levels of inflammatory cascade factors appear to inhibit the production of ADPN, suggesting bidirectional modulation. In addition, ADPN appears to have insulin-sensitizing action. It is known that a reduction in insulin signaling is associated with cognitive impairment. Based on this, it is of great interest to investigate the mechanism of restoration of the insulin signal in the brain as an action of ADPN, because it is useful for testing a possible pharmacological treatment for the improvement of cognitive decline. Anyway, if ADPN regulates neuronal functioning and cognitive performances by the glycemic metabolic system remains poorly explored. Moreover, although the mechanism is still unclear, women compared to men have a doubled risk of developing cognitive decline. Several studies have also supported that during the menopausal transition, the estrogen reduction can adversely affect the brain, in particular, verbal memory and verbal fluency. During the postmenopausal period, in obese and insulin-resistant individuals, ADPN serum levels are significantly reduced. Our recent study has evaluated the relationship between plasma ADPN levels and cognitive performances in menopausal women. Thus, the aim of this review is to summarize both the mechanisms and the effects of ADPN in the central nervous system and the relationship between plasma ADPN levels and cognitive performances, also in menopausal women.
Collapse
|
1108
|
Huth C, Bauer A, Zierer A, Sudduth-Klinger J, Meisinger C, Roden M, Peters A, Koenig W, Herder C, Thorand B. Biomarker-defined pathways for incident type 2 diabetes and coronary heart disease-a comparison in the MONICA/KORA study. Cardiovasc Diabetol 2020; 19:32. [PMID: 32164753 PMCID: PMC7066738 DOI: 10.1186/s12933-020-01003-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
Background Biomarkers may contribute to our understanding of the pathophysiology of various diseases. Type 2 diabetes (T2D) and coronary heart disease (CHD) share many clinical and lifestyle risk factors and several biomarkers are associated with both diseases. The current analysis aims to assess the relevance of biomarkers combined to pathway groups for the development of T2D and CHD in the same cohort. Methods Forty-seven serum biomarkers were measured in the MONICA/KORA case-cohort study using clinical chemistry assays and ultrasensitive molecular counting technology. The T2D (CHD) analyses included 689 (568) incident cases and 1850 (2004) non-cases from three population-based surveys. At baseline, the study participants were 35–74 years old. The median follow-up was 14 years. We computed Cox regression models for each biomarker, adjusted for age, sex, and survey. Additionally, we assigned the biomarkers to 19 etiological pathways based on information from literature. One age-, sex-, and survey-controlled average variable was built for each pathway. We used the R2PM coefficient of determination to assess the explained disease risk. Results The associations of many biomarkers, such as several cytokines or the iron marker soluble transferrin receptor (sTfR), were similar in strength for T2D and CHD, but we also observed important differences. Lipoprotein (a) (Lp(a)) and N-terminal pro B-type natriuretic peptide (NT-proBNP) even demonstrated opposite effect directions. All pathway variables together explained 49% of the T2D risk and 21% of the CHD risk. The insulin-like growth factor binding protein 2 (IGFBP-2, IGF/IGFBP system pathway) best explained the T2D risk (about 9% explained risk, independent of all other pathway variables). For CHD, the myocardial-injury- and lipid-related-pathways were most important and both explained about 4% of the CHD risk. Conclusions The biomarker-derived pathway variables explained a higher proportion of the T2D risk compared to CHD. The ranking of the pathways differed between the two diseases, with the IGF/IGFBP-system-pathway being most strongly associated with T2D and the myocardial-injury- and lipid-related-pathways with CHD. Our results help to better understand the pathophysiology of the two diseases, with the ultimate goal of pointing out targets for lifestyle intervention and drug development to ideally prevent both T2D and CHD development.
Collapse
Affiliation(s)
- Cornelia Huth
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany. .,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| | - Alina Bauer
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Astrid Zierer
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | | | - Christa Meisinger
- Chair of Epidemiology, Ludwig-Maximilians-Universität München, UNIKA-T Augsburg, Augsburg, Germany.,Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Wolfgang Koenig
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany.,Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Christian Herder
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
1109
|
Ma X, Dong L, Shao Q, Cheng Y, Lv S, Sun Y, Shen H, Wang Z, Zhou Y, Liu X. Triglyceride glucose index for predicting cardiovascular outcomes after percutaneous coronary intervention in patients with type 2 diabetes mellitus and acute coronary syndrome. Cardiovasc Diabetol 2020; 19:31. [PMID: 32156279 PMCID: PMC7063826 DOI: 10.1186/s12933-020-01006-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/24/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The triglyceride glucose (TyG) index, a simple surrogate estimate of insulin resistance, has been demonstrated to predict cardiovascular (CV) disease morbidity and mortality in the general population and many patient cohorts. However, to our knowledge, the prognostic usefulness of the TyG index after percutaneous coronary intervention (PCI) in patients with type 2 diabetes mellitus (T2DM) and acute coronary syndrome (ACS) has not been determined. This study aimed to evaluate the association of the TyG index with adverse CV outcomes in patients with T2DM and ACS who underwent PCI. METHODS The TyG index was calculated using the formula ln[fasting triglycerides (mg/dL) × fasting glucose (mg/dL)/2]. The primary endpoint was the composite of all-cause mortality, non-fatal stroke, non-fatal myocardial infarction, or unplanned repeat revascularization. The association between the TyG index and adverse CV outcomes was assessed by Cox proportional hazards regression analysis. RESULTS In total, 776 patients with T2DM and ACS who underwent PCI (mean age, 61 ± 10 years; men, 72.2%) were included in the final analysis. Over a median follow-up of 30 months, 188 patients (24.2%) had at least 1 primary endpoint event. The follow-up incidence of the primary endpoint rose with increasing TyG index tertiles. The multivariate Cox proportional hazards regression analysis adjusted for multiple confounders revealed a hazard ratio for the primary endpoint of 2.17 (95% CI 1.45-3.24; P for trend = 0.001) when the highest and lowest TyG index tertiles were compared. CONCLUSIONS The TyG index was significantly and positively associated with adverse CV outcomes, suggesting that the TyG index may be a valuable predictor of adverse CV outcomes after PCI in patients with T2DM and ACS.
Collapse
Affiliation(s)
- Xiaoteng Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China
| | - Lisha Dong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China
| | - Qiaoyu Shao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China
| | - Yujing Cheng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China
| | - Sai Lv
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China
| | - Yan Sun
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China
| | - Hua Shen
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China
| | - Zhijian Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China
| | - Xiaoli Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, 100029, China.
| |
Collapse
|
1110
|
Canonical Correlation between Behavioral-Psychological Variables and Predictors of Coronary Artery Disease Prognosis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17051608. [PMID: 32131511 PMCID: PMC7084809 DOI: 10.3390/ijerph17051608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/06/2020] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
Metabolic syndrome (MetS) and severity of coronary artery disease (CAD) are considered predictors of CAD prognosis. Unhealthy lifestyles and type-D personality are associated with MetS and are potential causes of primary and secondary CAD. In this cross-sectional descriptive study, we aimed to investigate the relationship between behavioral-psychological variables and predictors of CAD prognosis. The behavioral-psychological variable set contained six lifestyle categories and two type-D personality categories. Descriptive analyses, t-tests, analysis of variance, Pearson’s correlation, and canonical correlation were used. The behavioral-psychological variable set was related to the predictor set for CAD prognosis, with a significant canonical variate of 0.67 (45% overlapping variance). Significant pairs of canonical variates indicated that poor physical activity and weight control (−0.77), poor dietary habits (−0.78), alcohol consumption and cigarette smoking (−0.37), lack of sleep and rest (−0.40), stress (−0.64) in the lifestyle set, higher negative affectivity (0.52), and social inhibition (0.71) in the type-D personality set were associated with a high MetS score (0.59) and severity of CAD (0.91). A combination of behavioral and psychological variables was found to be important in predicting the prognosis of CAD; therefore, interventions aimed at preventing combinations of these variables may be effective in improving CAD prognosis.
Collapse
|
1111
|
McCrimmon RJ, Catarig AM, Frias JP, Lausvig NL, le Roux CW, Thielke D, Lingvay I. Effects of once-weekly semaglutide vs once-daily canagliflozin on body composition in type 2 diabetes: a substudy of the SUSTAIN 8 randomised controlled clinical trial. Diabetologia 2020; 63:473-485. [PMID: 31897524 PMCID: PMC6997246 DOI: 10.1007/s00125-019-05065-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Intra-abdominal or visceral obesity is associated with insulin resistance and an increased risk for cardiovascular disease. This study aimed to compare the effects of semaglutide 1.0 mg and canagliflozin 300 mg on body composition in a subset of participants from the SUSTAIN 8 Phase IIIB, randomised double-blind trial who underwent whole-body dual-energy x-ray absorptiometry (DXA) scanning. METHODS Adults (age ≥18 years) with type 2 diabetes, HbA1c 53-91 mmol/mol (7.0-10.5%), on a stable daily dose of metformin (≥1500 mg or maximum tolerated dose) and with an eGFR ≥60 ml min-1 [1.73 m]-2 were randomised 1:1 to semaglutide 1.0 mg once weekly and canagliflozin placebo once daily, or canagliflozin 300 mg once daily and semaglutide placebo once weekly. Body composition was assessed using whole-body DXA scans. The study participants and investigator remained blinded throughout the trial, and quality of DXA scans was evaluated in a blinded manner. Change from baseline to week 52 in total fat mass (kg) was the confirmatory efficacy endpoint. RESULTS A subset of 178 participants (semaglutide, n = 88; canagliflozin, n = 90) underwent DXA scanning at screening and were randomised into the substudy. Of these, 114 (semaglutide, n = 53; canagliflozin, n = 61) participants had observed end-of-treatment data included in the confirmatory efficacy analysis. Of the 178 participants in the substudy, numerical improvements in body composition (including fat mass, lean mass and visceral fat mass) were observed after 52 weeks with both treatments. Total fat mass (baseline 33.2 kg) was reduced by 3.4 kg and 2.6 kg with semaglutide and canagliflozin, respectively (estimated treatment difference: -0.79 [95% CI -2.10, 0.51]). Although total lean mass (baseline 51.3 kg) was also reduced by 2.3 kg and 1.5 kg with semaglutide and canagliflozin, respectively (estimated treatment difference: -0.78 [-1.61, 0.04]), the proportion of lean mass (baseline 59.4%) increased by 1.2%- and 1.1%-point, respectively (estimated treatment difference 0.14 [-0.89, 1.17]). Changes in visceral fat mass and overall changes in body composition (assessed by the fat to lean mass ratio) were comparable between the two treatment groups. CONCLUSIONS/INTERPRETATION In individuals with uncontrolled type 2 diabetes on stable-dose metformin therapy, the changes in body composition with semaglutide and canagliflozin were not significantly different. Although numerical improvements in body composition were observed following treatment in both treatment arms, the specific impact of both treatments on body composition in the absence of a placebo arm is speculative at this stage. TRIAL REGISTRATION ClinicalTrials.gov NCT03136484. FUNDING This trial was supported by Novo Nordisk A/S, Denmark.
Collapse
Affiliation(s)
- Rory J McCrimmon
- School of Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK.
| | | | - Juan P Frias
- National Research Institute, Los Angeles, CA, USA
| | | | - Carel W le Roux
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | | | - Ildiko Lingvay
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
1112
|
Demyanets S, Kaun C, Kaider A, Speidl W, Prager M, Oravec S, Hohensinner P, Wojta J, Rega-Kaun G. The pro-inflammatory marker soluble suppression of tumorigenicity-2 (ST2) is reduced especially in diabetic morbidly obese patients undergoing bariatric surgery. Cardiovasc Diabetol 2020; 19:26. [PMID: 32101157 PMCID: PMC7045735 DOI: 10.1186/s12933-020-01001-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND High soluble suppression of tumorigenicity-2 (sST2) is a marker of poor prognosis in chronic inflammatory conditions. ST2 and its ligand interleukin (IL)-33 are elevated in adipose tissue of obese individuals. We aimed to evaluate circulating sST2 and IL-33 as possible markers of metabolic benefit in morbidly overweight patients after Roux-en-Y gastric bypass (RYGB) bariatric surgery. METHODS sST2, IL-33, high sensitive IL-6, high sensitive C-reactive protein (hsCRP), leptin, cholesterol metabolism and liver parameters were measured in 80 morbidly obese individuals before and 1 year after bariatric surgery. RESULTS sST2 was higher (P = 0.03) in diabetics as compared to individuals without diabetes. Baseline sST2 was also higher in males than in females (P= 0.0002). One year after bariatric surgery, sST2 levels were decreased (median 120, IQR 59-176 pg/mL) as compared to sST2 before surgery (median 141, IQR 111-181, P = 0.0024), and the diabetic group showed most pronounced reduction in sST2 (P = 0.0016). An association was found between sST2 and liver function parameters before and after bariatric surgery, and between baseline sST2 and total cholesterol, triglyceride, total low density lipoprotein (LDL), small dense LDL, Apolipoprotein B as well as with small dense high density lipoproteins (HDL). In the subgroup of diabetic patients positive correlation between IL-33 and sST2 (r = 0.44, P = 0.05) was noticed. CONCLUSIONS Circulating sST2 is associated with markers of liver functions and lipid metabolism in severely obese patients and a reduction of sST2 was shown after successful bariatric surgery, most prominently in diabetic patients.
Collapse
Affiliation(s)
- Svitlana Demyanets
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christoph Kaun
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Alexandra Kaider
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Walter Speidl
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Manfred Prager
- Department of Surgery, Hospital Hietzing, Wolkersbergenstraße 1, 1130, Vienna, Austria
| | - Stanislav Oravec
- Krankenanstalten Dr. Dostal, Saarplatz 9, 1190, Vienna, Austria
- 2nd Department of Internal Medicine, Faculty of Medicine, Comenius University, Šafárikovo námestie 6, 814 99, Bratislava 1, Slovakia
| | - Philipp Hohensinner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Core Facilities, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Ludwig Boltzmann Institute for Cardiovascular Research, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Gersina Rega-Kaun
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- 5th Medical Department, Wilhelminenhospital, Montleartstraße 37, 1160, Vienna, Austria
| |
Collapse
|
1113
|
Wang S, Wong LY, Neumann D, Liu Y, Sun A, Antoons G, Strzelecka A, Glatz JF, Nabben M, Luiken JJ. Augmenting Vacuolar H +-ATPase Function Prevents Cardiomyocytes from Lipid-Overload Induced Dysfunction. Int J Mol Sci 2020; 21:ijms21041520. [PMID: 32102213 PMCID: PMC7073192 DOI: 10.3390/ijms21041520] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/17/2020] [Accepted: 02/21/2020] [Indexed: 12/14/2022] Open
Abstract
The diabetic heart is characterized by a shift in substrate utilization from glucose to lipids, which may ultimately lead to contractile dysfunction. This substrate shift is facilitated by increased translocation of lipid transporter CD36 (SR-B2) from endosomes to the sarcolemma resulting in increased lipid uptake. We previously showed that endosomal retention of CD36 is dependent on the proper functioning of vacuolar H+-ATPase (v-ATPase). Excess lipids trigger CD36 translocation through inhibition of v-ATPase function. Conversely, in yeast, glucose availability is known to enhance v-ATPase function, allowing us to hypothesize that glucose availability, via v-ATPase, may internalize CD36 and restore contractile function in lipid-overloaded cardiomyocytes. Increased glucose availability was achieved through (a) high glucose (25 mM) addition to the culture medium or (b) adenoviral overexpression of protein kinase-D1 (a kinase mediating GLUT4 translocation). In HL-1 cardiomyocytes, adult rat and human cardiomyocytes cultured under high-lipid conditions, each treatment stimulated v-ATPase re-assembly, endosomal acidification, endosomal CD36 retention and prevented myocellular lipid accumulation. Additionally, these treatments preserved insulin-stimulated GLUT4 translocation and glucose uptake as well as contractile force. The present findings reveal v-ATPase functions as a key regulator of cardiomyocyte substrate preference and as a novel potential treatment approach for the diabetic heart.
Collapse
Affiliation(s)
- Shujin Wang
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (S.W.); (L.-Y.W.); (Y.L.); (A.S.); (A.S.); (M.N.)
| | - Li-Yen Wong
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (S.W.); (L.-Y.W.); (Y.L.); (A.S.); (A.S.); (M.N.)
- Department of Clinical Genetics, Maastricht University Medical Center+, 6200-MD Maastricht, The Netherlands
| | - Dietbert Neumann
- Departments of Pathology, CARIM School for Cardiovascular Diseases, Maastricht University, 6200-MD Maastricht, The Netherlands;
| | - Yilin Liu
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (S.W.); (L.-Y.W.); (Y.L.); (A.S.); (A.S.); (M.N.)
| | - Aomin Sun
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (S.W.); (L.-Y.W.); (Y.L.); (A.S.); (A.S.); (M.N.)
| | - Gudrun Antoons
- Departments of Physiology, CARIM School for Cardiovascular Diseases, Maastricht University, 6200-MD Maastricht, The Netherlands;
| | - Agnieszka Strzelecka
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (S.W.); (L.-Y.W.); (Y.L.); (A.S.); (A.S.); (M.N.)
| | - Jan F.C. Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (S.W.); (L.-Y.W.); (Y.L.); (A.S.); (A.S.); (M.N.)
- Department of Clinical Genetics, Maastricht University Medical Center+, 6200-MD Maastricht, The Netherlands
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (S.W.); (L.-Y.W.); (Y.L.); (A.S.); (A.S.); (M.N.)
| | - Joost J.F.P. Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (S.W.); (L.-Y.W.); (Y.L.); (A.S.); (A.S.); (M.N.)
- Correspondence: ; Tel.: +31-43 3881209
| |
Collapse
|
1114
|
Souza LM, Mendonça CL, Assis RN, Oliveira Filho EF, Soares GSL, Souto RJC, Soares PC, Afonso JAB. Changes in cardiac biomarkers in goats naturally affected by pregnancy toxemia. Res Vet Sci 2020; 130:73-78. [PMID: 32146378 DOI: 10.1016/j.rvsc.2020.02.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 01/25/2023]
Abstract
Pregnancy toxemia (PT) is considered one of the most common metabolic diseases with high impact on the production of small ruminants. The objective of this study was investigate possible myocardial damage in goats affected with PT by the determination of serum myocardial biomarkers CK-MB and cTnI. A total of 44 goats affected with PT, and 10 apparently healthy goats (control group or CG) were used in the study. In goats with PT, the serum concentrations of cTnI (0.43 ng/mL) were significantly higher than that in CG goats (0.06 ng/mL). Although CK-MB showed no significant difference, it was approximately three times higher in animals with PT. The serum concentrations of insulin were significantly lower in PT goats (5.03 ppmol/L) compared to CG goats (10.66 pmol/L). The serum concentrations of cortisol in PT goats (155.41 nmol/L) were significantly higher than that in CG goats (36.58 nmol/L). Results of this study indicate that a clinically significant myocardial damage might occur in goats affected with PT leading to significant elevations in values of cTnI and CK-MB. Therefore, these parameters could be used as a potential prognostic indicator in goats affected with this important disease.
Collapse
Affiliation(s)
- L M Souza
- Federal Rural University of Pernambuco (UAG/UFRPE), Postgraduate Program in Sanitation and Reproduction of Ruminants, Av. Bom Pastor, s/n, CP 152-Boa Vista, 55292-278 Garanhuns, Pernambuco, Brazil.
| | - C L Mendonça
- Cattle Clinic, Campus Garanhuns/UFRPE, Av. Bom Pastor, s/n, CP 152-Boa Vista, 55292-278 Garanhuns, Pernambuco, Brazil
| | - R N Assis
- Federal Rural University of Pernambuco (UAG/UFRPE), Postgraduate Program in Sanitation and Reproduction of Ruminants, Av. Bom Pastor, s/n, CP 152-Boa Vista, 55292-278 Garanhuns, Pernambuco, Brazil
| | - E F Oliveira Filho
- Federal Rural University of Pernambuco, Postgraduate Program in Veterinary Medicine, Campus Recife/UFRPE, Av. Manuel de Medeiros, s/n-Dois Irmãos, 52171-900 Recife, Pernambuco, Brazil
| | - G S L Soares
- Federal Rural University of Pernambuco, Postgraduate Program in Veterinary Medicine, Campus Recife/UFRPE, Av. Manuel de Medeiros, s/n-Dois Irmãos, 52171-900 Recife, Pernambuco, Brazil
| | - R J C Souto
- Cattle Clinic, Campus Garanhuns/UFRPE, Av. Bom Pastor, s/n, CP 152-Boa Vista, 55292-278 Garanhuns, Pernambuco, Brazil
| | - P C Soares
- Department of Veterinary Medicine, Campus Recife/UFRPE, Av. Manuel de Medeiros, s/n-Dois Irmãos, 52171-900 Recife, Pernambuco, Brazil
| | - J A B Afonso
- Cattle Clinic, Campus Garanhuns/UFRPE, Av. Bom Pastor, s/n, CP 152-Boa Vista, 55292-278 Garanhuns, Pernambuco, Brazil
| |
Collapse
|
1115
|
Kreienkamp R, Gonzalo S. Metabolic Dysfunction in Hutchinson-Gilford Progeria Syndrome. Cells 2020; 9:cells9020395. [PMID: 32046343 PMCID: PMC7072593 DOI: 10.3390/cells9020395] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/22/2022] Open
Abstract
Hutchinson–Gilford Progeria Syndrome (HGPS) is a segmental premature aging disease causing patient death by early teenage years from cardiovascular dysfunction. Although HGPS does not totally recapitulate normal aging, it does harbor many similarities to the normal aging process, with patients also developing cardiovascular disease, alopecia, bone and joint abnormalities, and adipose changes. It is unsurprising, then, that as physicians and scientists have searched for treatments for HGPS, they have targeted many pathways known to be involved in normal aging, including inflammation, DNA damage, epigenetic changes, and stem cell exhaustion. Although less studied at a mechanistic level, severe metabolic problems are observed in HGPS patients. Interestingly, new research in animal models of HGPS has demonstrated impressive lifespan improvements secondary to metabolic interventions. As such, further understanding metabolism, its contribution to HGPS, and its therapeutic potential has far-reaching ramifications for this disease still lacking a robust treatment strategy.
Collapse
Affiliation(s)
- Ray Kreienkamp
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
- Department of Pediatrics Residency, Washington University Medical School, St. Louis, MO 63105, USA;
| | - Susana Gonzalo
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
- Correspondence: ; Tel.: +1-314-977-9244
| |
Collapse
|
1116
|
Wedervang-Resell K, Friis S, Lonning V, Smelror RE, Johannessen C, Agartz I, Ulven SM, Holven KB, Andreassen OA, Myhre AM. Lipid alterations in adolescents with early-onset psychosis may be independent of antipsychotic medication. Schizophr Res 2020; 216:295-301. [PMID: 31791814 DOI: 10.1016/j.schres.2019.11.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/21/2019] [Accepted: 11/22/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Dyslipidemia and insulin resistance (HOMA-IR) are cardiovascular risk factors prevalent in patients with psychosis. Whether these factors are intrinsic or affected by lifestyle or antipsychotic medication (AP) is unclear. Therefore, we investigated lipid profiles, HOMA-IR, and psychotic phenotypes in patients aged 12-18 years with early-onset psychosis (EOP) with and without AP exposure. METHOD We measured fasting total cholesterol (TC), high-density lipoprotein cholesterol (HDLC), triglycerides (TG), insulin, and glucose in patients with EOP (n = 39) and healthy controls (HC) (n = 66). Diet information was not available. Negative symptoms were assessed using the Positive and Negative Syndrome Scale (PANSS). We used univariate analysis of variance to compare TC/HDL-C ratios and TG and HOMA-IR values, controlling for body mass index (BMI) and AP exposure. We assessed the explained variance of having EOP using multiple regression analysis. RESULTS Patients with and without AP exposure had significantly higher TC/HDL-C (p = 0.003, p = 0.029) and TG values (p < 0.001, p = 0.021) than HC. Significantly increased HOMA-IR scores were found only in AP-exposed patients (p = 0.037). EOP significantly increased the explained variance for TC/HDL-C and TG, but not for HOMA-IR. Patients with a PANSS negative score > 21 had significantly higher levels of TG than those with low scores (p = 0.032). CONCLUSION Our results suggest that lipid alterations predate AP treatment in adolescents with EOP. Higher levels of negative symptoms and AP further increase metabolic risk. The preliminary findings propose that subclinical dyslipidemia may be intrinsic to EOP.
Collapse
Affiliation(s)
- Kirsten Wedervang-Resell
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Department of Psychiatric Research and Development, Oslo University Hospital, Oslo, Norway.
| | - Svein Friis
- Division of Mental Health and Addiction, Department of Psychiatric Research and Development, Oslo University Hospital, Oslo, Norway
| | - Vera Lonning
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Runar E Smelror
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Cecilie Johannessen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Ingrid Agartz
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Centre for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Stine M Ulven
- Department of Nutrition, Institute for Basic Medical Sciences, University of Oslo, PO Box 1046, 0317 Blindern, Oslo, Norway
| | - Kirsten B Holven
- Department of Nutrition, Institute for Basic Medical Sciences, University of Oslo, PO Box 1046, 0317 Blindern, Oslo, Norway
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne M Myhre
- Division of Mental Health and Addiction, Department of Psychiatric Research and Development, Oslo University Hospital, Oslo, Norway; Child and Adolescent Psychiatry Unit, Division of Mental Health and Addiction, Institute of clinical Medicine, University of Oslo, Norway
| |
Collapse
|
1117
|
Zatterale F, Longo M, Naderi J, Raciti GA, Desiderio A, Miele C, Beguinot F. Chronic Adipose Tissue Inflammation Linking Obesity to Insulin Resistance and Type 2 Diabetes. Front Physiol 2020; 10:1607. [PMID: 32063863 PMCID: PMC7000657 DOI: 10.3389/fphys.2019.01607] [Citation(s) in RCA: 594] [Impact Index Per Article: 118.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity is one of the major health burdens of the 21st century as it contributes to the growing prevalence of its related comorbidities, including insulin resistance and type 2 diabetes. Growing evidence suggests a critical role for overnutrition in the development of low-grade inflammation. Specifically, chronic inflammation in adipose tissue is considered a crucial risk factor for the development of insulin resistance and type 2 diabetes in obese individuals. The triggers for adipose tissue inflammation are still poorly defined. However, obesity-induced adipose tissue expansion provides a plethora of intrinsic signals (e.g., adipocyte death, hypoxia, and mechanical stress) capable of initiating the inflammatory response. Immune dysregulation in adipose tissue of obese subjects results in a chronic low-grade inflammation characterized by increased infiltration and activation of innate and adaptive immune cells. Macrophages are the most abundant innate immune cells infiltrating and accumulating into adipose tissue of obese individuals; they constitute up to 40% of all adipose tissue cells in obesity. In obesity, adipose tissue macrophages are polarized into pro-inflammatory M1 macrophages and secrete many pro-inflammatory cytokines capable of impairing insulin signaling, therefore promoting the progression of insulin resistance. Besides macrophages, many other immune cells (e.g., dendritic cells, mast cells, neutrophils, B cells, and T cells) reside in adipose tissue during obesity, playing a key role in the development of adipose tissue inflammation and insulin resistance. The association of obesity, adipose tissue inflammation, and metabolic diseases makes inflammatory pathways an appealing target for the treatment of obesity-related metabolic complications. In this review, we summarize the molecular mechanisms responsible for the obesity-induced adipose tissue inflammation and progression toward obesity-associated comorbidities and highlight the current therapeutic strategies.
Collapse
Affiliation(s)
- Federica Zatterale
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Michele Longo
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Jamal Naderi
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Environmental, Biological, and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Gregory Alexander Raciti
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Antonella Desiderio
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.,URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| |
Collapse
|
1118
|
Li RJ, Liu Y, Liu HQ, Li J. Ketogenic diets and protective mechanisms in epilepsy, metabolic disorders, cancer, neuronal loss, and muscle and nerve degeneration. J Food Biochem 2020; 44:e13140. [PMID: 31943235 DOI: 10.1111/jfbc.13140] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022]
Abstract
Ketogenic diet (KD), the "High-fat, low-carbohydrate, adequate-protein" diet strategy, replacing glucose with ketone bodies, is effective against several diseases, from intractable epileptic seizures, metabolic disorders, tumors, autosomal dominant polycystic kidney disease, and neurodegeneration to skeletal muscle atrophy and peripheral neuropathy. Key mechanisms include augmented mitochondrial efficiency, reduced oxidative stress, and regulated phospho-AMP-activated protein kinase, gamma-aminobutyric acid-glutamate, Na+/ K+ pump, leptin and adiponectin levels, ghrelin levels, lipogenesis, ketogenesis, lipolysis, and gluconeogenesis. In cancer cells, KD targets glucose metabolism, suppresses insulin-like growth factor-1 and PI3K/AKT/mTOR pathways, and reduces cancer cachexia and muscle waste and fatigue. An associated increased skeletal proliferator-activated receptor-γ coactivator-1α activity alters systemic ketone body homeostasis, contributing toward attenuated diabetic hyperketonemia. Antioxidative and anti-inflammatory properties enable KD enhance endurance and sports performances while preventing exercise-induced muscle and organ debility. KD reduces metabolic syndromes-associated allodynia and promotes peripheral axonal and sensory regeneration. This review enlightens effects of KD on various disease conditions. PRACTICAL APPLICATIONS: It is increasingly being realized that diet plays a very important role in our fight against several diseases. This can range from neurological disorders to diabetes and cancer. In this context, the potential of KD, the "High-fat, low-carbohydrate, adequate-protein" diet strategy, is increasingly being realized. In this article, we provide a comprehensive analysis of the benefits of KD against many diseases and discuss the underlying biochemical mechanisms. We hope that our write-up will stimulate further research on KD and help generate an interest for the populations to adopt this healthy diet. It can help overcome the problems associated with weight and dysregulated metabolism.
Collapse
Affiliation(s)
- Rui-Jun Li
- The Handsurgery Department, The First Hospital of Jilin University, Changchun, China
| | - Yang Liu
- The Handsurgery Department, The First Hospital of Jilin University, Changchun, China
| | - Huan-Qiu Liu
- The Anesthesia Department, The First Hospital of Jilin University, Changchun, China
| | - Ji Li
- The Anesthesia Department, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
1119
|
Belete TM. A Recent Achievement In the Discovery and Development of Novel Targets for the Treatment of Type-2 Diabetes Mellitus. J Exp Pharmacol 2020; 12:1-15. [PMID: 32021494 PMCID: PMC6959499 DOI: 10.2147/jep.s226113] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes (T2DM) is a chronic metabolic disorder. Impaired insulin secretion, enhanced hepatic glucose production, and suppressed peripheral glucose use are the main defects responsible for developing the disease. Besides, the pathophysiology of T2DM also includes enhanced glucagon secretion, decreased incretin secretion, increased renal glucose reabsorption, and adipocyte, and brain insulin resistance. The increasing prevalence of T2DM in the world beseeches an urgent need for better treatment options. The antidiabetic drugs focus on control of blood glucose concentration, but the future treatment goal is to delay disease progression and treatment failure, which causes poorer glycemic regulation. Recent treatment approaches target on several novel pathophysiological defects present in T2DM. Some of the promising novel targets being under clinical development include those that increase insulin sensitization (antagonists of glucocorticoids receptor), decreasing hepatic glucose production (glucagon receptor antagonist, inhibitors of glycogen phosphorylase and fructose-1,6-biphosphatase). This review summarizes studies that are available on novel targets being studied to treat T2DM with an emphasis on the small molecule drug design. The experience gathered from earlier studies and knowledge of T2DM pathways can guide the anti-diabetic drug development toward the discovery of drugs essential to treat T2DM.
Collapse
Affiliation(s)
- Tafere Mulaw Belete
- Department of Pharmacology, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
1120
|
Al-Bazz DY, Wilding JP. Dapagliflozin and cardiovascular outcomes in patients with Type 2 diabetes. Future Cardiol 2020; 16:77-88. [PMID: 31914812 DOI: 10.2217/fca-2019-0065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The relationship between cardiovascular disease, heart failure (HF) and Type-2 diabetes (T2DM) is widely recognized. Cardiovascular (CV) outcome trials are required for all new glucose-lowering agents to confirm safety with respect to CV risk. CV outcome trials with SGLT2i inhibitors have shown CV benefit, with reductions in major CV events and HF. This review focuses on the DECLARE-TIMI 58 trial with dapagliflozin in T2DM, which showed noninferiority for major adverse cardiovascular events and reduction in hospitalization for HF and associated CV mortality in a broad range of patients with T2DM. The DAPA-HF trial of dapagliflozin in people with HF with reduced ejection fraction with and without T2DM confirms benefits for those with HF.
Collapse
Affiliation(s)
- Dalal Y Al-Bazz
- Department of Diabetes and Endocrinology, Clinical Sciences Centre, Aintree University Hospital, Liverpool, L9 7AL, UK
| | - John Ph Wilding
- Obesity & Endocrinology Research, Institute of Ageing & Chronic Disease, University of Liverpool, Liverpool, L9 7AL, UK.,Department of Diabetes and Endocrinology, Clinical Sciences Centre, Aintree University Hospital, Liverpool, L9 7AL, UK
| |
Collapse
|
1121
|
The Dephosphorylation of p70S6 (Thr389) Kinase as a Marker of l-Glutamate-Induced Excitotoxicity Related to Diabetes Disturbances—an Unconventional In Vitro Model. Neurotox Res 2020; 37:628-639. [DOI: 10.1007/s12640-019-00155-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/05/2019] [Accepted: 12/19/2019] [Indexed: 12/15/2022]
|
1122
|
Davids SFG, Matsha TE, Peer N, Erasmus RT, Kengne AP. The 7-Year Change in the Prevalence of Insulin Resistance, Inflammatory Biomarkers, and Their Determinants in an Urban South African Population. J Diabetes Res 2020; 2020:3781214. [PMID: 32566678 PMCID: PMC7261324 DOI: 10.1155/2020/3781214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/29/2020] [Accepted: 05/12/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Insulin resistance (IR) and subclinical inflammation are involved in pathological pathways leading to the development of biological cardiovascular risk factors and subsequent cardiovascular events. Therefore, monitoring these processes can provide advanced information on the trajectory of cardiovascular risk profile of a population and inform prevention and control strategies. We investigated changes in IR and subclinical inflammation in a population from Cape Town, South Africa, between 2008/09 and 2014/16. METHODS In a total of 2503 (n = 797, 2008/09) and (n = 1706, 2014/16) participants, IR was calculated using five indices, i.e., insulin fasting, HOMA-IR, QUICKI, McAuley, and Matsuda while subclinical inflammation was measured using usCRP and gamma GT. Linear and logistic regression analyses and interaction tests were conducted. RESULTS The mean age of participants was 53.2 (2008/09) and 48.2 (2014/16), respectively. In females, IR prevalence significantly decreased between 2008/09 and 2014/2016 by all indices (p ≤ 0.021), while subclinical inflammation prevalence increased from 54.7% (2008/09) to 57.1% (2014/16) based on usCRP and 29.6% to 33.4% based on gamma GT. In a multivariate analysis adjusted for the year of study, age, and gender, prominent factors associated with increased IR or subclinical inflammation were obesity levels measured using waist circumference, glycated haemoglobin, and fasting insulin levels. CONCLUSIONS Over the 7-year period, subclinical inflammation increased and this was associated with IR and the metabolic syndrome components, both of which are strong predictors of CVDs. The decrease in IR over the year period reflects in part the much younger age in the second survey.
Collapse
Affiliation(s)
- Saarah Fatoma Gadija Davids
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- SAMRC/CPUT Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Tandi Edith Matsha
- SAMRC/CPUT Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Nasheeta Peer
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Rajiv Timothy Erasmus
- Department of Chemical Pathology, National Health Laboratory Service (NHLS) and Stellenbosch University, Cape Town, South Africa
| | - Andre Pascal Kengne
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
1123
|
Ainehchi N, Khaki A, Ouladsahebmadarek E, Hammadeh M, Farzadi L, Farshbaf-Khalili A, Asnaashari S, Khamnei HJ, Khaki AA, Shokoohi M. The effect of clomiphene citrate, herbal mixture, and herbal mixture along with clomiphene citrate on clinical and para-clinical parameters in infertile women with polycystic ovary syndrome: a randomized controlled clinical trial. Arch Med Sci 2020; 16:1304-1318. [PMID: 33224329 PMCID: PMC7667414 DOI: 10.5114/aoms.2020.93271] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 08/03/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION To evaluate the effect of a herbal mixture (i.e., Mentha spicata, Zingiber officinale, Cinnamomum zeylanicum, and Citrus sinensis) alone and in combination with clomiphene citrate (CC) compared to CC on the treatment of polycystic ovary syndrome (PCOS). MATERIAL AND METHODS This single-blind randomized clinical trial was conducted on 60 infertile participants with PCOS who were randomly divided into three groups. After spontaneous or progestin-induced withdrawal bleeding, group 1 (n = 20) received routine treatment with CC (50-150 mg) for three menstrual cycles from the 3rd to 5th day of menstruation for 5 days while group 2 (n = 20) and group 3 (n = 20) received herbal mixture capsules 700 mg/day and the herbal mixture along with CC for 3 months, respectively. Finally, several related parameters were measured, including the level of sex steroids, homeostatic model assessment for insulin resistance (HOMA-IR), lipid profile (primary outcomes), thyroid hormones, and clinical features. The analysis was based on intention-to-treat analysis. RESULTS No statistically significant differences were observed between the groups in terms of socio-demographic characteristics. However, after adjustment for baseline, luteinizing hormone (aMD = 4.9; 95% CI: 3.7-6.2), luteinizing hormone/follicle-stimulating hormone (aMD = 0.9; 95% CI: 0.7-1.2), total testosterone (aMD = -0.12; 95% CI: -0.2 to -0.01) in group 2 and free testosterone (aMD = -6.0; 95% CI: -9.7 to -2.3) in group 3 revealed a significant difference compared to group 1. In addition, HOMA-IR in group 2 (aMD = -1.3; 95% CI: -2.4 to -0.2) decreased significantly compared to group 1. Further, total cholesterol, triglycerides, low-density lipoprotein cholesterol, and very low-density lipoprotein cholesterol decreased significantly in group 2 (aMD = -21.8; 95% CI: -31.5 to -12.1; aMD = -29.9; 95% CI: -47.9 to -12.0; aMD = -21.2; 95% CI: -31.3 to -11.1; aMD = -5.1; 95% CI: -7.5 to -2.7) and group 3 (aMD = -18.3; 95% CI: -27.4 to -9.2; aMD = -26.9; 95% CI: -43.8 to -9.9; aMD = -21.4; 95% CI: -31.1 to -11.7; aMD = -5.9; 95% CI: -8.3 to -3.6) compared to group 1, respectively. However, high-density lipoproteins cholesterol in group 2 (aMD = 6.8; 95% CI: 2.9-10.7) and group 3 (aMD = 10.7; 95% CI: 7.2-14.7) increased remarkably compared to group 1. Overall, clinical outcomes improved significantly in all groups (p < 0.05). CONCLUSIONS In general, the herbal mixture along with CC was found to improve free testosterone, HOMA-IR, lipid profile, and clinical features of PCOS women.
Collapse
Affiliation(s)
- Nava Ainehchi
- Women’s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arash Khaki
- Women’s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Ouladsahebmadarek
- Women’s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohamad Hammadeh
- Department of Obstetrics and Gynecology, University of Saarland, Homburg, Germany
| | - Laya Farzadi
- Women’s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azizeh Farshbaf-Khalili
- Aging Research Institute, Physical Medicine and Rehabilitation Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Asnaashari
- Biotechnology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Amir Afshin Khaki
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Shokoohi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
1124
|
McElwain CJ, Tuboly E, McCarthy FP, McCarthy CM. Mechanisms of Endothelial Dysfunction in Pre-eclampsia and Gestational Diabetes Mellitus: Windows Into Future Cardiometabolic Health? Front Endocrinol (Lausanne) 2020; 11:655. [PMID: 33042016 PMCID: PMC7516342 DOI: 10.3389/fendo.2020.00655] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Placental insufficiency and adipose tissue dysregulation are postulated to play key roles in the pathophysiology of both pre-eclampsia (PE) and gestational diabetes mellitus (GDM). A dysfunctional release of deleterious signaling motifs can offset an increase in circulating oxidative stressors, pro-inflammatory factors and various cytokines. It has been previously postulated that endothelial dysfunction, instigated by signaling from endocrine organs such as the placenta and adipose tissue, may be a key mediator of the vasculopathy that is evident in both adverse obstetric complications. These signaling pathways also have significant effects on long term maternal cardiometabolic health outcomes, specifically cardiovascular disease, hypertension, and type II diabetes. Recent studies have noted that both PE and GDM are strongly associated with lower maternal flow-mediated dilation, however the exact pathways which link endothelial dysfunction to clinical outcomes in these complications remains in question. The current diagnostic regimen for both PE and GDM lacks specificity and consistency in relation to clinical guidelines. Furthermore, current therapeutic options rely largely on clinical symptom control such as antihypertensives and insulin therapy, rather than that of early intervention or prophylaxis. A better understanding of the pathogenic origin of these obstetric complications will allow for more targeted therapeutic interventions. In this review we will explore the complex signaling relationship between the placenta and adipose tissue in PE and GDM and investigate how these intricate pathways affect maternal endothelial function and, hence, play a role in acute pathophysiology and the development of future chronic maternal health outcomes.
Collapse
Affiliation(s)
- Colm J. McElwain
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
- *Correspondence: Colm J. McElwain
| | - Eszter Tuboly
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| |
Collapse
|
1125
|
Cao Y, Zhong M, Zhang Y, Zheng Z, Liu Y, Ni X, Han L, Song M, Zhang W, Wang Z. Presarcopenia Is an Independent Risk Factor for Carotid Atherosclerosis in Chinese Population with Metabolic Syndrome. Diabetes Metab Syndr Obes 2020; 13:81-88. [PMID: 32021354 PMCID: PMC6956996 DOI: 10.2147/dmso.s235335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 11/22/2019] [Indexed: 12/16/2022] Open
Abstract
PURPOSE To investigate the impacts of skeletal muscle mass on carotid atherosclerosis in Chinese adults with metabolic syndrome (MetS). METHODS One hundred and ninety-five subjects with MetS had the waist-to-height ratio (WHTR)≥0.5 for all. One hundred and eighty-four subjects without MetS were divided into 2 groups: Non-Mets obese group (WHTR ≥ 0.5, n = 118) and Non-MetS control group (WHTR < 0.5, n = 66). All the groups had no difference in age. Appendicular skeletal muscle mass was acquired and skeletal muscle mass index (SMI) was calculated. Carotid intima-media thickness (IMT) was assessed by ultrasonography. Each group was stratified according to the presence or absence of presarcopenia. RESULTS While most parameters showed an increasing trend with WHTR and MetS in both genders, SMI and HDL-C showed a decreasing trend. The prevalence of carotid atherosclerosis showed the same increasing trend. Multivariate logistic regression analyses showed SBP and presarcopenia were both independent risk factors for carotid atherosclerosis in MetS (OR 1.026, P < 0.001; OR 2.788, P = 0.001, respectively). There was no significant difference in IMT among the three groups with preserved muscle mass whether the participants suffered from obesity or MetS, while there was a significant difference between the two groups with presarcopenia (in male P = 0.020, in female P = 0.009, respectively). The area under the ROC curve (AUC) was 0.641 (P<0.001) for presarcopenia. CONCLUSION Obesity was a risk factor for sarcopenia independent of age, especially in subjects with metabolic syndrome. In individuals with MetS, our findings suggest that presarcopenia may be an independent risk factor for atherosclerosis and appendicular skeletal muscle mass had potential protective effects for carotid atherosclerosis regardless of gender.
Collapse
Affiliation(s)
- Yuan Cao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Yuke Zhang
- The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Department of Critical Care Medicine, Jinan, Shandong, People’s Republic of China
| | - Zijie Zheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Yapeng Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Xiaoning Ni
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Lu Han
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
- Department of General Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Ming Song
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
- Correspondence: Wei Zhang The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, No. 107, Wen Hua Xi Road, Jinan250012, People’s Republic of ChinaTel +86 185 6008 6586Fax +86 531 8616 9356 Email
| | - Zhihao Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, and The Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, and The Key Laboratory of Cardiovascular Proteomics of Shandong Province, Jinan, Shandong, People’s Republic of China
- Zhihao Wang The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, The Department of Geriatric Medicine, Qilu Hospital of Shandong University, and The Key Laboratory of Cardiovascular Proteomics of Shandong Province, No. 107, Wen Hua Xi Road, Jinan250012, People’s Republic of ChinaTel +86 185 6008 2268Fax +86 531 8616 9356 Email
| |
Collapse
|
1126
|
Habib SS, Alkahtani S, Alhussain M, Aljuhani O. Sarcopenia Coexisting with High Adiposity Exacerbates Insulin Resistance and Dyslipidemia in Saudi Adult Men. Diabetes Metab Syndr Obes 2020; 13:3089-3097. [PMID: 32922058 PMCID: PMC7457864 DOI: 10.2147/dmso.s260382] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/25/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE The associations between sarcopenia, adiposity indices and metabolic dysregulation still remain controversial. We aimed to assess and compare insulin resistance and metabolic profile in sarcopenic and non-sarcopenic obese Saudi adult men. METHODS This cross sectional study was conducted at the College of Sports Sciences, King Saud University, Riyadh. We recruited 312 Saudi adult male individuals and 288 were finally selected for the study. Body composition analysis and hand grip strength (HGS) were estimated by bioimpedance analysis (BIA) and dynamometer in all subjects, respectively. Fasting blood samples were collected for glucose (FBG), basal insulin (BI) and lipid profile. The subjects were divided into three groups based on the body composition parameters, appendicular lean mass (ALM) and body fat percentage (BF%), into non-obese (NonOb) [Normal ALM+<25 BF%], obese without sarcopenia (ObNonS) [Normal ALM+>25 BF%] and obese with sarcopenia (ObS) [Low ALM+>25 BF%]. RESULTS Obese subjects had significantly higher BI, HOMA-IR and HOMA-β compared to non-obese. Moreover, comparison between two obese groups revealed that both BI and HOMA-IR levels were higher in ObS subjects compared to ObNonS individuals revealing that sarcopenia exacerbates the insulin resistance profile. There was a significant trend of higher resistance and lower sensitivity from non-obese to obesity with sarcopenia. Total cholesterol (TC) and triglycerides (TG) were significantly higher and high density lipoprotein cholesterol (HDL-C) was significantly lower in sarcopenic obese subjects compared to non-sarcopenic obese individuals. The worsening effects were more significant at cutoff point of 7.46 on insulin indices and lipid profile showing that sarcopenia associated with obesity exacerbates the dyslipidemia. CONCLUSION Our study shows that obesity associated with sarcopenia exhibits significantly greater insulin resistance and dyslipidemia than sarcopenia or obesity per se. Therefore, sarcopenic obesity might be an independent risk factor for metabolic disease progression.
Collapse
Affiliation(s)
- Syed Shahid Habib
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Correspondence: Syed Shahid Habib Department of Physiology, College of Medicine, King Saud University, PO Box 2925, King Saud University Medical City, Riyadh11461, Kingdom of Saudi ArabiaTel +966 1-4671616Fax +966 1-4672567 Email
| | - Shaea Alkahtani
- Department of Exercise Physiology, College of Sport Sciences and Physical Activity, King Saud University, Riyadh, Saudi Arabia
| | - Maha Alhussain
- Department of Nutrition and Food Sciences, College of Agricultural and Food Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Osama Aljuhani
- Department of Physical Education, College of Sport Sciences and Physical Activity, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
1127
|
Brandhorst S, Longo VD. Dietary Restrictions and Nutrition in the Prevention and Treatment of Cardiovascular Disease. Circ Res 2019; 124:952-965. [PMID: 30870119 DOI: 10.1161/circresaha.118.313352] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in many developed countries and remains one of the major diseases strongly affected by the diet. Nutrition can affect CVD directly by contributing to the accumulation of vascular plaques and also indirectly by regulating the rate of aging. This review summarizes research on nutrition and CVD incidence based on a multipillar system that includes basic research focused on aging, epidemiological studies, clinical studies, and studies of centenarians. The relevant research linking nutrition and CVD with focus on macronutrients and aging will be highlighted. We will review some of the most relevant studies on nutrition and CVD treatment, also focusing on interventions known to delay aging. We will discuss both everyday dietary compositions, as well as intermittent and periodic fasting interventions with the potential to prevent and treat CVD.
Collapse
Affiliation(s)
- Sebastian Brandhorst
- From the Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles (S.B., V.D.L.)
| | - Valter D Longo
- From the Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles (S.B., V.D.L.).,Institute of Molecular Oncology, Italian Foundation for Cancer Research, Milan (V.D.L.)
| |
Collapse
|
1128
|
Impacts of lifestyle behavior and shift work on visceral fat accumulation and the presence of atherosclerosis in middle-aged male workers. Hypertens Res 2019; 43:235-245. [DOI: 10.1038/s41440-019-0362-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/16/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022]
|
1129
|
Matsumoto T, Takayanagi K, Kobayashi S, Kojima M, Taguchi K, Kobayashi T. Effect of Equol on Vasocontractions in Rat Carotid Arteries Treated with High Insulin. Biol Pharm Bull 2019; 42:1048-1053. [PMID: 31155582 DOI: 10.1248/bpb.b19-00051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous research has indicated that high insulin affects vascular function. Equol is an active metabolite of daidzein, an isoflavone produced from soy by intestinal microbial flora, with beneficial effects on the vascular system. This study investigated whether equol was beneficial for vascular function under high insulin conditions. Using organ culture techniques, rat carotid arteries were treated for 23 ± 1 h with a vehicle, high insulin (100 nM), or equol (100 µM) plus high insulin (100 nM). Vascular isometric forces were measured by the organ bath technique. In each endothelium-intact ring, the contractions induced by high-K+, noradrenaline, or by serotonin (5-HT) were similar for the vehicle, insulin, and equol + insulin treatments. Contractions induced by a selective 5-HT2A receptor agonist (TCB2) increased with insulin treatment (vs. vehicle), but less so with equol + insulin. Under basal conditions, a selective 5-HT2B receptor agonist (BW723C86) did not induce contraction; following precontraction by a thromboxane analog, it induced contraction but not relaxation. These responses were similar across the three treatments. Acetylcholine-induced relaxations were also similar for the three treatments. In the endothelium-denuded preparations, 5-HT-induced contraction was augmented with insulin treatment (vs. vehicle) but less so by equol + insulin treatment. These differences in 5-HT-induced contractions were eliminated by iberiotoxin, a large-conductance calcium-activated K+ channel (BKCa) inhibitor. These results suggest that equol exerts a preventive effect on the enhancement of 5-HT-induced contraction by high insulin (possibly mediated by the 5-HT2A receptor), and that these effects may be attributed to the activation of BKCa channels in vascular smooth muscle.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Keisuke Takayanagi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Shota Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Mihoka Kojima
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| |
Collapse
|
1130
|
Luo E, Wang D, Yan G, Qiao Y, Liu B, Hou J, Tang C. High triglyceride-glucose index is associated with poor prognosis in patients with acute ST-elevation myocardial infarction after percutaneous coronary intervention. Cardiovasc Diabetol 2019; 18:150. [PMID: 31722708 PMCID: PMC6852896 DOI: 10.1186/s12933-019-0957-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023] Open
Abstract
Background Insulin resistance (IR) is considered a pivotal risk factor for cardiometabolic diseases, and the triglyceride–glucose index (TyG index) has emerged as a reliable surrogate marker of IR. Although several recent studies have shown the association of the TyG index with vascular disease, no studies have further investigated the role of the TyG index in acute ST-elevation myocardial infarction (STEMI). The objective of the present study was to evaluate the potential role of the TyG index as a predictor of prognosis in STEMI patients after percutaneous coronary intervention (PCI). Methods The study included 1092 STEMI patients who underwent PCI. The patients were divided into 4 quartiles according to TyG index levels. Clinical characteristics, fasting plasma glucose (FPG), triglycerides (TGs), other biochemical parameters, and the incidence of major adverse cardiovascular and cerebral events (MACCEs) during the follow-up period were recorded. The TyG index was calculated using the following formula: ln[fasting TGs (mg/dL) × FPG (mg/dL)/2]. Results The incidence of MACCEs and all-cause mortality within 30 days, 6 months and 1 year after PCI were higher among STEMI patients with TyG index levels in the highest quartile. The TyG index was significantly associated with an increased risk of MACCEs in STEMI patients within 1 year after PCI, independent of confounding factors, with a value of 1.529 (95% CI 1.001–2.061; P = 0.003) for those in the highest quartile. The area under the curve (AUC) of the TyG index predicting the occurrence of MACCEs in STEMI patients after PCI was 0.685 (95% CI 0.610–0.761; P = 0.001). The results also revealed that Killip class > 1, anaemia, albumin, uric acid, number of stents and left ventricular ejection fraction (LVEF) were independent predictors of MACCEs in STEMI patients after PCI (all P < 0.05). Conclusions This study indicated an association between higher TyG index levels and increased risk of MACCEs in STEMI patients for the first time, and the TyG index might be a valid predictor of clinical outcomes in STEMI patients undergoing PCI. Trial Registration ChiCTR1900024577.
Collapse
Affiliation(s)
- Erfei Luo
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital, Southeast University, Hunan Road, Nanjing, 210009, Jiangsu, China.
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, Southeast University, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital, Southeast University, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Bo Liu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jiantong Hou
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, Southeast University, Hunan Road, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
1131
|
Ndisang JF. Twists and Turns in Cardio-metabolic Diseases and Related Complications. Curr Med Chem 2019; 26:4495-4497. [PMID: 31654562 DOI: 10.2174/092986732624190927115059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Joseph Fomusi Ndisang
- Department of Anatomy, Physiology & Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, Canada
| |
Collapse
|
1132
|
Jin JL, Cao YX, Liu HH, Zhang HW, Guo YL, Wu NQ, Zhu CG, Xu RX, Gao Y, Sun J, Dong Q, Li JJ. Impact of free fatty acids on prognosis in coronary artery disease patients under different glucose metabolism status. Cardiovasc Diabetol 2019; 18:134. [PMID: 31610783 PMCID: PMC6791018 DOI: 10.1186/s12933-019-0936-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The aim of the present study is to examine the effects of free fatty acids (FFAs) on major cardiovascular events (MACEs) in patients with stable coronary artery disease (CAD) and different glucose metabolism status. METHODS In this study, we consecutively enrolled 5443 patients from March 2011 to May 2015. Patients were categorized according to both status of glucose metabolism status [diabetes mellitus (DM), pre-diabetes (Pre-DM), normal glycaemia regulation (NGR)] and FFAs levels. All subjects were followed up for the occurrence of the MACEs. RESULTS During a median of 6.7 years' follow-up, 608 MACEs occurred. A twofold higher FFAs level was independently associated with MACEs after adjusting for confounding factors [Hazard Ratio (HR): 1.242, 95% confidence interval (CI) 1.084-1.424, p value = 0.002]. Adding FFAs to the Cox model increased the C-statistic by 0.015 (0.005-0.027). No significant difference in MACEs was observed between NGR and Pre-DM groups (p > 0.05). When patients were categorized by both status of glucose metabolism and FFAs levels, medium and high FFAs were associated with significantly higher risk of MACEs in Pre-DM [1.736 (1.018-2.959) and 1.779 (1.012-3.126), all p-value < 0.05] and DM [2.017 (1.164-3.494) and 2.795 (1.619-4.824), all p-value < 0.05]. CONCLUSIONS The present data indicated that baseline FFAs levels were associated with the prognosis in DM and Pre-DM patients with CAD, suggesting that FFAs may be a valuable predictor in patients with impaired glucose metabolism.
Collapse
Affiliation(s)
- Jing-Lu Jin
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Ye-Xuan Cao
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Hui-Hui Liu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Hui-Wen Zhang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Yuan-Lin Guo
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Na-Qiong Wu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Cheng-Gang Zhu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Rui-Xia Xu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Ying Gao
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Jing Sun
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Qian Dong
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Jian-Jun Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China.
| |
Collapse
|
1133
|
Kseneva SI, Borodulina EV, Udut VV, Fisenko VP. Mechanism Underlying the Formation of a Cluster of Metabolic Syndrome. Endocr Metab Immune Disord Drug Targets 2019; 20:564-569. [PMID: 31589129 DOI: 10.2174/1871530319666191007115214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND The concept of metabolic syndrome (MetS) as a cluster of risk factors of type 2 diabetes and cardiovascular diseases has undergone some evolutionary transformations over the past years. Integrating the autonomic dysfunction into the pathogenesis of MetS creates the possibility of including a range of nosologies affecting treatment and clinical manifestations of pathologies belonging to MetS into the MetS cluster. The purpose of this work is to determine the involvement of autonomic dysfunction in the pathogenesis of associated pathological conditions in patients and MetS. METHODS A cross-sectional study was conducted. The sample consisted of 158 patients with metabolic syndrome. The patients underwent a physical examination, including BMI; a blood chemistry test with the determination of the hormonal status (insulin, testosterone, dihydrotestosterone); a 24-hour monitoring of blood pressure (BP); an assessment of heart rate variability; studies showing the presence of gastric reflux (рН-measurement) or its damaging impact (endoscopy); men were tested with the IPSSQOL questionnaire and underwent transrectal ultrasound of the prostate and ultrasound of the bladder. RESULTS It is revealed that because of MetS, the occurrence of cardiac autonomic neuropathy reaches 37.5%. Some features of gastroesophageal reflux disease in patients with MetS are shown. Regurgitation prevails in the structure of complaints. In case of fibrogastroduodenoscopy, an endoscopynegative form of the disease occurs in 38%. According to the data of daily pH-measurement, when DeMeester score is high, in the supine position, 25% of the time accounts for alkaline reflux (рН > 7). It is found out that young men experience the enlargement of prostate volume and size; according to the IPSS questionnaire, the scores correspond to the initial manifestations of hyperplastic diseases of the prostate gland due to insulin resistance and normal level of androgens. CONCLUSION The paper demonstrates that the autonomic dysfunction of the nervous system (on a par with insulin resistance) is the main link in the development of MetS. This provides the basis for including the mentioned states - cardiac autonomic neuropathy, lower urinary tract symptoms, and gastroesophageal reflux disease - into the MetS cluster.
Collapse
Affiliation(s)
- Svetlana Igorevna Kseneva
- Ministry of Education and Science, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russian Federation
| | - Elena Valentinovna Borodulina
- Ministry of Education and Science, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russian Federation
| | - Vladimir Vasilievich Udut
- Ministry of Education and Science, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russian Federation.,Ministry of Education and Science National Research Tomsk State University, Tomsk, Russian Federation
| | - Vladimir Petrovich Fisenko
- Ministry of Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| |
Collapse
|
1134
|
Gupta AP, Syed AA, Garg R, Goand UK, Singh P, Riyazuddin M, Valicherla GR, Husain A, Gayen JR. Pancreastatin inhibitor PSTi8 attenuates hyperinsulinemia induced obesity and inflammation mediated insulin resistance via MAPK/NOX3-JNK pathway. Eur J Pharmacol 2019; 864:172723. [PMID: 31586632 DOI: 10.1016/j.ejphar.2019.172723] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/26/2019] [Accepted: 10/02/2019] [Indexed: 01/13/2023]
Abstract
Pancreastatin (PST), a chromogranin A derived peptide has anti-insulin effects and plays a significant role in obesity-induced insulin resistance. In obesity and type 2 diabetes mellitus, both insulin and PST level are elevated, but it is not clearly understood how anti-insulin effect of PST get regulated in hyperinsulinemic state. Simultaneously we have explored pancreastatin inhibitor PSTi8 against the native PST in the same hyperinsulinemic state. In in-vitro studies, we found that PST treatment increases lipid droplets and reactive oxygen species production in 3T3L1 adipocyte cells and theses effects of PST was found synergistic with chronic-insulin treatment. Treatment of PSTi8 in 3T3L1 adipocytes attenuates PST effect on lipid droplet formation and reactive oxygen species production. We further validated these findings in epididymal white adipose tissue of C57BL/6 mice, implanted with mini-osmotic insulin pump with and without PSTi8 for 4 weeks. We found that chronic hyperinsulinemia enhanced PST levels in circulation which in turn induces expression of various pro-inflammatory cytokines and oxidative stress. In addition, it also stimulated the expression of lipogenic genes, fat mass and body weight gain through the regulation of circulating adiponectin level. The change in PST mediated inflammatory and lipogenic parameters were attenuated by PSTi8 treatment, leading to enhanced insulin sensitivity and improved glucose homeostasis. PSTi8 rescue from PST mediated insulin resistance in adipose via inhibition of MAPK and NOX3-JNK stress signalling pathway which stimulates GLUT4 expression through activation of AKT-AS160 pathway. Thus PSTi8 may be a novel therapeutic agent for the treatment of hyperinsulinemia induced obesity and inflammation mediated insulin resistance.
Collapse
Affiliation(s)
- Anand P Gupta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Anees A Syed
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Richa Garg
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Umesh K Goand
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Pragati Singh
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Mohammed Riyazuddin
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Guru R Valicherla
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Athar Husain
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
1135
|
Malekahmadi M, Moradi Moghaddam O, Firouzi S, Daryabeygi-Khotbehsara R, Shariful Islam SM, Norouzy A, Soltani S. Effects of pycnogenol on cardiometabolic health: A systematic review and meta-analysis of randomized controlled trials. Pharmacol Res 2019; 150:104472. [PMID: 31585179 DOI: 10.1016/j.phrs.2019.104472] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/29/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022]
Abstract
AIM Clinical trials on the effect of pycnogenol supplementation on cardiometabolic health have been controversial. We conducted a systematic review and meta-analysis of randomized controlled trials (RCTs) to evaluate the potential effect of pycnogenol supplementation on cardiometabolic profile. METHODS PubMed, Scopus, and ISI Web of Science databases were searched until October 2018. RCTs that evaluated the effects of pycnogenol on cardiometabolic parameters were included. DerSimonian and Laird random-effect models were used to compute the weighted mean differences (WMDs) and 95% confidence intervals (CIs). RESULTS Twenty-four RCTs including 1594 participants were included in the meta-analysis. Pycnogenol significantly reduced fasting blood glucose (WMD: -5.86 mg/dl; 95% CI: -9.56, -2.15), glycated hemoglobin (WMD = -0.29%, 95%CI: -0.56, -0.01), systolic blood pressure (WMD: -2.54 mmhg; 95% CI: -4.08, -0.99), diastolic blood pressure (WMD: -1.76 mmhg; 95% CI: -3.12, -0.41), body mass index (WMD: -0.47 kg/m2; 95% CI: -0.90, -0.03), LDL cholesterol (WMD: -7.12 mg/dl; 95% CI: -13.66, -0.58) and increased HDL cholesterol (WMD: 3.27 mg/dl; 95% CI: 0.87, 5.66). CONCLUSION This meta-analysis suggests that pycnogenol may have a role in preventing cardiometabolic disease. However, further well-designed RCTs are recommended to evaluate its long-term effects and explore the optimal duration of use and dosage.
Collapse
Affiliation(s)
- Mahsa Malekahmadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran; Nutrition Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Omid Moradi Moghaddam
- Trauma and Injury Research Center, Critical Care Department, Rasoul-e-Akram Complex Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Safieh Firouzi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran; Nutrition Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Daryabeygi-Khotbehsara
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Australia
| | - Sheikh Mohammed Shariful Islam
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Australia
| | - Abdolreza Norouzy
- Nutrition Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sepideh Soltani
- Department of Nutrition, Faculty of Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
1136
|
Ali A, Bain S, Hicks D, Newland Jones P, Patel DC, Evans M, Fernando K, James J, Milne N, Viljoen A, Wilding J. SGLT2 Inhibitors: Cardiovascular Benefits Beyond HbA1c-Translating Evidence into Practice. Diabetes Ther 2019; 10:1595-1622. [PMID: 31290126 PMCID: PMC6778582 DOI: 10.1007/s13300-019-0657-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD), including heart failure (HF), is a leading cause of morbidity and mortality in people with type 2 diabetes mellitus (T2DM). CVD and T2DM share common risk factors for development and progression, and there is significant overlap between the conditions in terms of worsening outcomes. In assessing the cardiovascular (CV) safety profiles of anti-diabetic drugs, sodium-glucose co-transporter-2 inhibitor (SGLT2i) therapies have emerged with robust evidence for reducing the risk of adverse CVD outcomes in people with T2DM who have either established CVD or are at risk of developing CVD. A previous consensus document from the Improving Diabetes Steering Committee has examined the potential role of SGLT2is in T2DM management and considered the risk-benefit profile of the class and the appropriate place for these medicines within the T2DM pathway. This paper builds on these findings and presents practical guidance for maximising the pleiotropic benefits of this class of medicines in people with T2DM in terms of reducing adverse CVD outcomes. The Improving Diabetes Steering Committee aims to offer evidence-based practical guidance for the use of SGLT2i therapies in people with T2DM stratified by CVD risk. This is of particular importance currently because some treatment guidelines have not been updated to reflect recent evidence from cardiovascular outcomes trials (CVOTs) and real-world studies that complement the CVOTs. The Improving Diabetes Steering Committee seeks to support healthcare professionals (HCPs) in appropriate treatment selection for people with T2DM who are at risk of developing or have established CVD and examines the role of SGLT2i therapy for these people.Funding: Napp Pharmaceuticals Limited.
Collapse
Affiliation(s)
- Amar Ali
- Oakenhurst Medical Practice, Blackburn, UK
| | - Steve Bain
- Diabetes Research Unit Cymru, Swansea University, Swansea, UK
| | | | | | - Dipesh C Patel
- Department of Diabetes and Endocrinology, Division of Medicine, University College London, London, UK
| | - Marc Evans
- Department of Diabetes, University Hospital Llandough, Llandough, UK
| | | | - June James
- University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Nicola Milne
- CODES (Community Diabetes Education and Support), Manchester University NHS Foundation Trust, Manchester, UK
| | - Adie Viljoen
- Department of Metabolic Medicine/Chemical Pathology, Lister Hospital, Stevenage, UK
| | - John Wilding
- Obesity and Endocrinology Research, University of Liverpool, Liverpool, UK.
| |
Collapse
|
1137
|
Chan YH, Chang GJ, Lai YJ, Chen WJ, Chang SH, Hung LM, Kuo CT, Yeh YH. Atrial fibrillation and its arrhythmogenesis associated with insulin resistance. Cardiovasc Diabetol 2019; 18:125. [PMID: 31558158 PMCID: PMC6761716 DOI: 10.1186/s12933-019-0928-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/14/2019] [Indexed: 02/07/2023] Open
Abstract
Background Insulin resistance (IR) is considered as a risk factor for atrial fibrillation (AF) even before diabetes develops. The pathophysiology and underlying mechanism are largely unclear. Methods We investigated the corresponding mechanism in two IR models of rats fed 15-week high-fat (HFa) and high-fructose/cholesterol (HFr) diets. AF was evaluated and induced by burst atrial pacing. Isolated atrial myocytes were used for whole-cell patch clamp and calcium assessment. Ex vivo whole heart was used for optical mapping. Western blot and immunofluorescence were used for quantitative protein evaluation. Results Both HFa and HFr rat atria were vulnerable to AF evaluated by burst atrial pacing. Isolated atrial myocytes from HFa and HFr rats revealed significantly increased sarcoplasmic reticulum calcium content and diastolic calcium sparks. Whole-heart mapping showed prolonged calcium transient duration, conduction velocity reduction, and repetitive ectopic focal discharge in HFa and HFr atria. Protein analysis revealed increased TGF-β1 and collagen expression; increased superoxide production; abnormal upregulation of calcium-homeostasis-related proteins, including oxidized CaMKIIδ, phosphorylated-phospholamban, phosphorylated-RyR-2, and sodium-calcium exchanger; and increased Rac1 activity in both HFa and HFr atria. We observed that inhibition of CaMKII suppressed AF in both HF and HFr diet-fed rats. In vitro palmitate-induced IR neonatal cardiomyocytes and atrial fibroblasts expressed significantly more TGF-β1 than did controls, suggesting paracrine and autocrine effects on both myocytes and fibroblasts. Conclusions IR engenders both atrial structural remodeling and abnormal intracellular calcium homeostasis, contributing to increased AF susceptibility. The inhibition of CaMKII may be a potential therapeutic target for AF in insulin resistance.
Collapse
Affiliation(s)
- Yi-Hsin Chan
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,College of Medicine, Chang-Gung University, Taoyuan, Taiwan.,Microscopy Core Laboratory, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang-Gung University, Taoyuan, Taiwan
| | - Ying-Ju Lai
- Department of Respiratory Therapy, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,College of Medicine, Chang-Gung University, Taoyuan, Taiwan.,Center for Big Data Analytics and Statistics, Chang-Gung Memorial Hospital, Taoyuan, Taiwan
| | - Li-Man Hung
- Department of Biomedical Sciences, College of Medicine, Healthy and Aging Research Center, Chang-Gung University, Taoyuan, Taiwan
| | - Chi-Tai Kuo
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan. .,College of Medicine, Chang-Gung University, Taoyuan, Taiwan.
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang-Gung Memorial Hospital, Linkou, Taoyuan, Taiwan. .,College of Medicine, Chang-Gung University, Taoyuan, Taiwan.
| |
Collapse
|
1138
|
Yan Y, Li S, Liu Y, Bazzano L, He J, Mi J, Chen W. Temporal relationship between inflammation and insulin resistance and their joint effect on hyperglycemia: the Bogalusa Heart Study. Cardiovasc Diabetol 2019; 18:109. [PMID: 31443647 PMCID: PMC6706925 DOI: 10.1186/s12933-019-0913-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Inflammation and insulin resistance play crucial roles in the development of type 2 diabetes mellitus (T2DM). We aim to examine the temporal relationship between high-sensitivity C-reactive protein (hsCRP) and insulin resistance in non-diabetic adults and their joint effect on the development of hyperglycemia. METHODS The longitudinal cohort from the Bogalusa Heart Study consisted of 509 non-diabetic adults (360 whites and 149 blacks, mean age = 42.8 years at follow-up) who had hsCRP, fasting glucose and insulin measured twice at baseline and follow-up over 6.8 years. Cross-lagged panel model was used to examine the temporal relationship between hsCRP and homeostasis model assessment for insulin resistance (HOMA-IR). Information on incident T2DM was collected in a survey in 6.1 years after the follow-up survey. RESULTS After adjusting for race, sex, age, body mass index, smoking, alcohol drinking and follow-up years, the path coefficient from baseline hsCRP to follow-up HOMA-IR (β2 = 0.105, p = 0.009) was significant and greater than the path from baseline HOMA-IR to follow-up hsCRP (β1 = 0.005, p = 0.903), with p = 0.011 for the difference between β1 and β2. This one-directional path from baseline hsCRP to follow-up HOMA-IR was significant in the hyperglycemia group but not in the normoglycemia group. In addition, participants with high levels of baseline hsCRP and follow-up HOMA-IR had greater risks of T2DM (odds ratio, OR = 2.38, p = 0.035), pre-T2DM (OR = 2.27, p = 0.006) and hyperglycemia (OR = 2.18, p = 0.003) than those with low-low levels. CONCLUSIONS These findings suggest that elevated hsCRP is associated with future insulin resistance in non-diabetic adults, and their joint effect is predictive of the development of T2DM.
Collapse
Affiliation(s)
- Yinkun Yan
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
- Department of Non-communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, Room 1504G, New Orleans, LA, USA
| | - Shengxu Li
- Children's Minnesota Research Institute, Children's Hospitals and Clinics of Minnesota, Minneapolis, MN, USA
| | - Yang Liu
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, Room 1504G, New Orleans, LA, USA
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lydia Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, Room 1504G, New Orleans, LA, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, Room 1504G, New Orleans, LA, USA
| | - Jie Mi
- Department of Non-communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, Room 1504G, New Orleans, LA, USA.
| |
Collapse
|
1139
|
Rebollo-Hernanz M, Zhang Q, Aguilera Y, Martín-Cabrejas MA, Gonzalez de Mejia E. Relationship of the Phytochemicals from Coffee and Cocoa By-Products with their Potential to Modulate Biomarkers of Metabolic Syndrome In Vitro. Antioxidants (Basel) 2019; 8:E279. [PMID: 31387271 PMCID: PMC6721099 DOI: 10.3390/antiox8080279] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/28/2019] [Accepted: 08/02/2019] [Indexed: 12/18/2022] Open
Abstract
This study aimed to compare the phytochemicals from coffee and cocoa by-products and their relationship with the potential for reducing markers of inflammation, oxidative stress, adipogenesis, and insulin resistance in vitro. We characterized the phytochemical profile of extracts from coffee husk, coffee silverskin, and cocoa shell and evaluated their in vitro biological activity in RAW264.7 macrophages and 3T3-L1 adipocytes. Pearson correlations and principal component regressions were performed to find the contribution of phytochemicals and underlying mechanisms of action. Coffee husk and silverskin extracts were mainly composed of caffeine and chlorogenic acid. Major components in cocoa shell included theobromine and protocatechuic acid. Both coffee and cocoa by-product extracts effectively reduced inflammatory markers in macrophages and adipocytes (NO, PGE2, TNF-α, MCP-1, and IL-6) and the production of reactive oxygen species (21.5-66.4%). Protocatechuic and chlorogenic acids, together with caffeine, were suggested as main contributors against inflammation and oxidative stress. Furthermore, extracts reduced lipid accumulation (4.1-49.1%) in adipocytes by regulating lipolysis and inducing adipocyte browning. Gallic and chlorogenic acids were associated with reduced adipogenesis, and caffeine with adipocyte browning. Extracts from coffee and cocoa by-products also modulated the phosphorylation of insulin receptor signaling pathway and stimulated GLUT-4 translocation (52.4-72.9%), increasing glucose uptake. The insulin-sensitizing potential of the extracts was mainly associated with protocatechuic acid. For the first time, we identified the phytochemicals from coffee and cocoa by-products and offered new insights into their associations with biomarkers of inflammation, oxidative stress, adipogenesis, and insulin resistance in vitro.
Collapse
Affiliation(s)
- Miguel Rebollo-Hernanz
- Institute of Food Science Research, CIAL (UAM-CSIC), 28049 Madrid, Spain
- Department of Agricultural Chemistry and Food Science, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Qiaozhi Zhang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310000, China
| | - Yolanda Aguilera
- Institute of Food Science Research, CIAL (UAM-CSIC), 28049 Madrid, Spain
- Department of Agricultural Chemistry and Food Science, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Maria A Martín-Cabrejas
- Institute of Food Science Research, CIAL (UAM-CSIC), 28049 Madrid, Spain
- Department of Agricultural Chemistry and Food Science, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Elvira Gonzalez de Mejia
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
1140
|
Cobbs A, Chen X, Zhang Y, George J, Huang MB, Bond V, Thompson W, Zhao X. Saturated fatty acid stimulates production of extracellular vesicles by renal tubular epithelial cells. Mol Cell Biochem 2019; 458:113-124. [PMID: 30993495 PMCID: PMC7027953 DOI: 10.1007/s11010-019-03535-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/10/2019] [Indexed: 12/30/2022]
Abstract
Lipotoxicity, an accumulation of intracellular lipid metabolites, has been proposed as an important pathogenic mechanism contributing to kidney dysfunction in the context of metabolic disease. Palmitic acid, a predominant lipid derivative, can cause lipoapoptosis and the release of inflammatory extracellular vesicles (EVs) in hepatocytes, but the effect of lipids on EV production in chronic kidney disease remains vaguely explored. This study was aimed to investigate whether palmitic acid would stimulate EV release from renal proximal tubular epithelial cells. Human and rat proximal tubular epithelial cells, HK-2 and NRK-52E, were incubated with 1% bovine serum albumin (BSA), BSA-conjugated palmitic acid (PA), and BSA-conjugated oleic acid (OA) for 24-48 h. The EVs released into conditioned media were isolated by ultracentrifugation and quantified by nanoparticle-tracking analysis (NTA). According to NTA, the size distribution of EVs was 30-150 nm with similar mode sizes in all experimental groups. Moreover, BSA-induced EV release was significantly enhanced in the presence of PA, whereas EV release was not altered by the addition of OA. In NRK-52E cells, PA-enhanced EV release was associated with an induction of cell apoptosis reflected by an increase in cleaved caspase-3 protein by Western blot and Annexin V positive cells analyzed by flow cytometry. Additionally, confocal microscopy confirmed the uptake of lipid-induced EVs by recipient renal proximal tubular cells. Collectively, our results indicate that PA stimulates EV release from cultured proximal tubular epithelial cells. Thus, extended characterization of lipid-induced EVs may constitute new signaling paradigms contributing to chronic kidney disease pathology.
Collapse
Affiliation(s)
- Alyssa Cobbs
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Xiaoming Chen
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Yuanyuan Zhang
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Jasmine George
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Ming-Bo Huang
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Vincent Bond
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Winston Thompson
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Xueying Zhao
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| |
Collapse
|
1141
|
de Souza LM, de Mendonça CL, de ASSIS RN, Oliveira Filho EF, Araújo Gonçalves DN, Cavalcante Souto RJ, Soares PC, Bastos Afonso JA. CARDIAC BIOMARKERS TROPONIN I AND CK-MB IN EWES AFFECTED BY PREGNANCY TOXEMIA. Small Rumin Res 2019. [DOI: 10.1016/j.smallrumres.2019.06.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
1142
|
Ko NY, Lo YTC, Huang PC, Huang YC, Chang JL, Huang HB. Changes in insulin resistance mediate the associations between phthalate exposure and metabolic syndrome. ENVIRONMENTAL RESEARCH 2019; 175:434-441. [PMID: 31158561 DOI: 10.1016/j.envres.2019.04.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/19/2019] [Accepted: 04/20/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Information on the relationships between phthalate exposure, insulin resistance, and metabolic syndrome (MetS) in younger adults is limited. It is still unclear whether changes in insulin resistance represent an intermediate biological mechanism linking phthalate exposure and MetS. OBJECTIVE To investigate the associations between cumulative risk of phthalates (such as daily intake [DI] and hazard index [HI]), insulin resistance, and MetS in younger adults and to examine the mediating role of insulin resistance in the associations between phthalate exposure and MetS. METHODS Urinary phthalate metabolite levels, insulin resistance (by using the Homeostatic Model Assessment of estimated Insulin Resistance [HOMA-IR]), and MetS status were determined in 435 military personnel in Taiwan. We estimated the DI of five phthalates: dimethyl phthalate (DMP), diethyl phthalate, dibutyl phthalate, benzyl butyl phthalate (BBzP), and di (2-ethylhexyl) phthalate and the HI based on urinary phthalate metabolite levels. Cross-sectional associations between DI and HI, HOMA-IR, and the indicators of MetS were explored using logistic regression models. Mediation analysis was conducted to assess the role of insulin resistance in the associations between phthalate exposure and MetS. RESULTS Higher DIDMP was associated with an increased odds of high HOMA-IR and MetS (odds ratio [OR], 1.686; 95% confidence interval [CI], 1.079-2.634 for high HOMA-IR; OR, 2.329; 95% CI, 1.263-4.295 for MetS). The mediation analysis indicated that 43% of the association between higher DIDMP and MetS was mediated by HOMA-IR. Higher DIBBzP and HI were associated with an increased odds of abdominal obesity (OR, 1.816; 95% CI, 1.180-2.797 for the high DIBBzP group; OR, 1.700, 95% CI, 1.105-2.614 for the high HI groups). CONCLUSIONS Exposure to environmental phthalates may be positively associated with insulin resistance and MetS. Insulin resistance may mediate these associations between exposure to certain phthalates and MetS.
Collapse
Affiliation(s)
- Nai-Yueh Ko
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Ting C Lo
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Po-Chin Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli and Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yi-Chen Huang
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Junn-Liang Chang
- Department of Pathology & Laboratory Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Han-Bin Huang
- School of Public Health, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
1143
|
Jeon HY, Seo JA, Jung SH, Lee YJ, Han ET, Park WS, Hong SH, Kim YM, Ha KS. Insulin prevents pulmonary vascular leakage by inhibiting transglutaminase 2 in diabetic mice. Life Sci 2019; 233:116711. [PMID: 31374233 DOI: 10.1016/j.lfs.2019.116711] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/27/2019] [Accepted: 07/29/2019] [Indexed: 12/15/2022]
Abstract
AIMS Insulin is a central peptide hormone required for carbohydrate metabolism; however, its role in diabetes-associated pulmonary disease is unknown. Here, we investigated the preventative effect of insulin against hyperglycemia-induced pulmonary vascular leakage and its molecular mechanism of action in the lungs of diabetic mice. MAIN METHODS Vascular endothelial growth factor (VEGF) activated transglutaminase 2 (TGase2) by sequentially elevating intracellular Ca2+ and reactive oxygen species (ROS) levels in primary human pulmonary microvascular endothelial cells (HPMVECs). KEY FINDINGS Insulin inhibited VEGF-induced TGase2 activation, but did not affect intracellular Ca2+ elevation and ROS generation. Insulin prevented VEGF-induced vascular leakage by inhibiting TGase2-mediated c-Src phosphorylation, disassembly of VE-cadherin and β-catenin, and stress fiber formation. Insulin replacement therapy prevented hyperglycemia-induced TGase2 activation, but not ROS generation, in the lungs of diabetic mice. Insulin also prevented vascular leakage and cancer metastasis in the diabetic lung. Notably, vascular leakage was not detectable in the lungs of TGase2-null (Tgm2-/-) diabetic mice. SIGNIFICANCE These findings demonstrate that insulin prevents hyperglycemia-induced pulmonary vascular leakage in diabetic mice by inhibiting VEGF-induced TGase2 activation rather than ROS generation.
Collapse
Affiliation(s)
- Hye-Yoon Jeon
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Republic of Korea
| | - Jae-Ah Seo
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Republic of Korea
| | - Se-Hui Jung
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Republic of Korea
| | - Yeon-Ju Lee
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Republic of Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Republic of Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Republic of Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Kangwon-do 24341, Republic of Korea.
| |
Collapse
|
1144
|
Mendez CE, Walker RJ, Eiler CR, Mishriky BM, Egede LE. Insulin therapy in patients with type 2 diabetes and high insulin resistance is associated with increased risk of complications and mortality. Postgrad Med 2019; 131:376-382. [PMID: 31311382 DOI: 10.1080/00325481.2019.1643635] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Objective: To investigate the relationship between insulin use and clinical outcomes in patients with type 2 diabetes stratified by level of insulin resistance (IR).Methods: Cross sectional analysis of the NHANES database from 2001 to 2010. Sample was comprised of 3,124 individuals with diabetes, representing a US population of 16,713,593. Insulin use was self-reported. Fasting glucose and insulin levels were used to assess IR by HOMA-IR determination. Subjects were allocated within High or Low HOMA-IR groups based on the sample median. Outcome variables were mortality, major adverse cardiovascular events (MACE), and diabetic kidney disease (DKD). Logistic regression adjusting for covariates including glycemic control and comorbidities were performed.Results: In the adjusted model, insulin use was significantly associated with increased risk of mortality (OR: 2.39, 95% CI: 1.136-5.010) having a MACE (OR: 2.45, 95% CI: 1.137-4.550), and developing DKD (OR: 1.89, 95% CI: 1.119-3.198) in the high HOMA-IR group. The association between insulin use and the outcome variables was not statistically significant in patients within the low HOMA-IR group.Conclusions: Insulin use was associated with increased risk of mortality, MACE, and DKD in patients within the high IR group, but the association was not significant within the low IR group. Our findings indicate that insulin therapy could be less beneficial in patients with high IR. Prospective studies are needed to identify subsets of individuals with type 2 diabetes who would benefit the most from insulin therapy, and for which patients, insulin should be avoided.
Collapse
Affiliation(s)
- Carlos E Mendez
- Medical College of Wisconsin, Director Diabetes Program, Zablocki VA Medical Center, Milwaukee, WI, USA
| | - Rebekah J Walker
- Division of General Internal Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.,Center for Advancing Population Science, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christian R Eiler
- Center for Advancing Population Science, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Basem M Mishriky
- Department of Internal Medicine, East Carolina University, Greenville, NC, USA
| | - Leonard E Egede
- Division of General Internal Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.,Center for Advancing Population Science, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
1145
|
Jung TW, Ahn SH, Shin JW, Kim HC, Park ES, Abd El-Aty AM, Hacımüftüoğlu A, Song KH, Jeong JH. Protectin DX ameliorates palmitate-induced hepatic insulin resistance through AMPK/SIRT1-mediated modulation of fetuin-A and SeP expression. Clin Exp Pharmacol Physiol 2019; 46:898-909. [PMID: 31246318 DOI: 10.1111/1440-1681.13131] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/08/2019] [Accepted: 06/21/2019] [Indexed: 12/15/2022]
Abstract
The role as well as the molecular mechanisms of protectin DX (PDX) in the prevention of hepatic insulin resistance, a hallmark of type 2 diabetes, remains unknown. Therefore, the present study was designed to explore the direct impact of PDX on insulin resistance and to investigate the expression of fetuin-A and selenoprotein P (SeP), hepatokines that are involved in insulin signalling, in hepatocytes. Human serum levels of PDX as well as fetuin-A and SeP were determined by high-performance liquid chromatography (HPLC). Human primary hepatocytes were treated with palmitate and PDX. NF-κB phosphorylation as well as expression of insulin signalling associated genes and hepatokines were determined by Western blotting analysis. FOXO1 binding levels were measured by quantitative real-time PCR. Selected genes from candidate pathways were evaluated by small interfering (si) RNA-mediated gene suppression. Serum PDX levels were significantly (P < 0.05) downregulated, whereas serum fetuin-A and SeP levels were increased (P < 0.05) in obese subjects compared with healthy subjects. In in vitro experiments, PDX treatment increased AMP-activated protein kinase (AMPK) phosphorylation and SIRT1 expression and attenuated palmitate-induced fetuin-A and SeP expression and insulin resistance in hepatocytes. AMPK or SIRT1 siRNA mitigated the suppressive effects of PDX on palmitate-induced fetuin-A through NF-κB and SeP expression linked to FOXO1 and insulin resistance. Recombinant fetuin-A and SeP reversed the suppressive effects of fetuin-A and SeP expression on palmitate-mediated impairment of insulin signalling. The current finding provides novel insight into the underlying mechanism linking hepatokines to the pathogenesis of hepatic insulin resistance.
Collapse
Affiliation(s)
- Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Sung Ho Ahn
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Jong Wook Shin
- Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Korea
| | - Eon Sub Park
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.,Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey
| | - Ahmet Hacımüftüoğlu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey
| | - Ki Hak Song
- Department of Urology, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Korea
| |
Collapse
|
1146
|
Matsumoto T, Ohnishi H, Sato T, Miki T, Akasaka H, Hanawa N, Koyama M, Saitoh S, Miura T. Insulin Resistance is Associated with Longitudinal Changes of Cardiac Repolarization Heterogeneity in Apparently Healthy Subjects. Cardiol Ther 2019; 8:239-251. [PMID: 31273651 PMCID: PMC6828911 DOI: 10.1007/s40119-019-0140-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Increased heterogeneity in ventricular repolarization is a risk factor of sudden cardiac death, but its natural history is unclear. Here we examined whether insulin resistance is associated with longitudinal change in ventricular repolarization heterogeneity in apparently healthy subjects. METHODS The study subjects were participants in health checkups in cohort 1 and cohort 2, which were followed up for 6 years and 5 years, respectively. Subjects with diabetes, cardiovascular disease, or renal disease at baseline were excluded from the analyses. As indices of insulin resistance, the homeostasis model assessment of insulin resistance (HOMA-IR) and triglyceride to HDL-cholesterol ratio (TG/HDL-C) were used in cohort 1 and cohort 2, respectively. Heterogeneity in ventricular repolarization was assessed by heart rate-corrected Tpeak-Tend interval in V5 (cTpTe), QT interval, and QT dispersion. In regression analyses, parameters with a skewed distribution were normalized by logarithmic transformation or by Box-Cox transformation. RESULTS In longitudinal analyses, Box-Cox-transformed cTpTe at the end of follow-up was weakly correlated with log HOMA-IR at baseline in cohort 1 (n = 153, r = - 0.207, 95% CI - 0.354 to - 0.050, p = 0.010) and with log TG/HDL-C at baseline in cohort 2 (n = 738, r = - 0.098, 95% CI - 0.169 to - 0.026, p = 0.008). Multiple regression analysis showed that indices of insulin resistance, but not glycosylated hemoglobin (HbA1c) or plasma glucose, at baseline were significant explanatory variables for cTpTe at the end of follow-up. Neither QT interval nor QT dispersion was correlated with metabolic parameters. CONCLUSION Insulin resistance may be involved in the longitudinal increase of ventricular repolarization heterogeneity in apparently healthy subjects.
Collapse
Affiliation(s)
- Tamaki Matsumoto
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Ohnishi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Miki
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Akasaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | - Masayuki Koyama
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shigeyuki Saitoh
- Division of Medical and Behavioral Subjects, Department of Nursing, Sapporo Medical University School of Health Sciences, Sapporo, Japan
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| |
Collapse
|
1147
|
Close association between circulating high-sensitivity cardiac troponin I and metabolic syndrome in the general population. Hypertens Res 2019; 42:1768-1775. [PMID: 31222189 DOI: 10.1038/s41440-019-0283-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/30/2019] [Accepted: 05/23/2019] [Indexed: 01/10/2023]
Abstract
Individuals with metabolic syndrome reportedly have an increased risk of cardiovascular disease, although the association between asymptomatic myocardial damage and metabolic syndrome has not been sufficiently investigated. The present study investigated possible associations between circulating cardiac troponin and metabolic syndrome or related factors. Subjects undergoing their annual health checkups were enrolled in the study (n = 1242). Laboratory measurements included serum high-sensitivity cardiac troponin I (hs-cTnI) and plasma B-type natriuretic peptide (BNP). Individual salt intake was estimated by calculating 24-h urinary sodium excretion from spot urine. Subjects whose electrocardiograms revealed ST-T segment abnormalities or who had renal insufficiency or a history of cardiovascular events were excluded. Subjects with metabolic syndrome had higher hs-cTnI levels than those without, but their BNP levels were equivalent. hs-cTnI levels were significantly associated with the presence and components of metabolic syndrome. Logistic regression analysis with the endpoint of hs-cTnI levels higher than the median value identified metabolic syndrome as an independent determinant of increased hs-cTnI levels. Additionally, urinary salt excretion levels were increased in subjects with metabolic syndrome or any of its components. Logistic regression analysis with the endpoint of metabolic syndrome revealed that hs-cTnI levels were independently associated with the presence of metabolic syndrome. A close association between hs-cTnI levels and the presence of metabolic syndrome, at least partially mediated by increased salt intake, was confirmed to exist in the general population. The findings support the idea that patients with metabolic syndrome develop asymptomatic myocardial damage without obvious ischaemic findings, which leads to increased cardiovascular risk.
Collapse
|
1148
|
Mao Q, Zhao N, Wang Y, Li Y, Xiang C, Li L, Zheng W, Xu S, Zhao XH. Association of Cystatin C with Metabolic Syndrome and Its Prognostic Performance in Non-ST-Segment Elevation Acute Coronary Syndrome with Preserved Renal Function. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8541402. [PMID: 31317040 PMCID: PMC6601472 DOI: 10.1155/2019/8541402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 04/11/2019] [Indexed: 12/03/2022]
Abstract
OBJECTIVE The underlying mechanisms by which cystatin C affects cardiovascular disease (CVD) are not very clear. Metabolic syndrome (MetS) is a cluster of risk factors that increase the risk of CVD. Here, we aimed to investigate the association of cystatin C with metabolic syndrome and cardiovascular outcomes in non-ST-segment elevation acute coronary syndrome (NSTE-ACS) with preserved renal function. METHODS In total, 422 NSTE-ACS patients with preserved renal function were enrolled to examine the association of cystatin C with MetS. MetS was defined based on the NCEP-ATP-III guidelines. Major adverse cardiovascular events (MACEs) were also evaluated, which included cardiac death, nonfatal myocardial infarction (MI), target vessel revascularization (TVR), heart failure, and nonfatal stroke. All patients underwent a 12-month follow-up for MACEs after admission. RESULTS Cystatin C was significantly correlated with metabolic risk factors and inflammation markers. The prevalence of MetS and MACEs correlated with cystatin C levels. Cystatin C showed a strong diagnostic performance for cardiovascular risk factors and outcomes in ROC analysis. After adjustment for multiple risk factors, cystatin C level was independently associated with MetS (OR 2.299, 95% CI 1.251-4.225, and P = 0.007). During a 12-month follow-up, the patients with high cystatin C level and MetS had higher incidence of MACEs (Log-rank = 24.586, P < 0.001) and cardiac death (Log-rank = 9.890, P = 0.020) compared to the others. Multivariate Cox analysis indicated that cystatin C level was an independent predictor of MACEs (HR 2.609, 95% CI 1.295-5.257, and P = 0.007). CONCLUSION Cystatin C may be an independent predictor of metabolic syndrome and therefore valuable for management of NSTE-ACS patients. Further multicenter, large-scale studies are required to assess the implication of these results.
Collapse
Affiliation(s)
- Qi Mao
- Department of Cardiovascular Medicine, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Ning Zhao
- Department of Cardiovascular Medicine, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Yuqing Wang
- Department of Cardiovascular Medicine, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Youmei Li
- Department of Cardiovascular Medicine, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Chaojun Xiang
- Department of Cardiovascular Medicine, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Lufeng Li
- Department of Cardiovascular Medicine, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Wei Zheng
- Department of Cardiovascular Medicine, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Shangcheng Xu
- Department of Occupational Health, Army Medical University, Chongqing 400038, China
| | - Xiao-Hui Zhao
- Department of Cardiovascular Medicine, Institute of Cardiovascular Research, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
1149
|
Lin Z, Li Q, Sun Y, Huang J, Wang W, Fu J, Xu J, Zeng D. Interactions between genetic variants involved in the folate metabolic pathway and serum lipid, homocysteine levels on the risk of recurrent spontaneous abortion. Lipids Health Dis 2019; 18:143. [PMID: 31200713 PMCID: PMC6570969 DOI: 10.1186/s12944-019-1083-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 06/04/2019] [Indexed: 12/20/2022] Open
Abstract
Background The interaction between folate pathway gene polymorphisms and homocysteine, serum lipid leverls are poorly understood in patients with recurrent spontaneous abortion (RSA). The aim of this study is to explore the effects of folate pathway gene polymorphisms (the 5–10-methylenetetrahydrofolate reductase, MTHTR C677T, MTHFR A1298C and the methionine synthase reductase, MTRR A66G) and their interactions with homocysteine on serum lipid levels in patients with RSA. Methods A total of 403 RSA women and 342 healthy women were randomly selected. Genotyping of the MTHFR C677T, A1298C and MTRR A66G were performed by TaqMan-MGB technique. Serum homocysteine, folate, fasting glucose, fasting insulin, Interleukin 6, Tumor necrosis factorα (TNFα) and lipid profiles were measured according to the kits. Continuous variables were analyzed using 2-sample t-tests. Categorical variables were analyzed and compared by χ2 or Fisher’s exact tests. Unconditional logistic regression model was applied to test the interactions of gene polymorphisms on RSA. Results The distribution of genotype of CC, CT TT and T allele of MTHFR C677T, genotype of AA and C allele of MTHFR A1298C, and genotype of AA, AG and G allele of MTRR A66G were different between cases and controls (all p were < 0.05). There were significant interactions between MTHFR C677T-A1298C and MTHFR A1298C-MTRR A66G in RSA group and control group, with ORs of 1.62 (95%CI: 1.28–2.04, p < 0.001) and 1.55 (95%CI: 1.27–1.88, p < 0.001), respectively. Serum TNFα level and insulin resistant status (HOMR-IR) were higher in RSA group than in control group (p = 0.038, 0.001, respectively). All the three gene SNPs except MTRR 66AG gene variant had detrimental effects on HOMA-IR (all p were < 0.05). RSA group who carried the MTHFR 677CT, TT, CT/TT genotypes and MTRR 66AG, AG/GG genotypes had detrimental effects on serum homocysteine levels, the MTHFR 677CT, CT/TT genotype carriers had favorable effects on serum folate levels, the MTHFR 677TT, CT/TT, 1298 AC, AC/CC genotype carriers had detrimental effects on serum low-density lipoprotein cholesterol (LDL-C) levels, and the MTRR 66AG genotype carriers had lower high-density lipoprotein cholesterol (HDL-C) levels than the AA genotype carriers (all p were < 0.05). Conclusions Interaction between the MTHFR C677T, A1298C and MTHFR A1298C, MTRR A66G are observed in our RSA group. Besides, all the three gene SNPs except MTRR 66AG gene variant had detrimental effects on HOMA-IR. MTHFR C677T and MTRR A66G gene variants had detrimental effects on serum homocysteine levels and insulin resistance status, while MTHFR C677T, A1298C and MTRR A66G gene variants had detrimental effects on certain serum lipid profiles.
Collapse
Affiliation(s)
- Zhong Lin
- Department of Obstetrics and Gynecology, Liuzhou Maternity and Child Health Care Hospital, 50 Yingshan Road, Liuzhou, 545001, Guangxi, China
| | - Qianxi Li
- Department of Obstetrics and Gynecology, The Maternal & Child Health Hospital of Guangxi Zhuang Autonomous Region, Guangxi, 530003, China
| | - Yifan Sun
- Department of Clinical Laboratory, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, 545001, Guangxi, China
| | - Jingchun Huang
- Department of Laboratory Science, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Wan Wang
- Department of Laboratory Science, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Jinjian Fu
- Department of Laboratory, Liuzhou Maternity and Child Health Care Hospital, Liuzhou, 545001, Guangxi, China
| | - Jianhua Xu
- Department of Laboratory Science, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China. .,Laboratory of Oncology Science and Molecular Biology, ShunDe Hospital of Guangzhou University of Chinese Medicine, Shunde, 528333, Guangdong, China.
| | - Dingyuan Zeng
- Department of Obstetrics and Gynecology, Liuzhou Maternity and Child Health Care Hospital, 50 Yingshan Road, Liuzhou, 545001, Guangxi, China.
| |
Collapse
|
1150
|
The Triglyceride-Glucose Index Predicts Coronary Artery Disease Severity and Cardiovascular Outcomes in Patients with Non-ST-Segment Elevation Acute Coronary Syndrome. DISEASE MARKERS 2019; 2019:6891537. [PMID: 31281548 PMCID: PMC6594265 DOI: 10.1155/2019/6891537] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/27/2019] [Accepted: 03/18/2019] [Indexed: 01/06/2023]
Abstract
Non-ST-segment elevation acute coronary syndrome (NSTE-ACS) is the leading cause of morbidity and mortality from cardiovascular disease worldwide. Several recent studies have shown the relationship between the triglyceride-glucose (TyG) index and vascular disease; however, the role of the TyG index in NSTE-ACS has not been extensively assessed. Thus, we aimed to investigate the association of the TyG index with cardiovascular risk factors and outcomes in NSTE-ACS. Overall, 438 patients with NSTE-ACS were enrolled to examine the association of the TyG index with the SYNTAX score and major adverse cardiovascular events (MACEs). The TyG index was calculated as ln (fasting triglyceride (mg/dL) × fasting glucose (mg/dL)/2). The severity of coronary lesions was quantified by the SYNTAX score. MACEs included cardiac death, nonfatal myocardial infarction, target vessel revascularization, congestive heart failure, and nonfatal stroke. All the patients underwent a 12-month follow-up for MACEs after admission. Multivariate regression analysis identified metabolic risk factors as independent parameters correlated with the TyG index. The prevalence of glucose metabolism disorder, metabolic syndrome, and MACEs increased with increasing TyG index. The TyG index showed a strong diagnostic performance for cardiovascular risk factors and was independently associated with the SYNTAX score (OR 6.055, 95% CI 2.915–12.579, P < 0.001). The risk of MACEs (12.8% and 22.8% for the low TyG index and high TyG index groups, respectively; adjusted HR = 1.791, 95% CI 1.045–3.068, P = 0.034) significantly increased in the high TyG index group as compared with the low TyG index group. The multivariate Cox regression analysis further revealed that the TyG index was an independent predictor of MACEs (HR 1.878, 95% CI 1.130–3.121, P = 0.015). In conclusion, the TyG index might be an independent predictor of coronary artery disease severity and cardiovascular outcomes in NSTE-ACS.
Collapse
|