1
|
Ghasemitarei M, Ghorbi T, Yusupov M, Zhang Y, Zhao T, Shali P, Bogaerts A. Effects of Nitro-Oxidative Stress on Biomolecules: Part 1-Non-Reactive Molecular Dynamics Simulations. Biomolecules 2023; 13:1371. [PMID: 37759771 PMCID: PMC10527456 DOI: 10.3390/biom13091371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Plasma medicine, or the biomedical application of cold atmospheric plasma (CAP), is an expanding field within plasma research. CAP has demonstrated remarkable versatility in diverse biological applications, including cancer treatment, wound healing, microorganism inactivation, and skin disease therapy. However, the precise mechanisms underlying the effects of CAP remain incompletely understood. The therapeutic effects of CAP are largely attributed to the generation of reactive oxygen and nitrogen species (RONS), which play a crucial role in the biological responses induced by CAP. Specifically, RONS produced during CAP treatment have the ability to chemically modify cell membranes and membrane proteins, causing nitro-oxidative stress, thereby leading to changes in membrane permeability and disruption of cellular processes. To gain atomic-level insights into these interactions, non-reactive molecular dynamics (MD) simulations have emerged as a valuable tool. These simulations facilitate the examination of larger-scale system dynamics, including protein-protein and protein-membrane interactions. In this comprehensive review, we focus on the applications of non-reactive MD simulations in studying the effects of CAP on cellular components and interactions at the atomic level, providing a detailed overview of the potential of CAP in medicine. We also review the results of other MD studies that are not related to plasma medicine but explore the effects of nitro-oxidative stress on cellular components and are therefore important for a broader understanding of the underlying processes.
Collapse
Affiliation(s)
- Maryam Ghasemitarei
- Department of Physics, Sharif University of Technology, Tehran 14588-89694, Iran
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
| | - Tayebeh Ghorbi
- Department of Physics, Sharif University of Technology, Tehran 14588-89694, Iran
| | - Maksudbek Yusupov
- School of Engineering, New Uzbekistan University, Tashkent 100007, Uzbekistan
- School of Engineering, Central Asian University, Tashkent 111221, Uzbekistan
- Laboratory of Thermal Physics of Multiphase Systems, Arifov Institute of Ion-Plasma and Laser Technologies, Academy of Sciences of Uzbekistan, Tashkent 100125, Uzbekistan
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
| | - Yuantao Zhang
- School of Electrical Engineering, Shandong University, Jinan 250061, China
| | - Tong Zhao
- School of Electrical Engineering, Shandong University, Jinan 250061, China
| | - Parisa Shali
- Research Unit Plasma Technology, Department of Applied Physics, Faculty of Engineering and Agriculture, Ghent University, 9000 Ghent, Belgium
| | - Annemie Bogaerts
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
2
|
A New Electrocatalytic System Based on Copper (II) Chloride and Magnetic Molecularly Imprinted Polymer Nanoparticles in 3D Printed Microfluidic Flow Cell for Enzymeless and Low-Potential Cholesterol Detection. J Electroanal Chem (Lausanne) 2022. [DOI: 10.1016/j.jelechem.2022.116853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
3
|
Baila-Rueda L, Cenarro A, Lamiquiz-Moneo I, Marco-Benedi V, Gracia-Rubio I, Casamayor-Franco MC, Arbones-Mainar JM, Civeira F, Laclaustra M. Association of Cholesterol and Oxysterols in Adipose Tissue With Obesity and Metabolic Syndrome Traits. J Clin Endocrinol Metab 2022; 107:e3929-e3936. [PMID: 35453148 DOI: 10.1210/clinem/dgac188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Adipose tissue stores a substantial amount of body cholesterol in humans. Obesity is associated with decreased concentrations of serum cholesterol. During weight gain, adipose tissue dysfunction might be one of the causes of metabolic syndrome. The aim of this study is to evaluate cholesterol storage and oxidized metabolites in adipose tissue and their relationship with metabolic clinical characteristics. METHODS Concentrations of cholesterol and oxysterols (27-hydroxycholesterol and 24S-hydroxycholesterol) in subcutaneous and visceral adipose tissue were determined by high-performance liquid chromatography with tandem mass spectrometry in 19 adult women with body mass index between 23 and 40 kg/m2 from the FAT expandability (FATe) study. Tissue concentration values were correlated with biochemical and clinical characteristics using nonparametric statistics. RESULTS Insulin correlated directly with 24S-hydroxycholesterol in both adipose tissues and with 27-hydroxycholesterol in visceral tissue. Leptin correlated directly with 24S-hydroxycholesterol in subcutaneous adipose tissue. Tissue cholesterol correlated directly with 27-hydroxycholesterol in both adipose tissues and with 24S-hydroxycholesterol in visceral tissue, where cholesterol correlation with 24S-hydroxycholesterol was higher than with 27-hydroxycholesterol. In addition, some tendencies were observed: serum high-density lipoprotein cholesterol tended to be inversely correlated with visceral adipose tissue cholesterol; high-sensitivity C-reactive protein tended to be correlated directly with subcutaneous adipose 24S-hydroxycholesterol and inversely with visceral 27-hydroxycholesterol. CONCLUSIONS Adipose tissue oxysterols are associated with blood insulin and insulin resistance. Tissue cholesterol correlated more with 27-hydroxycholesterol in subcutaneous adipose tissue and with 24S-hydroxycholesterol in visceral adipose tissue. Levels of adipose 24S-hydroxycholesterol seem to be correlated with some metabolic syndrome symptoms and inflammation while adipose 27-hydroxycholesterol could represent some protection against them.
Collapse
Affiliation(s)
- Lucia Baila-Rueda
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Cenarro
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Itziar Lamiquiz-Moneo
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Zaragoza, Zaragoza, Spain
| | - Victoria Marco-Benedi
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Gracia-Rubio
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
| | - Maria Carmen Casamayor-Franco
- Servicio de Cirugía General y Aparato Digestivo, Unidad de Cirugía Endocrina, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Jose M Arbones-Mainar
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
- Adipocyte and Fat Biology Laboratory (AdipoFat), Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Centro de Investigación Biomédica en Red Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Fernando Civeira
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Zaragoza, Zaragoza, Spain
| | - Martin Laclaustra
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Zaragoza, Zaragoza, Spain
| |
Collapse
|
4
|
Li B, Cheng X, Aierken A, Du J, He W, Zhang M, Tan N, Kou Z, Peng S, Jia W, Tang H, Hua J. Melatonin Promotes the Therapeutic Effect of Mesenchymal Stem Cells on Type 2 Diabetes Mellitus by Regulating TGF-β Pathway. Front Cell Dev Biol 2021; 9:722365. [PMID: 34722505 PMCID: PMC8554153 DOI: 10.3389/fcell.2021.722365] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/27/2021] [Indexed: 12/26/2022] Open
Abstract
Abundant evidence proves the therapeutic effect of adipose-derived mesenchymal stem cells (ADMSCs) in the treatment of diabetes mellitus. However, the problems have not been solved that viability of ADMSCs were inconsistent and the cells quickly undergo senescence after in vitro cell culture. In addition, the therapeutic effect of ADMSCs is still not satisfactory. In this study, melatonin (MLT) was added to canine ADMSC culture medium, and the treated cells were used to treat type 2 diabetes mellitus (T2DM). Our research reveals that adding MLT to ADMSC culture medium can promote the viability of ADMSCs. This effect depends on the binding of MLT and MLT receptors, which activates the transforming growth factor β (TGF-β) pathway and then changes the cell cycle of ADMSCs and improves the viability of ADMSCs. Since ADMSCs were found to be used to treat T2DM by anti-inflammatory and anti-endoplasmic reticulum (ER) stress capabilities, our data demonstrate that MLT augment several effects of ADMSCs in remission hyperglycemia, insulin resistance, and liver glycogen metabolism in T2DM patients. This suggest that ADMSCs and MLT-ADMSCs is safe and vabulable for pet clinic.
Collapse
Affiliation(s)
- Balun Li
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Xuedi Cheng
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Aili Aierken
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Jiaxin Du
- Department of Animal Engineering, Yangling Vocational and Technical College, Xianyang, China.,Department of Veterinary Medicine, College of Animal Sciences, Institute of Preventive Veterinary Sciences, Zhejiang University, Hangzhou, China
| | - Wenlai He
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Mengfei Zhang
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Ning Tan
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Zheng Kou
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Sha Peng
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Wenwen Jia
- Shanghai East Hospital, East Hospital Affiliated to Tongji University, Shanghai, China
| | - Haiyang Tang
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Jinlian Hua
- Shaanxi Centre of Stem Cells Engineering and Technology, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| |
Collapse
|
5
|
Nonalcoholic Fatty Liver Disease: Focus on New Biomarkers and Lifestyle Interventions. Int J Mol Sci 2021; 22:ijms22083899. [PMID: 33918878 PMCID: PMC8069944 DOI: 10.3390/ijms22083899] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered a hepatic manifestation of metabolic syndrome, characterized from pathological changes in lipid and carbohydrate metabolism. Its main characteristics are excessive lipid accumulation and oxidative stress, which create a lipotoxic environment in hepatocytes leading to liver injury. Recently, many studies have focused on the identification of the genetic and epigenetic modifications that also contribute to NAFLD pathogenesis and their prognostic implications. The present review is aimed to discuss on cellular and metabolic alterations associated with NAFLD, which can be helpful to identify new noninvasive biomarkers. The identification of accumulated lipids in the cell membranes, as well as circulating cytokeratins and exosomes, provides new insights in understanding of NAFLD. This review also suggests that lifestyle modifications remain the main prevention and/or treatment for NAFLD.
Collapse
|
6
|
Tian J, Fu G, Xu Z, Chen X, Sun J, Jin B. Urinary exfoliated tumor single-cell metabolomics technology for establishing a drug resistance monitoring system for bladder cancer with intravesical chemotherapy. Med Hypotheses 2020; 143:110100. [DOI: 10.1016/j.mehy.2020.110100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022]
|
7
|
Hashizume Y, Tandia M. The reduction impact of monoglucosyl rutin on abdominal visceral fat: A randomized, placebo-controlled, double-blind, parallel-group. J Food Sci 2020; 85:3577-3589. [PMID: 32935866 DOI: 10.1111/1750-3841.15429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/29/2020] [Accepted: 08/10/2020] [Indexed: 11/30/2022]
Abstract
Water soluble α-glycosylated rutin (4G-α-D-glucopyranosyl rutin, monoglucosyl rutin, MR) was used in this study to evaluate its ability to reduce abdominal visceral fat (AVF). We conducted a study examining 66 healthy Japanese men and women with a body mass index of ≥23 and <30 kg/m2 for 8 weeks. The subjects were randomly assigned to groups via computer random numbers as follows: MR200 group (MR 200 mg/day), MR400 group (MR 400mg/day), or placebo group. The primary outcome was change in the AVF area after 8 weeks of intervention. The secondary outcomes were effects of MR on total fat and subcutaneous fat of umbilical area, lipid-related markers, and subjective symptoms. The per-protocol set analysis involved 18 subjects in the placebo group (7 males and 11 females), 20 subjects in the MR200 group (8 males and 12 females), and 20 subjects in the MR400 group (8 males and 12 females). AVF area in both the MR200 and MR400 groups was reduced at week 8, with changes from the baseline (week 0) significantly higher than the placebo group. Additionally, the MR400 group reported improved subjective symptoms concerning being "worried about abdominal fat" at week 4 compared with the placebo group. These results indicate that the consumption of MR (200 and 400 mg/day) for 8 weeks reduced AVF. PRACTICAL APPLICATION: Monoglucosyl rutin, an enzymatically modified form of rutin, is a highly stable and water-soluble flavonoid widely used in food and beverages to prevent oxidation. The present clinical study demonstrated that it may improve overall health by reducing abdominal visceral fat.
Collapse
Affiliation(s)
- Yushi Hashizume
- Toyo Sugar Refining Co., Ltd, Yoto Bldg., 18-20, Nihombashi-Koamicho, Chuo-ku, Tokyo, 103-0016, Japan
| | - Mahamadou Tandia
- Toyo Sugar Refining Co., Ltd, Yoto Bldg., 18-20, Nihombashi-Koamicho, Chuo-ku, Tokyo, 103-0016, Japan
| |
Collapse
|
8
|
Biondo LA, Teixeira AAS, de O. S. Ferreira KC, Neto JCR. Pharmacological Strategies for Insulin Sensitivity in Obesity and Cancer: Thiazolidinediones and Metformin. Curr Pharm Des 2020; 26:932-945. [PMID: 31969093 DOI: 10.2174/1381612826666200122124116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/21/2019] [Indexed: 12/19/2022]
Abstract
Background:
Chronic diseases, such as obesity and cancer, have high prevalence rates. Both diseases
have hyperinsulinemia, hyperglycemia, high levels of IGF-1 and inflammatory cytokines in common. Therefore,
these can be considered triggers for cancer development and growth. In addition, low-grade inflammation that
modulates the activation of immune cells, cellular metabolism, and production of cytokines and chemokines are
common in obesity, cancer, and insulin resistance. Pharmacological strategies are necessary when a change in
lifestyle does not improve glycemic homeostasis. In this regard, thiazolidinediones (TZD) possess multiple molecular
targets and regulate PPARγ in obesity and cancer related to insulin resistance, while metformin acts
through the AMPK pathway.
Objective:
The aim of this study was to review TZD and metformin as pharmacological treatments for insulin
resistance associated with obesity and cancer.
Conclusions:
Thiazolidinediones restored adiponectin secretion and leptin sensitivity, reduced lipid droplets in
hepatocytes and orexigen peptides in the hypothalamus. In cancer cells, TZD reduced proliferation, production of
reactive oxygen species, and inflammation by acting through the mTOR and NFκB pathways. Metformin has
similar effects, though these are AMPK-dependent. In addition, both drugs can be efficient against certain side
effects caused by chemotherapy.
Collapse
Affiliation(s)
- Luana A. Biondo
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Alexandre A. S. Teixeira
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Karen C. de O. S. Ferreira
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jose C. R. Neto
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
9
|
Svegliati-Baroni G, Pierantonelli I, Torquato P, Marinelli R, Ferreri C, Chatgilialoglu C, Bartolini D, Galli F. Lipidomic biomarkers and mechanisms of lipotoxicity in non-alcoholic fatty liver disease. Free Radic Biol Med 2019; 144:293-309. [PMID: 31152791 DOI: 10.1016/j.freeradbiomed.2019.05.029] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/13/2019] [Accepted: 05/27/2019] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents the most common form of chronic liver disease worldwide (about 25% of the general population) and 3-5% of patients develop non-alcoholic steatohepatitis (NASH), characterized by hepatocytes damage, inflammation and fibrosis, which increase the risk of developing liver failure, cirrhosis and hepatocellular carcinoma. The pathogenesis of NAFLD, particularly the mechanisms whereby a minority of patients develop a more severe phenotype, is still incompletely understood. In this review we examine the available literature on initial mechanisms of hepatocellular damage and inflammation, deriving from toxic effects of excess lipids. Accumulating data indicate that the total amount of triglycerides stored in the liver cells is not the main determinant of lipotoxicity and that specific lipid classes act as damaging agents. These lipotoxic species affect the cell behavior via multiple mechanisms, including activation of death receptors, endoplasmic reticulum stress, modification of mitochondrial function and oxidative stress. The gut microbiota, which provides signals through the intestine to the liver, is also reported to play a key role in lipotoxicity. Finally, we summarize the most recent lipidomic strategies utilized to explore the liver lipidome and its modifications in the course of NALFD. These include measures of lipid profiles in blood plasma and erythrocyte membranes that can surrogate to some extent lipid investigation in the liver.
Collapse
Affiliation(s)
- Gianluca Svegliati-Baroni
- Department of Gastroenterology, Università Politecnica Delle Marche, Ancona, Italy; Obesity Center, Università Politecnica Delle Marche, Ancona, Italy.
| | - Irene Pierantonelli
- Department of Gastroenterology, Università Politecnica Delle Marche, Ancona, Italy; Department of Gastroenterology, Senigallia Hospital, Senigallia, Italy
| | | | - Rita Marinelli
- Department of Pharmaceutical Sciences, University of Perugia, Italy
| | - Carla Ferreri
- ISOF, Consiglio Nazionale Delle Ricerche, Via P. Gobetti 101, 40129, Bologna, Italy
| | | | | | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Italy
| |
Collapse
|
10
|
Wishart DS. Metabolomics for Investigating Physiological and Pathophysiological Processes. Physiol Rev 2019; 99:1819-1875. [PMID: 31434538 DOI: 10.1152/physrev.00035.2018] [Citation(s) in RCA: 554] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Metabolomics uses advanced analytical chemistry techniques to enable the high-throughput characterization of metabolites from cells, organs, tissues, or biofluids. The rapid growth in metabolomics is leading to a renewed interest in metabolism and the role that small molecule metabolites play in many biological processes. As a result, traditional views of metabolites as being simply the "bricks and mortar" of cells or just the fuel for cellular energetics are being upended. Indeed, metabolites appear to have much more varied and far more important roles as signaling molecules, immune modulators, endogenous toxins, and environmental sensors. This review explores how metabolomics is yielding important new insights into a number of important biological and physiological processes. In particular, a major focus is on illustrating how metabolomics and discoveries made through metabolomics are improving our understanding of both normal physiology and the pathophysiology of many diseases. These discoveries are yielding new insights into how metabolites influence organ function, immune function, nutrient sensing, and gut physiology. Collectively, this work is leading to a much more unified and system-wide perspective of biology wherein metabolites, proteins, and genes are understood to interact synergistically to modify the actions and functions of organelles, organs, and organisms.
Collapse
Affiliation(s)
- David S Wishart
- Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
11
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
12
|
Herath RP, Siriwardana SR, Ekanayake CD, Abeysekara V, Kodithuwakku SUA, Herath HP. Non-alcoholic fatty liver disease and pregnancy complications among Sri Lankan women: A cross sectional analytical study. PLoS One 2019; 14:e0215326. [PMID: 30978266 PMCID: PMC6461248 DOI: 10.1371/journal.pone.0215326] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/30/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the commonest cause of liver disease worldwide and is the hepatic manifestation of metabolic syndrome. Effects of NAFLD on pregnancy is still unclear with few studies showing an association to gestational diabetes and pre-eclampsia. We aimed to describe the association between the NAFLD and pregnancy complications. This is the first study, to our knowledge, in a South Asian population. METHOD A cross sectional analytical study was done in Teaching Hospital, Ragama, Sri Lanka. Women carrying a singleton pregnancy, admitted for delivery were assessed for NAFLD with liver ultrasound scan. Data were extracted from interviewer administered questionnaire and antenatal and inpatient records. Pregnancy complications and labour outcomes were compared between the women with NAFLD and women without NAFLD (non-NAFLD). RESULTS Out of the 573 women who participated, 18.2% (n = 104) were found to have NAFLD. Out of them, 58 (55.8%), 32(30.8%), and 14(13.5%) had fatty liver grade 1,2 and 3 respectively. Women with NAFLD were 2 times more likely to develop gestational hypertension and pre-eclampsia compared to the women in the non-NAFLD group, after adjusting for BMI, age and Hyperglycaemia in pregnancy [Adjusted OR 2.09, (95% CI 1.07-4.10)]. There was no association between the grade of steatosis and a composite outcome of gestational hypertension and pre-eclampsia, within the NAFLD group. Composite outcome of gestational diabetes mellitus and diabetes in pregnancy diagnosed during pregnancy was a significant complication in the NAFLD group compared to non-NAFLD group in the bivariate analysis (27.2% vs 17.7%; p<0.05), but the significance disappeared after adjusting for confounders. The current study did not demonstrate a significant association between NAFLD with preterm labour, caesarean section rate, low birth weight, and Apgar score of the baby. CONCLUSION Women with NAFLD had a 2-fold higher risk of developing gestational hypertension and pre-eclampsia during pregnancy compared to women without NAFLD, after controlling for other confounding variables.
Collapse
Affiliation(s)
- Rasika Pradeep Herath
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Shirom R. Siriwardana
- Department of Anatomy, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Chanil D. Ekanayake
- Department of Clinical Sciences, General Sir John Kotelawala Defence University, Ratmalana, Sri Lanka
| | - Vikum Abeysekara
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Sajith U. A. Kodithuwakku
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Himali P. Herath
- Department of Community Medicine, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| |
Collapse
|
13
|
Zhou Y, Deng T, Zhang H, Guan Q, Zhao H, Yu C, Shao S, Zhao M, Xu J. Hypercholesterolaemia increases the risk of high‑turnover osteoporosis in men. Mol Med Rep 2019; 19:4603-4612. [PMID: 30957178 PMCID: PMC6522796 DOI: 10.3892/mmr.2019.10131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 03/08/2019] [Indexed: 01/23/2023] Open
Abstract
As the incidence of osteoporosis (OP) and hypercholesterolaemia in men has increased, male OP has drawn more attention from clinicians worldwide. The present study sought to investigate the effects of cholesterol on male bone. Between July 2015 and October 2015, 216 men (aged ≥18 years) were recruited for this cross‑sectional study. To test our clinical hypothesis, we designed two male animal models: Exogenous hypercholesterolaemia induced by a high‑cholesterol diet (HCD) and endogenous hypercholesterolaemia induced by apolipoprotein E (ApoE) knockout. Finally, the direct effects of cholesterol on osteoblasts were observed in cell experiments. In our clinical studies, men with hypercholesterolaemia displayed a lower bone mineral density (BMD) and increased beta collagen cross‑linking (beta‑CTX) and type I anterior collagen amino terminal peptide (PINP) levels compared to those of the control subjects. Serum cholesterol levels were a significant independent predictor of BMD, beta‑CTX and PINP and were negatively correlated with BMD and positively correlated with beta‑CTX and PINP levels. Our animal experimental results validated our clinical results, as they also indicated that hypercholesterolaemia damages bone microstructure and reduces bone strength. Cholesterol directly increased osteoblast functional gene expression in vitro. Hypercholesterolaemia increases the risk of high‑turnover osteoporosis in men at least in part by excessively promoting the activity of the remodelling pathway. In addition, hypercholesterolaemia damages the bone microstructure, resulting in osteopenia or OP and reduced bone strength, leading to a higher risk of fracture in men. We emphasize the importance of preventing and treating hypercholesterolaemia as well as monitoring bone metabolic markers and BMD in men with hypercholesterolemia for the effective prevention of bone loss and subsequent fracture. In addition, our findings provide a theoretical basis for the development of treatments for high cholesterol‑induced osteoporosis in men.
Collapse
Affiliation(s)
- Yanman Zhou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Tuo Deng
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Haiqing Zhang
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Qingbo Guan
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hongqiang Zhao
- Department of Internal Medicine, Laiwu People's Hospital, Laiwu, Shandong 271100, P.R. China
| | - Chunxiao Yu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shanshan Shao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Meng Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Jin Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
14
|
Maslov LN, Naryzhnaya NV, Boshchenko AA, Popov SV, Ivanov VV, Oeltgen PR. Is oxidative stress of adipocytes a cause or a consequence of the metabolic syndrome? JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY 2018; 15:1-5. [PMID: 30479968 PMCID: PMC6240632 DOI: 10.1016/j.jcte.2018.11.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/02/2018] [Accepted: 11/08/2018] [Indexed: 01/17/2023]
Abstract
Metabolic syndrome is accompanied by oxidative stress in animals and humans. The main source of ROS in experimental metabolic syndrome is NADPH oxidase and possibly adipocyte mitochondria. It is now documented that oxidative stress induces insulin resistance of adipocytes and increases secretion of leptin, MCP-1, IL-6, and TNF-α by adipocytes. It was established that oxidative stress induces a decrease in adiponectin production by adipocytes. It has also been shown that obesity itself can induce oxidative stress. Oxidative stress can cause an alteration of intracellular signaling in adipocytes that apparently leads to the formation of insulin resistance of adipocytes. Chronic stress, glucocorticoids, mineralocorticoids, angiotensin-II, TNF-α also play an important role in the pathogenesis of oxidative stress of adipocytes. Oxidative stress is not only a consequence of metabolic syndrome, but also a reason and a foundational link in the pathogenesis of the metabolic syndrome.
Collapse
Affiliation(s)
- Leonid N Maslov
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Natalia V Naryzhnaya
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Alla A Boshchenko
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Sergey V Popov
- Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | | | - Peter R Oeltgen
- Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
15
|
Russo A, Bartolini D, Mensà E, Torquato P, Albertini MC, Olivieri F, Testa R, Rossi S, Piroddi M, Cruciani G, De Feo P, Galli F. Physical Activity Modulates the Overexpression of the Inflammatory miR-146a-5p in Obese Patients. IUBMB Life 2018; 70:1012-1022. [PMID: 30212608 DOI: 10.1002/iub.1926] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/27/2018] [Accepted: 06/11/2018] [Indexed: 01/30/2023]
Abstract
Specific microRNAs (miRs), including the "angio-miR-126" and the "inflamma-miR-146a-5p," have been proposed as biomarkers and even therapeutic targets of obesity-associated metabolic diseases. Physical activity, a key measure of prevention for obesity and its complications, is reported to influence the expression of these miRs. In this study, we investigate whether a physical activity program proven to improve metabolic parameters in obese patients can correct the circulating levels of these miRs. Plasma miR-126 and miR-146a-5p were measured in a cohort of obese patients (n = 31, 16F + 15M) before and after the 3-month physical activity program of the CURIAMO trial (registration number for clinical trials: ACTRN12611000255987) and in 37 lean controls (24F + 13M). miR-146a-5p, but not miR-126, was significantly increased in obese patients as compared with lean controls and decreased in approximately two-thirds of the participants post-intervention with a response that positively correlated with pre-intervention levels of this miR. Waist circumference, the inflammatory cytokine IL-8 and lipid parameters, principally total cholesterol, showed the strongest correlation with both the baseline levels and post-intervention correction of miR-146a-5p. Post-hoc analysis of experimental data supports the use of miR-146a-5p as a biomarker and predictor of the clinical response to physical activity in obese patients. Furthermore, miR-146a-5p expression was confirmed to increase together with that of the inflammatory genes TLR4, NF-κB, IL-6, and TNF-α in LPS-stimulated human mononuclear leukocytes. In conclusion, the inflamma-miR-146a-5p can serve as a personalized predictor of clinical outcome in obese patients entering physical activity weight-reduction programs. © 2018 IUBMB Life, 70(10):1012-1022, 2018.
Collapse
Affiliation(s)
- Angelo Russo
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy.,"Centro Universitario di Ricerca Interdipartimentale sull' Attività Motoria", University of Perugia, Perugia, Italy
| | - Desireé Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Emanuela Mensà
- Experimental Models in Clinical Pathology, IRCCS INRCA National Institute, Ancona, Italy
| | - Pierangelo Torquato
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Fabiola Olivieri
- Center of Clinical Pathology and Innovative Therapy, Italian National Research Center on Aging (IRCCS INRCA), Ancona, Italy.,Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Roberto Testa
- Experimental Models in Clinical Pathology, IRCCS INRCA National Institute, Ancona, Italy
| | - Sara Rossi
- Center of Clinical Pathology and Innovative Therapy, Italian National Research Center on Aging (IRCCS INRCA), Ancona, Italy
| | - Marta Piroddi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Pierpaolo De Feo
- "Centro Universitario di Ricerca Interdipartimentale sull' Attività Motoria", University of Perugia, Perugia, Italy
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
16
|
Sadie-Van Gijsen H. Adipocyte biology: It is time to upgrade to a new model. J Cell Physiol 2018; 234:2399-2425. [PMID: 30192004 DOI: 10.1002/jcp.27266] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/25/2018] [Indexed: 12/15/2022]
Abstract
Globally, the obesity pandemic is profoundly affecting quality of life and economic productivity, but efforts to address this, especially on a pharmacological level, have generally proven unsuccessful to date, serving as a stark demonstration that our understanding of adipocyte biology and pathophysiology is incomplete. To deliver better insight into adipocyte function and obesity, we need improved adipocyte models with a high degree of fidelity in representing the in vivo state and with a diverse range of experimental applications. Adipocyte cell lines, especially 3T3-L1 cells, have been used extensively over many years, but these are limited in terms of relevance and versatility. In this review, I propose that primary adipose-derived stromal/stem cells (ASCs) present a superior model with which to study adipocyte biology ex vivo. In particular, ASCs afford us the opportunity to study adipocytes from different, functionally distinct, adipose depots and to investigate, by means of in vivo/ex vivo studies, the effects of many different physiological and pathophysiological factors, such as age, body weight, hormonal status, diet and nutraceuticals, as well as disease and pharmacological treatments, on the biology of adipocytes and their precursors. This study will give an overview of the characteristics of ASCs and published studies utilizing ASCs, to highlight the areas where our knowledge is lacking. More comprehensive studies in primary ASCs will contribute to an improved understanding of adipose tissue, in healthy and dysfunctional states, which will enhance our efforts to more successfully manage and treat obesity.
Collapse
Affiliation(s)
- Hanél Sadie-Van Gijsen
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa.,Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Parow, South Africa
| |
Collapse
|
17
|
Sottero B, Leonarduzzi G, Testa G, Gargiulo S, Poli G, Biasi F. Lipid Oxidation Derived Aldehydes and Oxysterols Between Health and Disease. EUR J LIPID SCI TECH 2018. [DOI: 10.1002/ejlt.201700047] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Barbara Sottero
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| |
Collapse
|
18
|
Murdolo G, Bartolini D, Tortoioli C, Piroddi M, Torquato P, Galli F. Selenium and Cancer Stem Cells. Adv Cancer Res 2017; 136:235-257. [PMID: 29054420 DOI: 10.1016/bs.acr.2017.07.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Selenium (Se) is an essential micronutrient that functions as "redox gatekeeper" and homeostasis factor of normal and cancer cells. Epidemiology and experimental studies, in the last years suggested that both inorganic and organic forms of Se may have favorable health effects. In this regard, a protective action of Se on cellular systems that may help preventing cancer cell differentiation has been demonstrated, while the hypothesis that Se compounds may cure cancer and its metastatic diffusion appears speculative and is still a matter of investigation. Indeed, the overall actions of Se compounds in carcinogenesis are controversial. The recognition that cancer is a stem cell disease instigated major paradigm shifts in our basic understanding of cancer and attracted a great deal of interest. Although current treatment approaches in cancer are grounded in the need to kill the majority of cancer cells, targeting cancer stem cells (CSCs) may hold great potential in improving cancer treatment. In this respect, Se compounds have been demonstrated modulating numerous signaling pathways involved in CSC biology and these findings are now stimulating further research on optimal Se concentrations, most effective and cancer-specific Se compounds, and inherent pathways involved in redox and metabolic regulation of CSCs. In this review, we summarize the current knowledge about the effects of Se compounds on CSCs, by focusing on redox-dependent pathways and main gene regulation checkpoints that affect self-renewal, differentiation, and migration responses in this subpopulation of cancer cells.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Section of Internal Medicine, Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy.
| | | | - Cristina Tortoioli
- Section of Internal Medicine, Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy
| | | | | | | |
Collapse
|
19
|
Scheideler M, Herzig S, Georgiadi A. Endocrine and autocrine/paracrine modulators of brown adipose tissue mass and activity as novel therapeutic strategies against obesity and type 2 diabetes. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0043/hmbci-2017-0043.xml. [PMID: 28850545 DOI: 10.1515/hmbci-2017-0043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/28/2017] [Indexed: 12/17/2022]
Abstract
The dramatically increasing world-wide prevalence of obesity is recognized as a risk factor for the development of various diseases. The growing research on the role of adipose tissue in controlling energy homeostasis and insulin sensitivity has revealed that the promotion of brown adipose tissue (BAT) activity and the browning of white adipose tissue (WAT) leads to multiple health benefits and prevents obesity and type 2 diabetes (T2D). Inducible thermogenic adipocytes do exist in adult humans and are linked with increased energy combustion and lower body fat mass. Thus brown adipocytes are currently placed at the center of attention for novel therapeutic strategies against metabolic diseases such as obesity and diabetes. Besides the classical, norepinephrine-mediated sympathetic recruitment and activation of thermogenic adipocytes, a number of novel circulating factors have been recently identified to have a positive or negative impact on thermogenic adipocyte formation and activity. In this review their mechanism of action and the plausible therapeutic applications will be summarized and discussed.
Collapse
|
20
|
Oliveira PS, Chaves VC, Bona NP, Soares MSP, Cardoso JDS, Vasconcellos FA, Tavares RG, Vizzotto M, Silva LMCD, Grecco FB, Gamaro GD, Spanevello RM, Lencina CL, Reginatto FH, Stefanello FM. Eugenia uniflora fruit (red type) standardized extract: a potential pharmacological tool to diet-induced metabolic syndrome damage management. Biomed Pharmacother 2017; 92:935-941. [DOI: 10.1016/j.biopha.2017.05.131] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/22/2017] [Accepted: 05/28/2017] [Indexed: 11/25/2022] Open
|
21
|
Baret P, Le Sage F, Planesse C, Meilhac O, Devin A, Bourdon E, Rondeau P. Glycated human albumin alters mitochondrial respiration in preadipocyte 3T3-L1 cells. Biofactors 2017; 43:577-592. [PMID: 28543688 DOI: 10.1002/biof.1367] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/02/2017] [Accepted: 04/20/2017] [Indexed: 12/22/2022]
Abstract
Diabetes and obesity are strongly associated with increased levels of circulating advanced glycation end products (AGEs) and reactive oxygen species (ROS). These two molecular phenomena affect the physiology of adipose tissue, a biological driver of the metabolic syndrome, leading to an inflammatory profile and insulin resistance, which could contribute to obesity/diabetes-associated complications, such as cardiovascular diseases. Herein, we investigated the impact of AGEs on mitochondrial bioenergetics in murine preadipocyte cells (3T3-L1) and cellular redox homeostasis. We show that incubation of preadipocytes with AGEs stimulates mitochondrial activity and respiration while inducing oxidative stress. This AGE-induced intracellular ROS production was blocked by diphenylene iodonium, an NAD(P)H oxidase inhibitor. In parallel, antioxidant enzymes (catalase, superoxide dismutase, and glutathione peroxidase) were found to be activated upon AGE treatment. Our results suggest that AGE-induced oxidative stress is generated by NAD(P)H oxidase and leads to a cellular proliferation arrest associated with enhanced mitochondrial metabolism and biogenesis, and with increased levels of ROS-detoxifying enzymes, as well. These new data show how AGEs may be involved in hyperglycemia-induced oxidative damage in preadipocytes and their potential links to diabetes progression. © 2017 BioFactors, 43(4):577-592, 2017.
Collapse
Affiliation(s)
- Pascal Baret
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
| | - Fanny Le Sage
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
| | - Cynthia Planesse
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
| | - Olivier Meilhac
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
- CHU de La Réunion, Centre d'Investigation Clinique, Saint-Denis, France
| | - Anne Devin
- CNRS, Institut de Biochimie et Génétique Cellulaires, UMR 5095, Université de Bordeaux, Bordeaux, France
| | - Emmanuel Bourdon
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
| | - Philippe Rondeau
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde, France
- Université de La Réunion, UMR 1188, Sainte-Clotilde, France
| |
Collapse
|
22
|
Hosoyamada Y, Yamada M. Effects of Dietary Fish Oil and Apple Polyphenol on the Concentration Serum Lipids and Excretion of Fecal Bile Acids in Rats. J Nutr Sci Vitaminol (Tokyo) 2017; 63:21-27. [PMID: 28367922 DOI: 10.3177/jnsv.63.21] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We studied the effects of fish oil and apple polyphenol combined with a high cholesterol diet in rats, and assessed serum and liver lipids concentrations, serum oxidative stress and fecal bile acid excretion. Young male rats were fed a diet containing the control (Control), apple polyphenol (AP), fish oil (FO) or fish oil+apple polyphenol (FO+AP) for 4 wk. The control diet contained a lard component. Posterior abdominal wall fat and testicle peripheral fat weights decreased in the FO+AP group compared to the AP group. The concentration of total cholesterol in the serum and liver decreased in the FO group and the FO+AP group compared to the Control and the AP groups. The concentration of adiponectin and biological antioxidant potential in the serum increased in the FO group compared to the other groups. The diacron-reactive oxygen metabolites in serum decreased in the FO group and the FO+AP group compared to the Control and the AP groups. The bile acid excretion in feces increased in the AP group, the FO group and the FO+AP group compared to the Control group. These results suggested that the combination of fish oil and apple polyphenol in the diet improved serum and liver lipids, which should assist in the prevention and improvement of metabolic syndrome.
Collapse
Affiliation(s)
- Yasue Hosoyamada
- Department of Nutrition, Faculty of Health Care Sciences, Chiba Prefectural University of Health Sciences
| | | |
Collapse
|
23
|
Silva SF, Levy D, Ruiz JLM, de Melo TC, Isaac C, Fidelis ML, Rodrigues A, Bydlowski SP. Oxysterols in adipose tissue-derived mesenchymal stem cell proliferation and death. J Steroid Biochem Mol Biol 2017; 169:164-175. [PMID: 27133385 DOI: 10.1016/j.jsbmb.2016.04.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/22/2016] [Accepted: 04/27/2016] [Indexed: 11/26/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells characterized by self-renewal and cellular differentiation capabilities. Oxysterols comprise a very heterogeneous group derived from cholesterol through enzymatic and non-enzymatic oxidation. Potent effects in cell death processes, including cytoxicity and apoptosis induction, were described in several cell lines. Very little is known about the effects of oxysterols in MSCs. 7-ketocholesterol (7-KC), one of the most important oxysterols, was shown to be cytotoxic to human adipose tissue-derived MSCs. Here, we describe the short-term (24h) cytotoxic effects of cholestan-3α-5β-6α-triol, 3,5 cholestan-7-one, (3α-5β-6α)- cholestane-3,6-diol, 7-oxocholest-5-en-3β-yl acetate, and 5β-6β epoxy-cholesterol, on MSCs derived from human adipose tissue. MSCs were isolated from adipose tissue obtained from three young, healthy women. Oxysterols, with the exception of 3,5 cholestan-7-one and 7-oxocholest-5-en-3β-yl acetate, led to a complex mode of cell death that include apoptosis, necrosis and autophagy, depending on the type of oxysterol and concentration, being cholestan-3α-5β-6α-triol the most effective. Inhibition of proliferation was also promoted by these oxysterols, but no changes in cell cycle were observed.
Collapse
Affiliation(s)
- Suelen Feitoza Silva
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av.Dr.Enéas de Carvalho Aguiar, 155, 1st floor, room 43, 05403-000, São Paulo/SP, Brazil
| | - Débora Levy
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av.Dr.Enéas de Carvalho Aguiar, 155, 1st floor, room 43, 05403-000, São Paulo/SP, Brazil
| | - Jorge Luis Maria Ruiz
- Federal University of Latin American Integration-UNILA, Life and Nature Science Institute, Av. Tarquinio Joslin dos Santos, 1000, Sala 105, CEP: 85870-901, Foz do Iguacu, Parana/PR, Brazil
| | - Thatiana Correa de Melo
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av.Dr.Enéas de Carvalho Aguiar, 155, 1st floor, room 43, 05403-000, São Paulo/SP, Brazil
| | - Cesar Isaac
- Cell Culture and Wound Healing Research Laboratory, Division of Plastic Surgery, Hospital das Clínicas, University of São Paulo Medical School, Av. Dr. Arnaldo, 455, 1st floor, 05403-000, São Paulo/SP, Brazil
| | - Maíra Luísa Fidelis
- Department of Earth and Exact Sciences, Federal University of São Paulo, São Paulo/SP, Brazil
| | - Alessandro Rodrigues
- Department of Earth and Exact Sciences, Federal University of São Paulo, São Paulo/SP, Brazil
| | - Sérgio Paulo Bydlowski
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av.Dr.Enéas de Carvalho Aguiar, 155, 1st floor, room 43, 05403-000, São Paulo/SP, Brazil.
| |
Collapse
|
24
|
Dong Y, Xu Z, Zhang Z, Yin X, Lin X, Li H, Zheng F. Impaired adipose expansion caused by liver X receptor activation is associated with insulin resistance in mice fed a high-fat diet. J Mol Endocrinol 2017; 58:141-154. [PMID: 28258092 DOI: 10.1530/jme-16-0196] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 12/23/2022]
Abstract
Liver X receptors (LXR) are deemed as potential drug targets for atherosclerosis, whereas a role in adipose tissue expansion and its relation to insulin sensitivity remains unclear. To assess the metabolic effects of LXR activation by the dual LXRα/β agonist T0901317, C57BL/6 mice fed a high-fat diet (HFD) were treated with T0901317 (30 mg/kg once daily by intraperitoneal injection) for 3 weeks. Differentiated 3T3-L1 adipocytes were used for analysing the effect of T0901317 on glucose uptake. The following results were obtained from this study. T0901317 reduced fat mass, accompanied by a massive fatty liver and lower serum adipokine levels in HFD mice. Increased adipocyte apoptosis was found in epididymal fat of T0901317-treated HFD mice. In addition, T0901317 treatment promoted basal lipolysis, but blunted the anti-lipolytic action of insulin. Furthermore, LXR activation antagonised PPARγ target genes in epididymal fat and PPARγ-PPRE-binding activity in 3T3-L1 adipocytes. Although the glucose tolerance was comparable to that in HFD mice, the insulin response during IPGTT was significantly higher and the insulin tolerance was significantly impaired in T0901317-treated HFD mice, indicating decreased insulin sensitivity by T0901317 administration, and which was further supported by impaired insulin signalling found in epididymal fat and decreased insulin-induced glucose uptake in 3T3-L1 adipocytes by T0901317 administration. In conclusion, these findings reveal that LXR activation impairs adipose expansion by increasing adipocyte apoptosis, lipolysis and antagonising PPARγ-mediated transcriptional activity, which contributes to decreased insulin sensitivity in whole body. The potential of LXR activation being a therapeutic target for atherosclerosis might be limited by the possibility of exacerbating insulin resistance.
Collapse
Affiliation(s)
- Yueting Dong
- Department of EndocrinologySir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zhiye Xu
- Department of EndocrinologySir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ziyi Zhang
- Department of EndocrinologySir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xueyao Yin
- Department of EndocrinologySir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xihua Lin
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang ProvinceSir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hong Li
- Department of EndocrinologySir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Fenping Zheng
- Department of EndocrinologySir Run Run Shaw Hospital Affiliated with School of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
25
|
Piroddi M, Albini A, Fabiani R, Giovannelli L, Luceri C, Natella F, Rosignoli P, Rossi T, Taticchi A, Servili M, Galli F. Nutrigenomics of extra-virgin olive oil: A review. Biofactors 2017; 43:17-41. [PMID: 27580701 DOI: 10.1002/biof.1318] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 07/08/2016] [Accepted: 07/08/2016] [Indexed: 12/11/2022]
Abstract
Nutrigenomics data on the functional components of olive oil are still sparse, but rapidly increasing. Olive oil is the main source of fat and health-promoting component of the Mediterranean diet. Positive effects have been observed on genes involved in the pathobiology of most prevalent age- and lifestyle-related human conditions, such as cancer, cardiovascular disease and neurodegeneration. Other effects on health-promoting genes have been identified for bioactive components of olives and olive leafs. Omics technologies are offering unique opportunities to identify nutritional and health biomarkers associated with these gene responses, the use of which in personalized and even predictive protocols of investigation, is a main breakthrough in modern medicine and nutrition. Gene regulation properties of the functional components of olive oil, such as oleic acid, biophenols and vitamin E, point to a role for these molecules as natural homeostatic and even hormetic factors with applications as prevention agents in conditions of premature and pathologic aging. Therapeutic applications can be foreseen in conditions of chronic inflammation, and particularly in cancer, which will be discussed in detail in this review paper as major clinical target of nutritional interventions with olive oil and its functional components. © 2016 BioFactors, 43(1):17-41, 2017.
Collapse
Affiliation(s)
- Marta Piroddi
- Department of Pharmaceutical Sciences, Nutrition and Clinical Biochemistry Lab, University of Perugia, Italy
| | - Adriana Albini
- IRCCS MultiMedica, Scientific and Technology Pole, Milan, Italy
| | - Roberto Fabiani
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Italy
| | - Lisa Giovannelli
- NEUROFARBA - Section of Phamacology and Toxicology, University of Firenze, Italy
| | - Cristina Luceri
- NEUROFARBA - Section of Phamacology and Toxicology, University of Firenze, Italy
| | - Fausta Natella
- CREA-NUT, Consiglio per La Ricerca in Agricoltura E L'Analisi Dell'Economia Agraria, Food and Nutrition Research Centre, via Ardeatina 546, 00178, Roma, Italy
| | - Patrizia Rosignoli
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Italy
| | - Teresa Rossi
- Research and Statistics, Department, IRCCS "Tecnologie Avanzate E Modelli Assistenziali in Oncologia", Laboratory of Translational Research, Arcispedale S. Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - Agnese Taticchi
- Department of Agricultural Food and Environmental Sciences, University of Perugia, Italy
| | - Maurizio Servili
- Department of Agricultural Food and Environmental Sciences, University of Perugia, Italy
| | - Francesco Galli
- Department of Pharmaceutical Sciences, Nutrition and Clinical Biochemistry Lab, University of Perugia, Italy
| |
Collapse
|
26
|
Hydrogen Sulfide in the Adipose Tissue-Physiology, Pathology and a Target for Pharmacotherapy. Molecules 2016; 22:molecules22010063. [PMID: 28042862 PMCID: PMC6155731 DOI: 10.3390/molecules22010063] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/21/2016] [Accepted: 12/29/2016] [Indexed: 12/15/2022] Open
Abstract
Hydrogen sulfide (H₂S) is synthesized in the adipose tissue mainly by cystathionine γ-lyase (CSE). Several studies have demonstrated that H₂S is involved in adipogenesis, that is the differentiation of preadipocytes to adipocytes, most likely by inhibiting phosphodiesterases and increasing cyclic AMP concentration. The effect of H₂S on adipose tissue insulin sensitivity and glucose uptake is controversial. Some studies suggest that H₂S inhibits insulin-induced glucose uptake and that excess of H₂S contributes to adipose tissue insulin resistance in metabolic syndrome. In contrast, other studies have demonstrated that H₂S stimulates glucose uptake and its deficiency contributes to insulin resistance. Similarly, the effect of H₂S on adipose tissue lipolysis is controversial. H₂S produced by perivascular adipose tissue decreases vascular tone by activating ATP-sensitive and/or voltage-gated potassium channels in smooth muscle cells. Experimental obesity induced by high calorie diet has a time dependent effect on H₂S in perivascular adipose tissue; short and long-term obesity increase and decrease H₂S production, respectively. Hyperglycemia has been consistently demonstrated to suppress CSE-H₂S pathway in various adipose tissue depots. Finally, H₂S deficiency may contribute to adipose tissue inflammation associated with obesity/metabolic syndrome.
Collapse
|
27
|
Murdolo G, Piroddi M, Tortoioli C, Bartolini D, Schmelz M, Luchetti F, Canonico B, Papa S, Zerbinati C, Iuliano L, Galli F. Free Radical-derived Oxysterols: Novel Adipokines Modulating Adipogenic Differentiation of Adipose Precursor Cells. J Clin Endocrinol Metab 2016; 101:4974-4983. [PMID: 27710239 DOI: 10.1210/jc.2016-2918] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
CONTEXT Increased oxidative stress in adipose tissue emerges as an inducer of obesity-linked insulin resistance. Here we tested whether free-radical derived oxysterols are formed by, and accumulate in, human adipocytes. Moreover, we asked whether increased accumulation of oxysterols characterizes the adipose cells of obese patients with type 2 diabetes (T2D) (OBT2D) compared with lean, nondiabetic controls (CTRLs). Finally, we studied the effects of the free radical-derived oxysterols on adipogenic differentiation of adipose-derived stem cells (ASCs). MAIN OUTCOME MEASURES Adipocytes and ASCs were isolated from sc abdominal adipose tissue biopsy in four OBT2D and four CTRL subjects. Oxysterols in adipocytes were detected by gas chromatography/mass spectrometry. The cellular and molecular effects of oxysterols were then evaluated on primary cultures of ASCs focusing on cell viability, adipogenic differentiation, and "canonical" WNT and MAPK signaling pathways. RESULTS 7-ketocholesterol (7κ-C) and 7β-hydroxycholesterol were unambiguously detected in adipocytes, which showed higher oxysterol accumulation (P < .01) in OBT2D, as compared with CTRL individuals. Notably, the accumulation of oxysterols in adipocytes was predicted by the adipose cell size of the donor (R2 = 0.582; P < .01). Challenging ASCs with free radical-derived type I (7κ-C) and type II (5,6-Secosterol) oxysterols led to a time- and concentration-dependent decrease of cell viability. Meaningfully, at a non-toxic concentration (1μM), these bioactive lipids hampered adipogenic differentiation of ASCs by sequential activation of WNT/β-catenin, p38-MAPK, ERK1/2, and JNK signaling pathways. CONCLUSION Free radical-derived oxysterols accumulate in the "diabetic" fat and may act as novel adipokines modulating the adipogenic potential of undifferentiated adipose precursor cells.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Marta Piroddi
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Cristina Tortoioli
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Desirée Bartolini
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Martin Schmelz
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Francesca Luchetti
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Barbara Canonico
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Stefano Papa
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Chiara Zerbinati
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Luigi Iuliano
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Francesco Galli
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| |
Collapse
|
28
|
Dipeptidyl peptidase- IV inhibitor alogliptin improves stress-induced insulin resistance and prothrombotic state in a murine model. Psychoneuroendocrinology 2016; 73:186-195. [PMID: 27509090 DOI: 10.1016/j.psyneuen.2016.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/04/2016] [Accepted: 08/02/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Stress evokes lipolytic release of free fatty acid (FFA) and low-grade inflammation in visceral adipose tissue, mediated by increased adipokine secretion, and contributes to glucose metabolism disorder and prothrombotic state. We tested the hypothesis that alogliptin, a dipeptidyl peptidase-4 inhibitor, can ameliorate the biological effects of chronic stress in mice. METHOD AND RESULTS C57BL/6J mice were subjected to 2-week intermittent restraint stress and orally treated with vehicle or alogliptin (dose: 15 or 45mg/kg/day). Plasma levels of lipids, proinflammatory cytokines (monocyte chemoattractant protein-1, tumor necrosis factor-α, and interleukin-6), and 8-hydroxydeoxyguanosine were measured with enzyme-linked immunosorbent assay. Monocyte/macrophage accumulation in inguinal white adipose tissue (WAT) was examined by CD11b-positive cell count and mRNA expression of CD68 and F4/80 was examined by immunohistochemistry and RT-PCR, respectively. The mRNA levels of the above-mentioned proinflammatory cytokines, NADPH oxidase 4, adiponectin, and coagulation factors (plasminogen activation inhibitor-1 and tissue factor) in WAT were also assessed with RT-PCR. Glucose metabolism was assessed by glucose and insulin tolerance tests, plasma levels of DPP-4 activity, glucagon-like peptide-1, expression of DPP-4, insulin receptor substrate-1 and glucose transporter 4 in WAT and skeletal muscle. Alogliptin administration suppressed stress-induced FFA release, oxidative stress, adipose tissue inflammation, DPP-4 activation, and prothrombotic state in a dose-dependent manner, and improved insulin sensitivity in stressed mice. CONCLUSIONS The results indicate that alogliptin improves stress-induced prothrombotic state and insulin resistance; suggesting that alogliptin could have beneficial therapeutic effects against cardiovascular complications in diabetic patients under stress.
Collapse
|
29
|
Shimabukuro M, Okawa C, Yamada H, Yanagi S, Uematsu E, Sugasawa N, Kurobe H, Hirata Y, Kim-Kaneyama JR, Lei XF, Takao S, Tanaka Y, Fukuda D, Yagi S, Soeki T, Kitagawa T, Masuzaki H, Sato M, Sata M. The pathophysiological role of oxidized cholesterols in epicardial fat accumulation and cardiac dysfunction: a study in swine fed a high caloric diet with an inhibitor of intestinal cholesterol absorption, ezetimibe. J Nutr Biochem 2016; 35:66-73. [DOI: 10.1016/j.jnutbio.2016.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/29/2016] [Accepted: 05/19/2016] [Indexed: 12/25/2022]
|
30
|
Boyer F, Diotel N, Girard D, Rondeau P, Essop MF, Bourdon E. Enhanced oxidative stress in adipose tissue from diabetic mice, possible contribution of glycated albumin. Biochem Biophys Res Commun 2016; 473:154-160. [DOI: 10.1016/j.bbrc.2016.03.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 03/16/2016] [Indexed: 12/16/2022]
|
31
|
Black PN, Ahowesso C, Montefusco D, Saini N, DiRusso CC. Fatty Acid Transport Proteins: Targeting FATP2 as a Gatekeeper Involved in the Transport of Exogenous Fatty Acids. MEDCHEMCOMM 2016; 7:612-622. [PMID: 27446528 DOI: 10.1039/c6md00043f] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The fatty acid transport proteins (FATP) are classified as members of the Solute Carrier 27 (Slc27) family of proteins based on their ability to function in the transport of exogenous fatty acids. These proteins, when localized to the plasma membrane or at intracellular membrane junctions with the endoplasmic reticulum, function as a gate in the regulated transport of fatty acids and thus represent a therapeutic target to delimit the acquisition of fatty acids that contribute to disease as in the case of fatty acid overload. To date, FATP1, FATP2, and FATP4 have been used as targets in the selection of small molecule inhibitors with the goal of treating insulin resistance and attenuating dietary absorption of fatty acids. Several studies targeting FATP1 and FATP4 were based on the intrinsic acyl CoA synthetase activity of these proteins and not on transport directly. While several classes of compounds were identified as potential inhibitors of fatty acid transport, in vivo studies using a mouse model failed to provide evidence these compounds were effective in blocking or attenuating fatty acid transport. Studies targeting FATP2 employed a naturally occurring splice variant, FATP2b, which lacks intrinsic acyl CoA synthetase due to the deletion of exon 3, yet is fully functional in fatty acid transport. These studies identified two compounds, 5'-bromo-5-phenyl-spiro[3H-1,3,4-thiadiazole-2,3'-indoline]-2'-one), now referred to as Lipofermata, and 2-benzyl-3-(4-chlorophenyl)-5-(4-nitrophenyl)pyrazolo[1,5-a]pyrimidin-7(4H)-one, now called Grassofermata, that are effective fatty acid transport inhibitors both in vitro using a series of model cell lines and in vivo using a mouse model.
Collapse
Affiliation(s)
- Paul N Black
- Department of Biochemistry, University of Nebraska, Lincoln, NE
| | | | | | - Nipun Saini
- Department of Biochemistry, University of Nebraska, Lincoln, NE
| | | |
Collapse
|
32
|
Guillemot-Legris O, Mutemberezi V, Cani PD, Muccioli GG. Obesity is associated with changes in oxysterol metabolism and levels in mice liver, hypothalamus, adipose tissue and plasma. Sci Rep 2016; 6:19694. [PMID: 26795945 PMCID: PMC4726335 DOI: 10.1038/srep19694] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 12/16/2015] [Indexed: 12/21/2022] Open
Abstract
Oxysterols are bioactive lipids derived from cholesterol that are linked to inflammatory processes. Because obesity and metabolic syndrome are characterized by inflammation and altered cholesterol metabolism, we sought to investigate the variations of oxysterol levels and their metabolic pathways induced by obesity in the liver, hypothalamus, adipose tissue and plasma. To this end, we used diet-induced and genetic (ob/ob and db/db) models of obesity. Among the oxysterols measured, we found that 4β-oxysterol levels were consistently decreased in the high-fat diet study, at different time-points, and in the ob/ob model. Overall, we did not find any correlation between cytochromes mRNA expression and variations of oxysterol levels. We also measured the levels of hepatic primary bile acids, in these three models and found similar profiles between HFD and ob/ob mice. However, although they are downstream metabolites of oxysterols, the variations in bile acid levels did not reflect the variations of their precursors. Our data show that, when considering oxysterol metabolism, the high-fat diet and ob/ob models are more closely related when compared to the db/db model. However, we were able to discriminate between lean and obese phenotypes based on liver oxysterol (4β-hydroxycholesterol, 27- hydroxycholesterol, 7-hydroxycholestenone) levels and enzyme (CYP3A11, CYP27A1, CYP7A1) expression.
Collapse
Affiliation(s)
- Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Valentin Mutemberezi
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| |
Collapse
|
33
|
Mutemberezi V, Masquelier J, Guillemot-Legris O, Muccioli GG. Development and validation of an HPLC-MS method for the simultaneous quantification of key oxysterols, endocannabinoids, and ceramides: variations in metabolic syndrome. Anal Bioanal Chem 2015; 408:733-45. [DOI: 10.1007/s00216-015-9150-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/15/2015] [Accepted: 10/26/2015] [Indexed: 12/14/2022]
|
34
|
Li J, Papadopoulos V, Vihma V. Steroid biosynthesis in adipose tissue. Steroids 2015; 103:89-104. [PMID: 25846979 DOI: 10.1016/j.steroids.2015.03.016] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/20/2015] [Accepted: 03/24/2015] [Indexed: 12/25/2022]
Abstract
Tissue-specific expression of steroidogenic enzymes allows the modulation of active steroid levels in a local manner. Thus, the measurement of local steroid concentrations, rather than the circulating levels, has been recognized as a more accurate indicator of the steroid action within a specific tissue. Adipose tissue, one of the largest endocrine tissues in the human body, has been established as an important site for steroid storage and metabolism. Locally produced steroids, through the enzymatic conversion from steroid precursors delivered to adipose tissue, have been proven to either functionally regulate adipose tissue metabolism, or quantitatively contribute to the whole body's steroid levels. Most recently, it has been suggested that adipose tissue may contain the steroidogenic machinery necessary for the initiation of steroid biosynthesis de novo from cholesterol. This review summarizes the evidence indicating the presence of the entire steroidogenic apparatus in adipose tissue and discusses the potential roles of local steroid products in modulating adipose tissue activity and other metabolic parameters.
Collapse
Affiliation(s)
- Jiehan Li
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Department of Biochemistry, McGill University, Montreal, Canada
| | - Vassilios Papadopoulos
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Department of Biochemistry, McGill University, Montreal, Canada.
| | - Veera Vihma
- Folkhälsan Research Center, Helsinki, Finland; University of Helsinki and Helsinki University Central Hospital, Heart and Lung Center, Helsinki, Finland.
| |
Collapse
|
35
|
van der Burg JW, Allred EN, Kuban K, O'Shea TM, Dammann O, Leviton A. Maternal obesity and development of the preterm newborn at 2 years. Acta Paediatr 2015; 104:900-3. [PMID: 25982514 DOI: 10.1111/apa.13038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 04/14/2015] [Accepted: 05/07/2015] [Indexed: 11/28/2022]
Abstract
AIM To evaluate to what extent extremely preterm children (<28 weeks' gestational age) of overweight (BMI 25-29) or obese (BMI ≥30) women are at increased risk of adverse development at 2 years measured with the Bayley Scales of Infant Development II in a multicenter prospective cohort study. METHODS Heights and prepregnancy weights of the mothers of 852 preterm born children were collected and included in multinomial logistic regression models. RESULTS Compared to newborns born to mothers with normal BMIs, newborns of obese mothers, but not those of overweight mothers, were more likely to have Bayley Scales indices more than 3 standard deviations below the reference mean (mental: OR = 2.1; 95% CI: 1.3, 3.5) (motor: OR = 1.7; 95% CI: 1.1, 2.7). These associations were even more prominent in children who did not have the intermittent or sustained systemic inflammation profile previously shown to be associated with severely impaired development (mental: OR = 4.6; 95% CI: 1.6, 14) (motor: OR = 3.7; 95% CI: 1.5, 8.9). CONCLUSION Maternal obesity is associated with an increased risk of impaired offspring development. Some of this impaired development cannot be attributed to confounding due to immaturity, socio-economic correlates or neonatal systemic inflammation.
Collapse
Affiliation(s)
- Jelske W. van der Burg
- Health and Life Sciences; Faculty of Earth and Life Sciences; VU University; Amsterdam The Netherlands
- Department of Public Health and Community Medicine; Tufts University School of Medicine; Boston MA USA
| | - Elizabeth N. Allred
- Neuroepidemiology Unit; Department of Neurology; Boston Children's Hospital; Harvard University; Boston MA USA
| | - Karl Kuban
- Division of Neurology; Department of Pediatrics; Boston Medical Center and Boston University; Boston MA USA
| | - T Michael O'Shea
- Department of Pediatrics; Wake Forest University; Winston-Salem NC USA
| | - Olaf Dammann
- Department of Public Health and Community Medicine; Tufts University School of Medicine; Boston MA USA
- Neuroepidemiology Unit; Department of Neurology; Boston Children's Hospital; Harvard University; Boston MA USA
- Perinatal Epidemiology Unit; Hannover Medical School; Hannover Germany
| | - Alan Leviton
- Neuroepidemiology Unit; Department of Neurology; Boston Children's Hospital; Harvard University; Boston MA USA
| |
Collapse
|
36
|
Kulig W, Olżyńska A, Jurkiewicz P, Kantola AM, Komulainen S, Manna M, Pourmousa M, Vazdar M, Cwiklik L, Rog T, Khelashvili G, Harries D, Telkki VV, Hof M, Vattulainen I, Jungwirth P. Cholesterol under oxidative stress-How lipid membranes sense oxidation as cholesterol is being replaced by oxysterols. Free Radic Biol Med 2015; 84:30-41. [PMID: 25795515 DOI: 10.1016/j.freeradbiomed.2015.03.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/06/2015] [Accepted: 03/09/2015] [Indexed: 02/06/2023]
Abstract
The behavior of oxysterols in phospholipid membranes and their effects on membrane properties were investigated by means of dynamic light scattering, fluorescence spectroscopy, NMR, and extensive atomistic simulations. Two families of oxysterols were scrutinized-tail-oxidized sterols, which are mostly produced by enzymatic processes, and ring-oxidized sterols, formed mostly via reactions with free radicals. The former family of sterols was found to behave similar to cholesterol in terms of molecular orientation, roughly parallel to the bilayer normal, leading to increasing membrane stiffness and suppression of its membrane permeability. In contrast, ring-oxidized sterols behave quantitatively differently from cholesterol. They acquire tilted orientations and therefore disrupt the bilayer structure with potential implications for signaling and other biochemical processes in the membranes.
Collapse
Affiliation(s)
- Waldemar Kulig
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland.
| | - Agnieszka Olżyńska
- J. Heyrovský Institute of Physical Chemistry, Academy of Sciences of the Czech Republic, v. v. i., Dolejskova 3, 18223 Prague 8, Czech Republic
| | - Piotr Jurkiewicz
- J. Heyrovský Institute of Physical Chemistry, Academy of Sciences of the Czech Republic, v. v. i., Dolejskova 3, 18223 Prague 8, Czech Republic.
| | - Anu M Kantola
- Department of Physics and Chemistry, University of Oulu, P.O. Box 3000, FI-90014 Oulu, Finland
| | - Sanna Komulainen
- Department of Physics and Chemistry, University of Oulu, P.O. Box 3000, FI-90014 Oulu, Finland
| | - Moutusi Manna
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| | - Mohsen Pourmousa
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| | - Mario Vazdar
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland; Rudjer Bošković Institute, Division of Organic Chemistry and Biochemistry, POB 180, HR-10002 Zagreb, Croatia
| | - Lukasz Cwiklik
- J. Heyrovský Institute of Physical Chemistry, Academy of Sciences of the Czech Republic, v. v. i., Dolejskova 3, 18223 Prague 8, Czech Republic; Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 16610 Prague 6, Czech Republic.
| | - Tomasz Rog
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| | | | - Daniel Harries
- Institute of Chemistry and the Fritz Haber Research Center, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Ville-Veikko Telkki
- Department of Physics and Chemistry, University of Oulu, P.O. Box 3000, FI-90014 Oulu, Finland
| | - Martin Hof
- J. Heyrovský Institute of Physical Chemistry, Academy of Sciences of the Czech Republic, v. v. i., Dolejskova 3, 18223 Prague 8, Czech Republic
| | - Ilpo Vattulainen
- Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland; MEMPHYS-Center for Biomembrane Physics, University of Southern Denmark, Odense, Denmark
| | - Pavel Jungwirth
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 16610 Prague 6, Czech Republic; Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| |
Collapse
|
37
|
Weber D, Ni Z, Vetter D, Hoffmann R, Fedorova M. Electrochemical oxidation of cholesterol: An easy way to generate numerous oxysterols in short reaction times. EUR J LIPID SCI TECH 2015. [DOI: 10.1002/ejlt.201500101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Dieter Weber
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy; Leipzig University; Leipzig Germany
- Center for Biotechnology and Biomedicine (BBZ); Leipzig University; Leipzig Germany
| | - Zhixu Ni
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy; Leipzig University; Leipzig Germany
- Center for Biotechnology and Biomedicine (BBZ); Leipzig University; Leipzig Germany
| | | | - Ralf Hoffmann
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy; Leipzig University; Leipzig Germany
- Center for Biotechnology and Biomedicine (BBZ); Leipzig University; Leipzig Germany
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy; Leipzig University; Leipzig Germany
- Center for Biotechnology and Biomedicine (BBZ); Leipzig University; Leipzig Germany
| |
Collapse
|
38
|
Shim U, Kim HN, Sung YA, Kim HL. Pathway Analysis of Metabolic Syndrome Using a Genome-Wide Association Study of Korea Associated Resource (KARE) Cohorts. Genomics Inform 2014; 12:195-202. [PMID: 25705158 PMCID: PMC4330254 DOI: 10.5808/gi.2014.12.4.195] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/05/2014] [Accepted: 09/12/2014] [Indexed: 01/07/2023] Open
Abstract
Metabolic syndrome (MetS) is a complex disorder related to insulin resistance, obesity, and inflammation. Genetic and environmental factors also contribute to the development of MetS, and through genome-wide association studies (GWASs), important susceptibility loci have been identified. However, GWASs focus more on individual single-nucleotide polymorphisms (SNPs), explaining only a small portion of genetic heritability. To overcome this limitation, pathway analyses are being applied to GWAS datasets. The aim of this study is to elucidate the biological pathways involved in the pathogenesis of MetS through pathway analysis. Cohort data from the Korea Associated Resource (KARE) was used for analysis, which include 8,842 individuals (age, 52.2 ± 8.9 years; body mass index, 24.6 ± 3.2 kg/m(2)). A total of 312,121 autosomal SNPs were obtained after quality control. Pathway analysis was conducted using Meta-analysis Gene-Set Enrichment of Variant Associations (MAGENTA) to discover the biological pathways associated with MetS. In the discovery phase, SNPs from chromosome 12, including rs11066280, rs2074356, and rs12229654, were associated with MetS (p < 5 × 10(-6)), and rs11066280 satisfied the Bonferroni-corrected cutoff (unadjusted p < 1.38 × 10(-7), Bonferroni-adjusted p < 0.05). Through pathway analysis, biological pathways, including electron carrier activity, signaling by platelet-derived growth factor (PDGF), the mitogen-activated protein kinase kinase kinase cascade, PDGF binding, peroxisome proliferator-activated receptor (PPAR) signaling, and DNA repair, were associated with MetS. Through pathway analysis of MetS, pathways related with PDGF, mitogen-activated protein kinase, and PPAR signaling, as well as nucleic acid binding, protein secretion, and DNA repair, were identified. Further studies will be needed to clarify the genetic pathogenesis leading to MetS.
Collapse
Affiliation(s)
- Unjin Shim
- Department of Internal Medicine, Seoul Seonam Hospital, Ewha Womans University Medical Center, Seoul 158-070, Korea
| | - Han-Na Kim
- Department of Biochemistry, Ewha Womans University School of Medicine, Seoul 158-710, Korea
| | - Yeon-Ah Sung
- Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul 158-710, Korea
| | - Hyung-Lae Kim
- Department of Biochemistry, Ewha Womans University School of Medicine, Seoul 158-710, Korea
| |
Collapse
|
39
|
Dietary high-fat lard intake induces thyroid dysfunction and abnormal morphology in rats. Acta Pharmacol Sin 2014; 35:1411-20. [PMID: 25263336 DOI: 10.1038/aps.2014.82] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 07/05/2014] [Indexed: 12/11/2022] Open
Abstract
AIM Excess dietary fat intake can induce lipotoxicity in non-adipose tissues. The aim of this study was to observe the effects of dietary high-fat lard intake on thyroid in rats. METHODS Male Sprague-Dawley rats were fed a high-fat lard diet for 24 weeks, and then the rats were fed a normal control diet (acute dietary modification) or the high-fat lard diet for another 6 weeks. The serum lipid profile, total thyroxine (TT4), free thyroxine (FT4) and thyrotropin (TSH) levels were determined at the 12, 18, 24 and 30 weeks. High-frequency ultrasound scanning of the thyroid glands was performed at the 24 or 30 weeks. After the rats were sacrificed, the thyroid glands were collected for histological and immunohistochemical analyses. RESULTS The high-fat lard diet significantly increased triglyceride levels in both the serum and thyroid, and decreased serum TT4 and FT4 levels in parallel with elevated serum TSH levels. Ultrasonic imaging revealed enlarged thyroid glands with lowered echotexture and relatively heterogeneous features in the high-fat lard fed rats. The thyroid glands from the high-fat lard fed rats exhibited enlarged follicle cavities and flattened follicular epithelial cells under light microscopy, and dilated endoplasmic reticulum cisternae, twisted nuclei, fewer microvilli and secretory vesicles under transmission electron microscopy. Furthermore, the thyroid glands from the high-fat lard fed rats showed markedly low levels of thyroid hormone synthesis-related proteins TTF-1 and NIS. Acute dietary modification by withdrawal of the high-fat lard diet for 6 weeks failed to ameliorate the high-fat lard diet-induced thyroid changes. CONCLUSION Dietary high-fat lard intake induces significant thyroid dysfunction and abnormal morphology in rats, which can not be corrected by short-term dietary modification.
Collapse
|
40
|
Polycystic Ovary Syndrome as a Paradigm for Prehypertension, Prediabetes, and Preobesity. Curr Hypertens Rep 2014; 16:500. [DOI: 10.1007/s11906-014-0500-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
41
|
Machado MV, Cortez-Pinto H. Non-alcoholic fatty liver disease: what the clinician needs to know. World J Gastroenterol 2014; 20:12956-80. [PMID: 25278691 PMCID: PMC4177476 DOI: 10.3748/wjg.v20.i36.12956] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/21/2014] [Accepted: 05/25/2014] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most frequent cause of liver disease in the Western world. Furthermore, it is increasing worldwide, paralleling the obesity pandemic. Though highly frequent, only about one fifth of affected subjects are at risk of developing the progressive form of the disease, non-alcoholic steatohepatitis with fibrosis. Even in the latter, liver disease is slowly progressive, though, since it is so prevalent, it is already the third cause of liver transplantation in the United States, and it is predicted to get to the top of the ranking in few years. Of relevance, fatty liver is also associated with increased overall mortality and particularly increased cardiovascular mortality. The literature and amount of published papers on NAFLD is increasing as fast as its prevalence, which makes it difficult to keep updated in this topic. This review aims to summarize the latest knowledge on NAFLD, in order to help clinicians understanding its pathogenesis and advances on diagnosis and treatment.
Collapse
|
42
|
Luchetti F, Canonico B, Bartolini D, Arcangeletti M, Ciffolilli S, Murdolo G, Piroddi M, Papa S, Reiter RJ, Galli F. Melatonin regulates mesenchymal stem cell differentiation: a review. J Pineal Res 2014; 56:382-97. [PMID: 24650016 DOI: 10.1111/jpi.12133] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/14/2014] [Indexed: 12/12/2022]
Abstract
Among the numerous functions of melatonin, the control of survival and differentiation of mesenchymal stem cells (MSCs) has been recently proposed. MSCs are a heterogeneous population of multipotent elements resident in tissues such as bone marrow, muscle, and adipose tissue, which are primarily involved in developmental and regeneration processes, gaining thus increasing interest for tissue repair and restoration therapeutic protocols. Receptor-dependent and receptor-independent responses to melatonin are suggested to occur in these cells. These involve antioxidant or redox-dependent functions of this indolamine as well as secondary effects resulting from autocrine and paracrine responses. Inflammatory cytokines and adipokines, proangiogenic/mitogenic stimuli, and other mediators that influence the differentiation processes may affect the survival and functional integrity of these mesenchymal precursor cells. In this scenario, melatonin seems to regulate signaling pathways that drive commitment and differentiation of MSC into osteogenic, chondrogenic, adipogenic, or myogenic lineages. Common pathways suggested to be involved as master regulators of these processes are the Wnt/β-catenin pathway, the MAPKs and the, TGF-β signaling. In this respect melatonin emerges a novel and potential modulator of MSC lineage commitment and adipogenic differentiation. These and other aspects of the physiological and pharmacological effects of melatonin as regulator of MSC are discussed in this review.
Collapse
Affiliation(s)
- Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Levy D, Ruiz JLM, Celestino AT, Silva SF, Ferreira AK, Isaac C, Bydlowski SP. Short-term effects of 7-ketocholesterol on human adipose tissue mesenchymal stem cells in vitro. Biochem Biophys Res Commun 2014; 446:720-5. [PMID: 24491549 DOI: 10.1016/j.bbrc.2014.01.132] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 01/25/2014] [Indexed: 01/27/2023]
Abstract
Oxysterols comprise a very heterogeneous group derived from cholesterol through enzymatic and non-enzymatic oxidation. Among them, 7-ketocholesterol (7-KC) is one of the most important. It has potent effects in cell death processes, including cytoxicity and apoptosis induction. Mesenchymal stem cells (MSCs) are multipotent cells characterized by self-renewal and cellular differentiation capabilities. Very little is known about the effects of oxysterols in MSCs. Here, we describe the short-term cytotoxic effect of 7-ketocholesterol on MSCs derived from human adipose tissue. MSCs were isolated from adipose tissue obtained from two young, healthy women. After 24 h incubation with 7-KC, mitochondrial hyperpolarization was observed, followed by a slight increase in the level of apoptosis and changes in actin organization. Finally, the IC50 of 7-KC was higher in these cells than has been observed or described in other normal or cancer cell lines.
Collapse
Affiliation(s)
- Débora Levy
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil
| | - Jorge Luis Maria Ruiz
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil
| | - Andrea Turbuck Celestino
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil
| | - Suelen Feitoza Silva
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil
| | - Adilson Kleber Ferreira
- Departament of Immunology, Laboratory of Tumor Immunology, Institute of Biomedical Science, University of Sao Paulo, Av. Prof. Lineu Prestes, 1730-Room 254, 05508-900 São Paulo, SP, Brazil
| | - Cesar Isaac
- Cell Culture and Wound Healing Research Laboratory, Division of Plastic Surgery, University of São Paulo, Av. Dr. Arnaldo, 455, 1st Floor, 05403-000 São Paulo, SP, Brazil
| | - Sérgio Paulo Bydlowski
- Laboratory of Genetics and Molecular Hematology (LIM31), University of São Paulo School of Medicine, Av. Dr. Enéas de Carvalho Aguiar,155, 1st Floor, Room 43, 05403-000 São Paulo, SP, Brazil.
| |
Collapse
|