201
|
Abstract
Nuclear, casein kinase and cyclin-dependent kinase substrate (NUCKS), a protein similar to the HMG (high-mobility group) protein family, is one of the most modified proteins in the mammalian proteome. Although very little is known about the biological roles of NUCKS, emerging clinical evidence suggests that this protein can be a biomarker and therapeutic target in various human ailments, including several types of cancer. An inverse correlation between NUCKS protein levels and body mass index in humans has also been observed. Depletion of NUCKS in mice has been reported to lead to obesity and impaired glucose homoeostasis. Genome-wide genomic and proteomic approaches have revealed that NUCKS is a chromatin regulator that affects transcription. The time is now ripe for further understanding of the role of this novel biomarker of cancer and the metabolic syndrome, and how its sundry modifications can affect its function. Such studies could reveal how NUCKS could be a link between physiological cues and human ailments.
Collapse
|
202
|
Poitou C, Perret C, Mathieu F, Truong V, Blum Y, Durand H, Alili R, Chelghoum N, Pelloux V, Aron-Wisnewsky J, Torcivia A, Bouillot JL, Parks BW, Ninio E, Clément K, Tiret L. Bariatric Surgery Induces Disruption in Inflammatory Signaling Pathways Mediated by Immune Cells in Adipose Tissue: A RNA-Seq Study. PLoS One 2015; 10:e0125718. [PMID: 25938420 PMCID: PMC4418598 DOI: 10.1371/journal.pone.0125718] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/17/2015] [Indexed: 11/30/2022] Open
Abstract
Background Bariatric surgery is associated to improvements in obesity-associated comorbidities thought to be mediated by a decrease of adipose inflammation. However, the molecular mechanisms behind these beneficial effects are poorly understood. Methodology/Principal Findings We analyzed RNA-seq expression profiles in adipose tissue from 22 obese women before and 3 months after surgery. Of 15,972 detected genes, 1214 were differentially expressed after surgery at a 5% false discovery rate. Upregulated genes were mostly involved in the basal cellular machinery. Downregulated genes were enriched in metabolic functions of adipose tissue. At baseline, 26 modules of coexpressed genes were identified. The four most stable modules reflected the innate and adaptive immune responses of adipose tissue. A first module reflecting a non-specific signature of innate immune cells, mainly macrophages, was highly conserved after surgery with the exception of DUSP2 and CD300C. A second module reflected the adaptive immune response elicited by T lymphocytes; after surgery, a disconnection was observed between genes involved in T-cell signaling and mediators of the signal transduction such as CXCR1, CXCR2, GPR97, CCR7 and IL7R. A third module reflected neutrophil-mediated inflammation; after surgery, several genes were dissociated from the module, including S100A8, S100A12, CD300E, VNN2, TUBB1 and FAM65B. We also identified a dense network of 19 genes involved in the interferon-signaling pathway which was strongly preserved after surgery, with the exception of DDX60, an antiviral factor involved in RIG-I-mediated interferon signaling. A similar loss of connection was observed in lean mice compared to their obese counterparts. Conclusions/Significance These results suggest that improvements of the inflammatory state following surgery might be explained by a disruption of immuno-inflammatory cascades involving a few crucial molecules which could serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Christine Poitou
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Nutriomics team, F-75005, Paris, France
| | - Claire Perret
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Genomics and Pathophysiology of Cardiovascular Diseases team, F-75013, Paris, France
| | - François Mathieu
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Genomics and Pathophysiology of Cardiovascular Diseases team, F-75013, Paris, France
| | - Vinh Truong
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Genomics and Pathophysiology of Cardiovascular Diseases team, F-75013, Paris, France
| | - Yuna Blum
- Department of Medicine/Division of Cardiology, University of California Los Angeles, Los Angeles, California 90095, United States of America
| | - Hervé Durand
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Genomics and Pathophysiology of Cardiovascular Diseases team, F-75013, Paris, France
| | - Rohia Alili
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Nutriomics team, F-75005, Paris, France
| | - Nadjim Chelghoum
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Post-Genomic Platform of Pitié-Salpêtrière (P3S), F-75013, Paris, France
| | - Véronique Pelloux
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Nutriomics team, F-75005, Paris, France
| | - Judith Aron-Wisnewsky
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Nutriomics team, F-75005, Paris, France
| | - Adriana Torcivia
- Assistance Publique-Hôpitaux de Paris, Department of Visceral Surgery, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | - Jean-Luc Bouillot
- Assistance Publique-Hôpitaux de Paris, Department of General, Digestive and Metabolic Surgery, Ambroise-Paré Hospital, F- 92100, Boulogne-Billancourt, France
| | - Brian W. Parks
- Department of Medicine/Division of Cardiology, University of California Los Angeles, Los Angeles, California 90095, United States of America
| | - Ewa Ninio
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Genomics and Pathophysiology of Cardiovascular Diseases team, F-75013, Paris, France
| | - Karine Clément
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Nutriomics team, F-75005, Paris, France
| | - Laurence Tiret
- Institute of Cardiometabolism And Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, F-75013, Paris, France
- Sorbonne Universités, University Pierre et Marie Curie (UPMC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Genomics and Pathophysiology of Cardiovascular Diseases team, F-75013, Paris, France
- * E-mail:
| |
Collapse
|
203
|
Sun L, Deng H, He L, Hu X, Huang Q, Xue J, Chen J, Shi X, Xu Y. The relationship between NR2E1 and subclinical inflammation in newly diagnosed type 2 diabetic patients. J Diabetes Complications 2015; 29:589-94. [PMID: 25813674 DOI: 10.1016/j.jdiacomp.2014.12.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/25/2014] [Accepted: 12/26/2014] [Indexed: 02/06/2023]
Abstract
AIMS To evaluate the expression level of NR2E1 (nuclear receptor subfamily 2,group E,member 1) and its correlation with type 2 diabetes (T2DM). METHODS Plasma and peripheral blood mononuclear cells (PBMCs) were collected from 54 T2DM and 88 healthy individuals. The levels of free fatty acids (FFAs), total cholesterol (TC), triglyceride (TG), high density lipoprotein (HDL-c), low density lipoprotein (LDL-c), fasting insulin (FIN), and fasting blood glucose (FBG) were measured. The insulin resistance index was calculated using the homeostasis model assessment (HOMA). NR2E1 in PBMCs were analyzed using real-time RT-PCR and Western blots. Tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) in the plasma were measured by enzyme-linked immunosorbent assay (ELISA). PBMCs isolated from healthy volunteers were treated with glucose or palmitate for 24h, followed by analysis for the expression level of NR2E1.The amount of TNF-α and IL-6 secreted into the supernatant were measured by ELISA. RESULTS FIN, FBG, HOMA and TNF-α, IL-6 were significantly higher in diabetic patients, compared to the control group. Levels of NR2E1 were significantly higher in the PBMCs isolated from the diabetic group, compared to the control group. NR2E1 expression was positively correlated with FBG, FIN, HOMA, FFAs, TNF-α and IL-6. Glucose and palmitate treatment significantly increased NR2E1 gene expression and inflammatory cytokines production in PBMCs in vitro. CONCLUSIONS Increased NR2E1 level may be closely associated with inflammation and disorder of lipid and glucose metabolism in diabetic patients.
Collapse
Affiliation(s)
- Li Sun
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Haohua Deng
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Lanjie He
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Xuemei Hu
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Qi Huang
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Junli Xue
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Jin Chen
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Xiaoli Shi
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China
| | - Yancheng Xu
- Department of Endocrinology Zhongnan Hospital of Wuhan University, 169# Donghu Road, Wuhan, Hubei 430071 China.
| |
Collapse
|
204
|
Pantham P, Aye ILMH, Powell TL. Inflammation in maternal obesity and gestational diabetes mellitus. Placenta 2015; 36:709-15. [PMID: 25972077 DOI: 10.1016/j.placenta.2015.04.006] [Citation(s) in RCA: 405] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/08/2015] [Accepted: 04/13/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND The prevalence of maternal obesity is rising rapidly worldwide and constitutes a major obstetric problem, increasing mortality and morbidity in both mother and offspring. Obese women are predisposed to pregnancy complications such as gestational diabetes mellitus (GDM), and children of obese mothers are more likely to develop cardiovascular and metabolic disease in later life. Maternal obesity and GDM may be associated with a state of chronic, low-grade inflammation termed "metainflammation", as opposed to an acute inflammatory response. This inflammatory environment may be one mechanism by which offspring of obese women are programmed to develop adult disorders. METHODS Herein we review the evidence that maternal obesity and GDM are associated with changes in the maternal, fetal and placental inflammatory profile. RESULTS Maternal inflammation in obesity and GDM may not always be associated with fetal inflammation. CONCLUSION We propose that the placenta 'senses' and adapts to the maternal inflammatory environment, and plays a central role as both a target and producer of inflammatory mediators. In this manner, maternal obesity and GDM may indirectly program the fetus for later disease by influencing placental function.
Collapse
Affiliation(s)
- P Pantham
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - I L M H Aye
- Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - T L Powell
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
205
|
Akıncılar SC, Low KC, Liu CY, Yan TD, Oji A, Ikawa M, Li S, Tergaonkar V. Quantitative assessment of telomerase components in cancer cell lines. FEBS Lett 2015; 589:974-84. [PMID: 25749370 DOI: 10.1016/j.febslet.2015.02.035] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/06/2015] [Accepted: 02/26/2015] [Indexed: 12/22/2022]
Abstract
Besides its canonical function of catalyzing the formation of telomeric repeats, many groups have recently reported non-canonical functions of hTERT in particular, and telomerase in general. Regulating transcription is the central basis of non-canonical functions of telomerase. However, unlike reverse transcriptase activity of telomerase that requires only a few molecules of enzymatically active hTERT, non-canonical functions of hTERT or other telomerase components theoretically require several hundred copies. Here, we provide the first direct quantification of all the telomerase components in human cancer cell lines. We demonstrate that telomerase components do not exist in a 1:1 stoichiometric ratio, and there are several hundred copies of hTERT in cells. This provides the molecular basis of hTERT to function in other signaling cascades, including transcription.
Collapse
Affiliation(s)
- Semih Can Akıncılar
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
| | - Kee Chung Low
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Chia Yi Liu
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Ting Dong Yan
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Asami Oji
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan; Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan; Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Shang Li
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore.
| |
Collapse
|
206
|
The multifaceted role of curcumin in cancer prevention and treatment. Molecules 2015; 20:2728-69. [PMID: 25665066 PMCID: PMC6272781 DOI: 10.3390/molecules20022728] [Citation(s) in RCA: 314] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/30/2015] [Indexed: 02/07/2023] Open
Abstract
Despite significant advances in treatment modalities over the last decade, neither the incidence of the disease nor the mortality due to cancer has altered in the last thirty years. Available anti-cancer drugs exhibit limited efficacy, associated with severe side effects, and are also expensive. Thus identification of pharmacological agents that do not have these disadvantages is required. Curcumin, a polyphenolic compound derived from turmeric (Curcumin longa), is one such agent that has been extensively studied over the last three to four decades for its potential anti-inflammatory and/or anti-cancer effects. Curcumin has been found to suppress initiation, progression, and metastasis of a variety of tumors. These anti-cancer effects are predominantly mediated through its negative regulation of various transcription factors, growth factors, inflammatory cytokines, protein kinases, and other oncogenic molecules. It also abrogates proliferation of cancer cells by arresting them at different phases of the cell cycle and/or by inducing their apoptosis. The current review focuses on the diverse molecular targets modulated by curcumin that contribute to its efficacy against various human cancers.
Collapse
|
207
|
Shin EM, Osato M, Kumar AP, Tergaonkar V. RNA helicase DP103 and TAK1: a new connection in cancer. Mol Cell Oncol 2015; 2:e985911. [PMID: 27308465 PMCID: PMC4905301 DOI: 10.4161/23723556.2014.985911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/06/2014] [Accepted: 11/06/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Eun Myoung Shin
- Cancer Science Institute of Singapore; National University of Singapore; Singapore ; Institute of Molecular and Cellular Biology; ASTAR, Singapore
| | - Motomi Osato
- Cancer Science Institute of Singapore; National University of Singapore; Singapore ; Institute of Bioengineering and Nanotechnology; ASTAR; Singapore; Department of Paediatrics; Yong Loo Lin School of Medicine; National University of Singapore; Singapore; International Research Center for Medical Sciences; Kumamoto University; Kumamoto, Japan
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore; National University of Singapore; Singapore ; Department of Pharmacology; Yong Loo Lin School of Medicine; National University of Singapore; Singapore; National University Cancer Institute; Singapore; School of Biomedical Sciences; Faculty of Health Sciences; Curtin University; Perth, Western Australia, Australia; Department of Biological Sciences; University of North Texas; Denton, TX, USA
| | - Vinay Tergaonkar
- Institute of Molecular and Cellular Biology; ASTAR, Singapore; Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| |
Collapse
|
208
|
Al Hannan F, Culligan KG. Human resistin and the RELM of Inflammation in diabesity. Diabetol Metab Syndr 2015; 7:54. [PMID: 26097512 PMCID: PMC4474570 DOI: 10.1186/s13098-015-0050-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 06/05/2015] [Indexed: 12/11/2022] Open
Abstract
The initial discovery of resistin and resistin-like molecules (RELMs) in rodents suggested a role for these adipocytokines in molecular linkage of obesity, Type 2 Diabetes mellitus and metabolic syndrome. Since then, it became apparent that the story of resistin and RELMs was very much of mice and men. The putative role of this adipokine family evolved from that of a conveyor of insulin resistance in rodents to instigator of inflammatory processes in humans. Structural dissimilarity, variance in distribution profiles and a lack of corroborating evidence for functional similarities separate the biological functions of resistin in humans from that of rodents. Although present in gross visceral fat deposits in humans, resistin is a component of inflammation, being released from infiltrating white blood cells of the sub-clinical chronic low grade inflammatory response accompanying obesity, rather than from the adipocyte itself. This led researchers to further explore the functions of the resistin family of proteins in inflammatory-related conditions such as atherosclerosis, as well as in cancers such as endometrial and gastric cancers. Although elevated levels of resistin have been found in these conditions, whether it is causative or as a result of these conditions still remains to be determined.
Collapse
Affiliation(s)
- Fatima Al Hannan
- />Department of Biomedical Sciences, Royal College of Surgeons in Ireland – Bahrain, Building No. 2441, Road 2835, Busaiteen, Kingdom of Bahrain
| | - Kevin Gerard Culligan
- />Department of Biomedical Sciences, Royal College of Surgeons in Ireland – Bahrain, Building No. 2441, Road 2835, Busaiteen, Kingdom of Bahrain
- />Royal College of Surgeons in Ireland – Bahrain, PO Box 15503, Adliya, Kingdom of Bahrain
| |
Collapse
|
209
|
Bakar MHA, Sarmidi MR, Kai CK, Huri HZ, Yaakob H. Amelioration of mitochondrial dysfunction-induced insulin resistance in differentiated 3T3-L1 adipocytes via inhibition of NF-κB pathways. Int J Mol Sci 2014; 15:22227-57. [PMID: 25474091 PMCID: PMC4284705 DOI: 10.3390/ijms151222227] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 11/14/2014] [Accepted: 11/15/2014] [Indexed: 01/14/2023] Open
Abstract
A growing body of evidence suggests that activation of nuclear factor kappa B (NF-κB) signaling pathways is among the inflammatory mechanism involved in the development of insulin resistance and chronic low-grade inflammation in adipose tissues derived from obese animal and human subjects. Nevertheless, little is known about the roles of NF-κB pathways in regulating mitochondrial function of the adipose tissues. In the present study, we sought to investigate the direct effects of celastrol (potent NF-κB inhibitor) upon mitochondrial dysfunction-induced insulin resistance in 3T3-L1 adipocytes. Celastrol ameliorates mitochondrial dysfunction by altering mitochondrial fusion and fission in adipocytes. The levels of oxidative DNA damage, protein carbonylation and lipid peroxidation were down-regulated. Further, the morphology and quantification of intracellular lipid droplets revealed the decrease of intracellular lipid accumulation with reduced lipolysis. Moreover, massive production of the pro-inflammatory mediators tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were markedly depleted. Insulin-stimulated glucose uptake activity was restored with the enhancement of insulin signaling pathways. This study signified that the treatments modulated towards knockdown of NF-κB transcription factor may counteract these metabolic insults exacerbated in our model of synergy between mitochondrial dysfunction and inflammation. These results demonstrate for the first time that NF-κB inhibition modulates mitochondrial dysfunction induced insulin resistance in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Mohamad Hafizi Abu Bakar
- Department of Bioprocess Engineering, Faculty of Chemical Engineering, University Teknologi Malaysia, Skudai 81310, Malaysia.
| | - Mohamad Roji Sarmidi
- Institute of Bioproduct Development, University Teknologi Malaysia, Skudai 81310, Malaysia.
| | - Cheng Kian Kai
- Department of Bioprocess Engineering, Faculty of Chemical Engineering, University Teknologi Malaysia, Skudai 81310, Malaysia.
| | - Hasniza Zaman Huri
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Harisun Yaakob
- Innovation Centre in Agritechnology for Advanced Bioprocessing (ICA), University Teknologi Malaysia, Skudai 81310, Malaysia.
| |
Collapse
|
210
|
Gremese E, Tolusso B, Gigante MR, Ferraccioli G. Obesity as a risk and severity factor in rheumatic diseases (autoimmune chronic inflammatory diseases). Front Immunol 2014; 5:576. [PMID: 25426122 PMCID: PMC4227519 DOI: 10.3389/fimmu.2014.00576] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/27/2014] [Indexed: 12/31/2022] Open
Abstract
The growing body of evidence recognizing the adipose tissue (AT) as an active endocrine organ secreting bioactive mediators involved in metabolic and inflammatory disorders, together with the global epidemic of overweight and obesity, rise obesity as a hot topic of current research. The chronic state of low-grade inflammation present in the obese condition and the multiple pleiotropic effects of adipokines on the immune system has been implicated in the pathogenesis of several inflammatory conditions including rheumatic autoimmune and inflammatory diseases. We will discuss the main relevant evidences on the role of the AT on immune and inflammatory networks and the more recent evidences regarding the effects of obesity on the incidence and outcomes of the major autoimmune chronic inflammatory diseases.
Collapse
Affiliation(s)
- Elisa Gremese
- Division of Rheumatology, Institute of Rheumatology and Affine Sciences, Catholic University of the Sacred Heart , Rome , Italy
| | - Barbara Tolusso
- Division of Rheumatology, Institute of Rheumatology and Affine Sciences, Catholic University of the Sacred Heart , Rome , Italy
| | - Maria Rita Gigante
- Division of Rheumatology, Institute of Rheumatology and Affine Sciences, Catholic University of the Sacred Heart , Rome , Italy
| | - Gianfranco Ferraccioli
- Division of Rheumatology, Institute of Rheumatology and Affine Sciences, Catholic University of the Sacred Heart , Rome , Italy
| |
Collapse
|
211
|
Theaflavin-3, 3′-digallate, a black tea polyphenol, attenuates adipocyte-activated inflammatory response of macrophage associated with the switch of M1/M2-like phenotype. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.09.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
212
|
Santosa S, Swain J, Tchkonia T, Kirkland JL, Jensen MD. Inflammatory characteristics of adipose tissue collected by surgical excision vs needle aspiration. Int J Obes (Lond) 2014; 39:874-6. [PMID: 25319743 PMCID: PMC4400182 DOI: 10.1038/ijo.2014.185] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/03/2014] [Accepted: 09/24/2014] [Indexed: 11/10/2022]
Abstract
Subcutaneous adipose tissue can be obtained for research during an elective, clinically indicated operation by standard surgical excision approaches and by needle aspiration in pure research settings. Whether measurements of inflammatory markers and cells made in tissue collected these two different ways are comparable is debatable. We sought to determine whether these two techniques yield systematically different results for measurements of inflammation, cellular senescence, and adipose tissue composition. Twelve subjects undergoing surgery participated. At the time of surgery abdominal subcutaneous adipose tissue from adjacent sites was removed by excision and needle aspiration. Stromovascular cell composition (flow cytometry), the number of senescent cells (senescence-associated-β-galactosidase staining), and IL-6, IL-1, TNF-α, MCP1 mRNA (RT-PCR) were measured in each sample. We found no statistically significant differences between the two sample collection approaches for any of the parameters measured. We conclude that these two methods of obtaining adipose tissue do not systematically differ in the results of cytokine mRNA content, cellular senescence, or stromovascular cell composition.
Collapse
Affiliation(s)
- S Santosa
- 1] Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA [2] Department of Exercise Science, Concordia University, Montreal, Québec, Canada
| | - J Swain
- 1] Department of Surgery, Mayo Clinic, Rochester, MN, USA [2] Scottsdale Healthcare Bariatric Center, Scottsdale, AZ, USA
| | - T Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - J L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - M D Jensen
- Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
213
|
Rose S, Stansky E, Dagur PK, Samsel L, Weiner E, Jahanshad A, Doveikis J, Naik HB, Playford MP, McCoy JP, Mehta NN. Characterization of immune cells in psoriatic adipose tissue. J Transl Med 2014; 12:258. [PMID: 25224267 PMCID: PMC4197293 DOI: 10.1186/s12967-014-0258-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 09/09/2014] [Indexed: 12/21/2022] Open
Abstract
Background Adipose tissue normally contains immune cells that regulate adipocyte function and contribute to metabolic disorders including obesity and diabetes mellitus. Psoriasis is associated with increased risk for metabolic disease, which may in part be due to adipose dysfunction, which has not been investigated in psoriasis. There is currently no standardized method for immunophenotyping human adipose tissue. In prior studies, characteristic phenotypic markers of immune cell populations identified in animal models or in other human tissues have been applied in a similar manner to human adipose tissue. Rarely have these populations been verified with confirmatory methodologies or functional studies. Thus, we performed a comprehensive phenotypic and functional analysis of immune cell populations in psoriatic adipose tissue. Methods Conventional and imaging flow cytometry were used to define immune cell populations in biopsy specimens of psoriatic adipose tissue (n = 30) including T cells, B cells, NK cells, NKT cells, neutrophils, and macrophages. Relationships between adipose immune cell types and body mass index were determined using Spearman regression analysis, and multivariate linear regression analysis was performed to adjust for cardiometabolic disease risk factors. Results These analyses revealed a wide range of cell surface receptors on adipose tissue macrophages, which may serve a dual purpose in immunity and metabolism. Further, both CD16+CD56Lo and CD16-CD56Hi NK cells were found to correlate inversely with body mass index. The relationship between the predominant CD16+CD56Lo NK cell population and body mass index persisted after adjusting for age, sex, diabetes, and tobacco use. Conclusions Together, these studies enhance our understanding of adipose immune cell phenotype and function, and demonstrate that examination of adipose tissue may provide greater insight into cardiometabolic pathophysiology in psoriasis. Electronic supplementary material The online version of this article (doi:10.1186/s12967-014-0258-2) contains supplementary material, which is available to authorized users.
Collapse
|
214
|
Abstract
Adipose tissue (AT) lies at the crossroad of nutrition, metabolism, and immunity; AT inflammation was proposed as a central mechanism connecting obesity with its metabolic and vascular complications. Resident immune cells constitute the second largest AT cellular component after adipocytes and as such play important roles in the maintenance of AT homeostasis. Obesity-induced changes in their number and activity result in the activation of local and later systemic inflammatory response, marking the transition from simple adiposity to diseases such as type 2 diabetes mellitus, arterial hypertension, and ischemic heart disease. This review has focused on the various subsets of immune cells in AT and their role in the development of AT inflammation and obesity-induced insulin resistance.
Collapse
Affiliation(s)
- Milos Mraz
- Third Department of Medicine - Department of Endocrinology and MetabolismGeneral University Hospital, First Faculty of Medicine of Charles University in Prague, U nemocnice 1, 128 00 Prague 2, Czech Republic
| | - Martin Haluzik
- Third Department of Medicine - Department of Endocrinology and MetabolismGeneral University Hospital, First Faculty of Medicine of Charles University in Prague, U nemocnice 1, 128 00 Prague 2, Czech Republic
| |
Collapse
|
215
|
Shin EM, Hay HS, Lee MH, Goh JN, Tan TZ, Sen YP, Lim SW, Yousef EM, Ong HT, Thike AA, Kong X, Wu Z, Mendoz E, Sun W, Salto-Tellez M, Lim CT, Lobie PE, Lim YP, Yap CT, Zeng Q, Sethi G, Lee MB, Tan P, Goh BC, Miller LD, Thiery JP, Zhu T, Gaboury L, Tan PH, Hui KM, Yip GWC, Miyamoto S, Kumar AP, Tergaonkar V. DEAD-box helicase DP103 defines metastatic potential of human breast cancers. J Clin Invest 2014; 124:3807-24. [PMID: 25083991 DOI: 10.1172/jci73451] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 05/23/2014] [Indexed: 12/12/2022] Open
Abstract
Despite advancement in breast cancer treatment, 30% of patients with early breast cancers experience relapse with distant metastasis. It is a challenge to identify patients at risk for relapse; therefore, the identification of markers and therapeutic targets for metastatic breast cancers is imperative. Here, we identified DP103 as a biomarker and metastasis-driving oncogene in human breast cancers and determined that DP103 elevates matrix metallopeptidase 9 (MMP9) levels, which are associated with metastasis and invasion through activation of NF-κB. In turn, NF-κB signaling positively activated DP103 expression. Furthermore, DP103 enhanced TGF-β-activated kinase-1 (TAK1) phosphorylation of NF-κB-activating IκB kinase 2 (IKK2), leading to increased NF-κB activity. Reduction of DP103 expression in invasive breast cancer cells reduced phosphorylation of IKK2, abrogated NF-κB-mediated MMP9 expression, and impeded metastasis in a murine xenograft model. In breast cancer patient tissues, elevated levels of DP103 correlated with enhanced MMP9, reduced overall survival, and reduced survival after relapse. Together, these data indicate that a positive DP103/NF-κB feedback loop promotes constitutive NF-κB activation in invasive breast cancers and activation of this pathway is linked to cancer progression and the acquisition of chemotherapy resistance. Furthermore, our results suggest that DP103 has potential as a therapeutic target for breast cancer treatment.
Collapse
|
216
|
Rho protein GTPases and their interactions with NFκB: crossroads of inflammation and matrix biology. Biosci Rep 2014; 34:BSR20140021. [PMID: 24877606 PMCID: PMC4069681 DOI: 10.1042/bsr20140021] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The RhoGTPases, with RhoA, Cdc42 and Rac being major members, are a group of key ubiquitous proteins present in all eukaryotic organisms that subserve such important functions as cell migration, adhesion and differentiation. The NFκB (nuclear factor κB) is a family of constitutive and inducible transcription factors that through their diverse target genes, play a major role in processes such as cytokine expression, stress regulation, cell division and transformation. Research over the past decade has uncovered new molecular links between the RhoGTPases and the NFκB pathway, with the RhoGTPases playing a positive or negative regulatory role on NFκB activation depending on the context. The RhoA–NFκB interaction has been shown to be important in cytokine-activated NFκB processes, such as those induced by TNFα (tumour necrosis factor α). On the other hand, Rac is important for activating the NFκB response downstream of integrin activation, such as after phagocytosis. Specific residues of Rac1 are important for triggering NFκB activation, and mutations do obliterate this response. Other upstream triggers of the RhoGTPase–NFκB interactions include the suppressive p120 catenin, with implications for skin inflammation. The networks described here are not only important areas for further research, but are also significant for discovery of targets for translational medicine.
Collapse
|
217
|
Rakhshandehroo M, Gijzel SMW, Siersbæk R, Broekema MF, de Haar C, Schipper HS, Boes M, Mandrup S, Kalkhoven E. CD1d-mediated presentation of endogenous lipid antigens by adipocytes requires microsomal triglyceride transfer protein. J Biol Chem 2014; 289:22128-39. [PMID: 24966328 DOI: 10.1074/jbc.m114.551242] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Obesity-induced adipose tissue (AT) dysfunction results in a chronic low-grade inflammation that predisposes to the development of insulin resistance and type 2 diabetes. During the development of obesity, the AT-resident immune cell profile alters to create a pro-inflammatory state. Very recently, CD1d-restricted invariant (i) natural killer T (NKT) cells, a unique subset of lymphocytes that are reactive to so called lipid antigens, were implicated in AT homeostasis. Interestingly, recent data also suggest that human and mouse adipocytes can present such lipid antigens to iNKT cells in a CD1d-dependent fashion, but little is known about the lipid antigen presentation machinery in adipocytes. Here we show that CD1d, as well as the lipid antigen loading machinery genes pro-saposin (Psap), Niemann Pick type C2 (Npc2), α-galactosidase (Gla), are up-regulated in early adipogenesis, and are transcriptionally controlled by CCAAT/enhancer-binding protein (C/EBP)-β and -δ. Moreover, adipocyte-induced Th1 and Th2 cytokine release by iNKT cells also occurred in the absence of exogenous ligands, suggesting the display of endogenous lipid antigen-D1d complexes by 3T3-L1 adipocytes. Furthermore, we identified microsomal triglyceride transfer protein, which we show is also under the transcriptional regulation of C/EBPβ and -δ, as a novel player in the presentation of endogenous lipid antigens by adipocytes. Overall, our findings indicate that adipocytes can function as non-professional lipid antigen presenting cells, which may present an important aspect of adipocyte-immune cell communication in the regulation of whole body energy metabolism and immune homeostasis.
Collapse
Affiliation(s)
| | - Sanne M W Gijzel
- From the Molecular Cancer Research, Center for Molecular Medicine and
| | - Rasmus Siersbæk
- the Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | | | - Colin de Haar
- the Department of Pediatric Immunology, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands and
| | - Henk S Schipper
- From the Molecular Cancer Research, Center for Molecular Medicine and the Department of Pediatric Immunology, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands and
| | - Marianne Boes
- the Department of Pediatric Immunology, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands and
| | - Susanne Mandrup
- the Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Eric Kalkhoven
- From the Molecular Cancer Research, Center for Molecular Medicine and
| |
Collapse
|
218
|
Qiu B, Shi X, Wong ET, Lim J, Bezzi M, Low D, Zhou Q, Akıncılar SC, Lakshmanan M, Swa HLF, Tham JML, Gunaratne J, Cheng KKY, Hong W, Lam KSL, Ikawa M, Guccione E, Xu A, Han W, Tergaonkar V. NUCKS is a positive transcriptional regulator of insulin signaling. Cell Rep 2014; 7:1876-86. [PMID: 24931609 DOI: 10.1016/j.celrep.2014.05.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 03/17/2014] [Accepted: 05/14/2014] [Indexed: 12/24/2022] Open
Abstract
Although much is known about the molecular players in insulin signaling, there is scant information about transcriptional regulation of its key components. We now find that NUCKS is a transcriptional regulator of the insulin signaling components, including the insulin receptor (IR). Knockdown of NUCKS leads to impaired insulin signaling in endocrine cells. NUCKS knockout mice exhibit decreased insulin signaling and increased body weight/fat mass along with impaired glucose tolerance and reduced insulin sensitivity, all of which are further exacerbated by a high-fat diet (HFD). Genome-wide ChIP-seq identifies metabolism and insulin signaling as NUCKS targets. Importantly, NUCKS is downregulated in individuals with a high body mass index and in HFD-fed mice, and conversely, its levels increase upon starvation. Altogether, NUCKS is a physiological regulator of energy homeostasis and glucose metabolism that works by regulating chromatin accessibility and RNA polymerase II recruitment to the promoters of IR and other insulin pathway modulators.
Collapse
Affiliation(s)
- Beiying Qiu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Xiaohe Shi
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138667, Singapore
| | - Ee Tsin Wong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Joy Lim
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138667, Singapore
| | - Marco Bezzi
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Diana Low
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Qiling Zhou
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Semih Can Akıncılar
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Manikandan Lakshmanan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Hannah L F Swa
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Jill Mae Lan Tham
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Jayantha Gunaratne
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Kenneth K Y Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, Hong Kong, China
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Karen S L Lam
- State Key Laboratory of Pharmaceutical Biotechnology, Hong Kong, China
| | | | - Ernesto Guccione
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Weiping Han
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138667, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| | - Vinay Tergaonkar
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138673, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
219
|
Zhou X, Yuan F, Ji WJ, Guo ZZ, Zhang L, Lu RY, Liu X, Liu HM, Zhang WC, Jiang TM, Zhang Z, Li YM. High-salt intake induced visceral adipose tissue hypoxia and its association with circulating monocyte subsets in humans. Obesity (Silver Spring) 2014; 22:1470-6. [PMID: 24493236 DOI: 10.1002/oby.20716] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 01/29/2014] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To investigate the feasibility of blood oxygen level dependent magnetic resonance imaging (BOLD-MRI) in evaluating human visceral adipose tissue (AT) oxygenation induced by salt loading/depletion and its association with changes in circulating monocyte subsets. METHODS A dietary intervention study was performed in 23 healthy volunteers beginning with a 3-day usual diet followed by a 7-day high-salt diet (≥15 g NaCl/day) and a 7-day low-salt diet (≤5 g NaCl/day). BOLD-MRI was used to evaluate oxygenation in perirenal AT. RESULTS Salt loading led to a consistent AT hypoxia (increase in the R2* signal, 25.2 ± 0.90 s(-1) vs. baseline 21.5 ± 0.71 s(-1) , P < 0.001) and suppression of circulating renin-angiotensin-aldosterone system (RAAS), as well as an expansion of the CD14++CD16+ monocytes and monocyte pro-inflammatory activation. In salt depletion phase, the hypoxic state of AT and the expanded CD14++CD16+ monocyte pool were regressed to baseline levels, accompanied by a rebound activation of RAAS. Moreover, AT oxygenation level was positively correlated with the CD14++CD16+ monocytes (r = 0.419, P < 0.001). CONCLUSIONS This work provides proof-of-principle evidence supporting the feasibility of BOLD-MRI in monitoring visceral AT oxygenation in humans induced by dietary salt loading/depletion. In addition, the CD14++CD16+ monocytes may participate in the pathogenesis of high-salt intake induced AT hypoxia.
Collapse
Affiliation(s)
- Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury Institute of Cardiovascular Disease and Heart Center Pingjin Hospital, Logistics University of the Chinese People's Armed Police Forces, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Hahn WS, Kuzmicic J, Burrill JS, Donoghue MA, Foncea R, Jensen MD, Lavandero S, Arriaga EA, Bernlohr DA. Proinflammatory cytokines differentially regulate adipocyte mitochondrial metabolism, oxidative stress, and dynamics. Am J Physiol Endocrinol Metab 2014; 306:E1033-45. [PMID: 24595304 PMCID: PMC4010657 DOI: 10.1152/ajpendo.00422.2013] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Proinflammatory cytokines differentially regulate adipocyte mitochondrial metabolism, oxidative stress, and dynamics. Macrophage infiltration of adipose tissue and the chronic low-grade production of inflammatory cytokines have been mechanistically linked to the development of insulin resistance, the forerunner of type 2 diabetes mellitus. In this study, we evaluated the chronic effects of TNFα, IL-6, and IL-1β on adipocyte mitochondrial metabolism and morphology using the 3T3-L1 model cell system. TNFα treatment of cultured adipocytes led to significant changes in mitochondrial bioenergetics, including increased proton leak, decreased ΔΨm, increased basal respiration, and decreased ATP turnover. In contrast, although IL-6 and IL-1β decreased maximal respiratory capacity, they had no effect on ΔΨm and varied effects on ATP turnover, proton leak, or basal respiration. Only TNFα treatment of 3T3-L1 cells led to an increase in oxidative stress (as measured by superoxide anion production and protein carbonylation) and C16 ceramide synthesis. Treatment of 3T3-L1 adipocytes with cytokines led to decreased mRNA expression of key transcription factors and control proteins implicated in mitochondrial biogenesis, including PGC-1α and eNOS as well as deceased expression of COX IV and Cyt C. Whereas each cytokine led to effects on expression of mitochondrial markers, TNFα exclusively led to mitochondrial fragmentation and decreased the total level of OPA1 while increasing OPA1 cleavage, without expression of levels of mitofusin 2, DRP-1, or mitofilin being affected. In summary, these results indicate that inflammatory cytokines have unique and specialized effects on adipocyte metabolism, but each leads to decreased mitochondrial function and a reprogramming of fat cell biology.
Collapse
Affiliation(s)
- Wendy S Hahn
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Pecht T, Gutman-Tirosh A, Bashan N, Rudich A. Peripheral blood leucocyte subclasses as potential biomarkers of adipose tissue inflammation and obesity subphenotypes in humans. Obes Rev 2014; 15:322-37. [PMID: 24251825 DOI: 10.1111/obr.12133] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 10/01/2013] [Accepted: 10/18/2013] [Indexed: 12/14/2022]
Abstract
While obesity is clearly accepted as a major risk factor for cardio-metabolic morbidity, it is also apparent that some obese patients largely escape this association, forming a unique obese subphenotype(s). Current approaches to define such subphenotypes include clinical biomarkers that largely reflect already manifested comorbidities, such as markers of dyslipidaemia, hyperglycaemia and impaired regulation of vascular tone, and anthropometric or imaging-based assessment of adipose tissue distribution. Low-grade inflammation, evident both systemically and within adipose tissue (particularly intra-abdominal fat depots), seems to characterize the more cardio-metabolically morbid forms of obesity. Indeed, several systemic inflammatory markers (C-reactive protein), adipokines (retinol-binding protein 4, adiponectin) and cytokines have been shown to correlate in humans with adipose tissue inflammation and with obesity-associated health risks. Circulating leucocytes constitute a diverse group of cells that form a major arm of the immune system. They are both major sources of cytokines and likely also of infiltrating adipose tissue immune cells in obesity. In the present review, we summarize currently available literature on 'classical' blood white cell classes and on more specific leucocyte subclasses present in the circulation in human obesity. We critically raise the possibility that leucocytes may constitute clinically available markers for the more morbidity-associated obesity subphenotype(s), and when available, for intra-abdominal adipose tissue inflammation.
Collapse
Affiliation(s)
- T Pecht
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; The National Institute of Biotechnology (NIBN) in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | |
Collapse
|
222
|
Activation and regulation of the pattern recognition receptors in obesity-induced adipose tissue inflammation and insulin resistance. Nutrients 2013; 5:3757-78. [PMID: 24064574 PMCID: PMC3798933 DOI: 10.3390/nu5093757] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 08/14/2013] [Accepted: 09/11/2013] [Indexed: 12/19/2022] Open
Abstract
Obesity-associated chronic tissue inflammation is a key contributing factor to type 2 diabetes mellitus, and a number of studies have clearly demonstrated that the immune system and metabolism are highly integrated. Recent advances in deciphering the various immune cells and signaling networks that link the immune and metabolic systems have contributed to our understanding of the pathogenesis of obesity-associated inflammation. Other recent studies have suggested that pattern recognition receptors in the innate immune system recognize various kinds of endogenous and exogenous ligands, and have a crucial role in initiating or promoting obesity-associated chronic inflammation. Importantly, these mediators act on insulin target cells or on insulin-producing cells impairing insulin sensitivity and its secretion. Here, we discuss how various pattern recognition receptors in the immune system underlie the etiology of obesity-associated inflammation and insulin resistance, with a particular focus on the TLR (Toll-like receptor) family protein Radioprotective 105 (RP105)/myeloid differentiation protein-1 (MD-1).
Collapse
|