701
|
Izaguirre M, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Becerril S, Valentí V, Moncada R, Unamuno X, Silva C, de la Higuera M, Salvador J, Monreal I, Frühbeck G, Catalán V. GLP-1 Limits Adipocyte Inflammation and Its Low Circulating Pre-Operative Concentrations Predict Worse Type 2 Diabetes Remission after Bariatric Surgery in Obese Patients. J Clin Med 2019; 8:E479. [PMID: 30970605 PMCID: PMC6518381 DOI: 10.3390/jcm8040479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/01/2019] [Accepted: 04/08/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Glucagon-like peptide (GLP)-1 has been proposed as a key candidate in glucose improvements after bariatric surgery. Our aim was to explore the role of GLP-1 in surgically-induced type 2 diabetes (T2D) improvement and its capacity to regulate human adipocyte inflammation. METHODS Basal circulating concentrations of GLP-1 as well as during an oral glucose tolerance test (OGTT) were measured in lean and obese volunteers with and without T2D (n = 93). In addition, GLP-1 levels were determined before and after weight loss achieved by Roux-en-Y gastric bypass (RYGB) (n = 77). The impact of GLP-1 on inflammation signalling pathways was also evaluated. RESULTS We show that the reduced (p < 0.05) circulating levels of GLP-1 in obese T2D patients increased (p < 0.05) after RYGB. The area under the curve was significantly lower in obese patients with (p < 0.01) and without (p < 0.05) T2D compared to lean volunteers while obese patients with T2D exhibited decreased GLP-1 levels at baseline (p < 0.05) and 120 min (p < 0.01) after the OGTT. Importantly, higher (p < 0.05) pre-operative GLP-1 concentrations were found in patients with T2D remission after RYGB. We also revealed that exendin-4, a GLP-1 agonist, downregulated the expression of inflammation-related genes (IL1B, IL6, IL8, TNF) and, conversely, upregulated the mRNA levels of ADIPOQ in human visceral adipocytes. Furthermore, exendin-4 blocked (p < 0.05) LPS-induced inflammation in human adipocytes via downregulating the expression and secretion of key inflammatory markers. CONCLUSIONS Our data indicate that GLP-1 may contribute to glycemic control and exert a role in T2D remission after RYGB. GLP-1 is also involved in limiting inflammation in human visceral adipocytes.
Collapse
Affiliation(s)
- Maitane Izaguirre
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
- Department of Surgery, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
- Department of Anesthesia, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
| | - Camilo Silva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona/Madrid, Spain.
| | - Magdalena de la Higuera
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona/Madrid, Spain.
| | - Javier Salvador
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona/Madrid, Spain.
| | - Ignacio Monreal
- Department of Biochemistry, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona/Madrid, Spain.
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| |
Collapse
|
702
|
Kwon Y, Park S. Microvascular Outcomes in Patients With Diabetes After Bariatric Surgery. Ann Intern Med 2019; 170:507. [PMID: 30934069 DOI: 10.7326/l19-0014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Yeongkeun Kwon
- Korea University College of Medicine, Seoul, Korea (Y.K., S.P.)
| | - Sungsoo Park
- Korea University College of Medicine, Seoul, Korea (Y.K., S.P.)
| |
Collapse
|
703
|
Guilherme A, Henriques F, Bedard AH, Czech MP. Molecular pathways linking adipose innervation to insulin action in obesity and diabetes mellitus. Nat Rev Endocrinol 2019; 15:207-225. [PMID: 30733616 PMCID: PMC7073451 DOI: 10.1038/s41574-019-0165-y] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adipose tissue comprises adipocytes and many other cell types that engage in dynamic crosstalk in a highly innervated and vascularized tissue matrix. Although adipose tissue has been studied for decades, it has been appreciated only in the past 5 years that extensive arborization of nerve fibres has a dominant role in regulating the function of adipose tissue. This Review summarizes the latest literature, which suggests that adipocytes signal to local sensory nerve fibres in response to perturbations in lipolysis and lipogenesis. Such adipocyte signalling to the central nervous system causes sympathetic output to distant adipose depots and potentially other metabolic tissues to regulate systemic glucose homeostasis. Paracrine factors identified in the past few years that mediate such adipocyte-neuron crosstalk are also reviewed. Similarly, immune cells and endothelial cells within adipose tissue communicate with local nerve fibres to modulate neurotransmitter tone, blood flow, adipocyte differentiation and energy expenditure, including adipose browning to produce heat. This understudied field of neurometabolism related to adipose tissue biology has great potential to reveal new mechanistic insights and potential therapeutic strategies for obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Adilson Guilherme
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Felipe Henriques
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Alexander H Bedard
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
704
|
Association between Irisin, hs-CRP, and Metabolic Status in Children and Adolescents with Type 2 Diabetes Mellitus. Mediators Inflamm 2019; 2019:6737318. [PMID: 31015797 PMCID: PMC6446111 DOI: 10.1155/2019/6737318] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/28/2019] [Accepted: 02/07/2019] [Indexed: 02/07/2023] Open
Abstract
Proinflammatory cytokines and the novel myokine irisin, a cleavage product of FNDC5, have been found to play a role in obesity and type 2 diabetes mellitus (T2DM). Irisin has been shown to increase browning of adipose tissue, thermogenesis, energy expenditure, and insulin sensitivity, yet its association with inflammatory markers is still limited. Circulating irisin has been found to be increased in obesity, while in adult subjects with T2DM decreased levels have been found. However, data establishing the association of circulating irisin in children and adolescents with T2DM has not been described in the literature. The objective of this study was to determine irisin plasma concentration and its association with metabolic and adiposity markers and with hs-CRP, a surrogate marker of inflammation used in clinical practice, in a pediatric population with T2DM. A cross-sample of 40 Mexican children and adolescents aged 7-17 were recruited, 20 diagnosed with T2DM and 20 healthy controls. Plasma irisin levels were found to be lower in the T2DM group compared with controls, which could be attributed to a reduced PGC-1α activity in muscle tissue with a consequent decrease in FNDC5 and irisin expression. Irisin concentration was found to be positively correlated with HDL-c, LDL-c, and total cholesterol, while negatively correlated with BMI, waist circumference, and triglycerides. However, after multiple regression analysis, only HDL-c correlation remained significant. hs-CRP was higher in the T2DM group and positively associated with adiposity markers, unfavorable lipid profile, insulin levels, and HOMA-IR, but no association with irisin was found. Given the favorable metabolic effects attributed to irisin, the low plasma levels found in children and adolescents with T2DM could exacerbate the inflammatory and metabolic imbalances and the intrinsic cardiovascular risk of this disease. We propose an "irisin-proinflammatory/anti-inflammatory axis" to explain the role of irisin as a metabolic regulator in obesity and T2DM.
Collapse
|
705
|
Henry NL, Unger JM, Till C, Schott AF, Crew KD, Lew DL, Fisch MJ, Moinpour CM, Wade JL, Hershman DL. Association between body mass index and response to duloxetine for aromatase inhibitor-associated musculoskeletal symptoms in SWOG S1202. Cancer 2019; 125:2123-2129. [PMID: 30861098 DOI: 10.1002/cncr.32024] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/18/2019] [Accepted: 01/25/2019] [Indexed: 11/06/2022]
Abstract
BACKGROUND Aromatase inhibitor (AI)-associated musculoskeletal symptoms (AIMSS) negatively impact adherence to and persistence with therapy. In SWOG S1202, patients with AIMSS who were treated with duloxetine, a serotonin norepinephrine reuptake inhibitor, reported improvement in pain by 12 weeks compared with placebo. Based on the authors' prior observation that responses to pain interventions differ between obese and nonobese patients, the current study examined whether response to duloxetine therapy differed by obesity status. METHODS In SWOG S1202, a total of 299 AI-treated postmenopausal women with stage I to III (AJCC 7th Edition) breast cancer who developed new or worsening average pain were enrolled, randomized to duloxetine or placebo, and treated for 12 weeks. Patient-reported outcomes were obtained at baseline and through 12 weeks. Patients were categorized into nonobese (body mass index [BMI] <30 kg/m2 ) or obese (BMI ≥30 kg/m2 ). The authors tested the interaction between intervention and obesity with respect to average pain at 12 weeks in the 289 eligible patients, using a P value of .05 to indicate statistical significance. RESULTS In approximately 54% of evaluable patients with a BMI ≥30 kg/m2 , the reduction in the mean average pain score between baseline and 12 weeks was statistically significantly greater for patients treated with duloxetine compared with those receiving placebo (-2.73 vs -1.64 points; P = .003). Conversely, in the nonobese patients, the reduction in the mean average pain score was similar in the 2 cohorts (-2.46 vs -2.34 points; P = .75). The P value for interaction was .02, thereby meeting the threshold criteria of the current study. Similar findings were evident for other pain-related patient-reported outcomes. CONCLUSIONS In this trial, obese patients with AIMSS obtained more analgesic benefit from duloxetine compared with nonobese patients. Additional studies are warranted to determine the biologic basis for these findings.
Collapse
Affiliation(s)
- N Lynn Henry
- Department of Internal Medicine, University of Utah Huntsman Cancer Institute, Salt Lake City, Utah
| | - Joseph M Unger
- SWOG Statistics and Data Management Center, Seattle, Washington.,Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Cathee Till
- SWOG Statistics and Data Management Center, Seattle, Washington.,Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Anne F Schott
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | - Danika L Lew
- SWOG Statistics and Data Management Center, Seattle, Washington.,Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Carol M Moinpour
- Cancer Prevention Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - James L Wade
- Heartland National Cancer Institute Community Oncology Research Program, Decatur, Illinois
| | - Dawn L Hershman
- Department of Medicine, Columbia University, New York, New York
| |
Collapse
|
706
|
Wang N, Zhao TT, Li SM, Sun X, Li ZC, Li YH, Li DS, Wang WF. Fibroblast Growth Factor 21 Exerts its Anti-inflammatory Effects on Multiple Cell Types of Adipose Tissue in Obesity. Obesity (Silver Spring) 2019; 27:399-408. [PMID: 30703283 DOI: 10.1002/oby.22376] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/21/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Obesity-related, chronic, low-grade inflammation has been identified as a key factor in the development of many metabolic diseases, such as type 2 diabetes and cardiovascular diseases. Adipocytes, preadipocytes, and macrophages have been implicated in initiating inflammation in adipose tissue. This study aims to investigate the effects of fibroblast growth factor-21 (FGF-21) on obesity-related inflammation and its mechanisms in vivo and in vitro. METHODS Monosodium glutamate (MSG) was used to induce obesity in mice and subsequently treated the mice with or without FGF-21. Primary adipocytes and stromal vascular fraction cells were isolated from MSG-obesity mice for additional experiments. RESULTS Results obtained by ELISA and real-time polymerase chain reaction showed that FGF-21 efficiently ameliorated obesity-related inflammation in MSG-obesity mice. This study demonstrated that preadipocytes and adipocytes responded to anti-inflammatory effects of FGF-21. In vitro, 3 T3-L1 preadipocytes lacking β-klotho did not respond to FGF-21 under glucose uptake. Interestingly, the treatment of 3 T3-L1 preadipocytes with FGF-21 significantly attenuated lipopolysaccharide-induced inflammatory response. CONCLUSIONS Our study showed that FGF-21-induced glucose uptake and FGF-21-related anti-inflammatory effects are mediated by different signaling pathways. Moreover, FGF-21 showed anti-inflammatory effects on preadipocytes; these effects are mediated by the fibroblast growth factor receptor substrate 2/ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Nan Wang
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Ting-Ting Zhao
- Aier School of Ophthalmology, Central South University, Changsha, China
- Aier Eye Hospital Group, Harbin, China
| | - Si-Ming Li
- College of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Xu Sun
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Zi-Cheng Li
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Yan-Hua Li
- School of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - De-Shan Li
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Wen-Fei Wang
- School of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
707
|
Atawia RT, Bunch KL, Toque HA, Caldwell RB, Caldwell RW. Mechanisms of obesity-induced metabolic and vascular dysfunctions. FRONT BIOSCI-LANDMRK 2019; 24:890-934. [PMID: 30844720 PMCID: PMC6689231 DOI: 10.2741/4758] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity has reached epidemic proportions and its prevalence is climbing. Obesity is characterized by hypertrophied adipocytes with a dysregulated adipokine secretion profile, increased recruitment of inflammatory cells, and impaired metabolic homeostasis that eventually results in the development of systemic insulin resistance, a phenotype of type 2 diabetes. Nitric oxide synthase (NOS) is an enzyme that converts L-arginine to nitric oxide (NO), which functions to maintain vascular and adipocyte homeostasis. Arginase is a ureohydrolase enzyme that competes with NOS for L-arginine. Arginase activity/expression is upregulated in obesity, which results in diminished bioavailability of NO, impairing both adipocyte and vascular endothelial cell function. Given the emerging role of NO in the regulation of adipocyte physiology and metabolic capacity, this review explores the interplay between arginase and NO, and their effect on the development of metabolic disorders, cardiovascular diseases, and mitochondrial dysfunction in obesity. A comprehensive understanding of the mechanisms involved in the development of obesity-induced metabolic and vascular dysfunction is necessary for the identification of more effective and tailored therapeutic avenues for their prevention and treatment.
Collapse
Affiliation(s)
- Reem T Atawia
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Katharine L Bunch
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology,and Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Robert W Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904,USA,
| |
Collapse
|
708
|
Abstract
Obesity is associated with both increased cancer incidence and progression in multiple tumour types, and is estimated to contribute to up to 20% of cancer-related deaths. These associations are driven, in part, by metabolic and inflammatory changes in adipose tissue that disrupt physiological homeostasis both within local tissues and systemically. However, the mechanisms underlying the obesity-cancer relationship are poorly understood. In this Review, we describe how the adipose tissue microenvironment (ATME) evolves during body-weight gain, and how these changes might influence tumour initiation and progression. We focus on multiple facets of ATME physiology, including inflammation, vascularity and fibrosis, and discuss therapeutic interventions that have the potential to normalize the ATME, which might be translationally relevant for cancer prevention and therapy. Given that the prevalence of obesity is increasing on an international scale, translational research initiatives are urgently needed to provide mechanistic explanations for the obesity-cancer relationship, and how to best identify high-risk individuals without relying on crude measures, such as BMI.
Collapse
Affiliation(s)
- Daniela F Quail
- Goodman Cancer Research Centre, Department of Physiology, McGill University, Montreal, Quebec, Canada.
| | | |
Collapse
|
709
|
Costa-Almeida R, Reis RL, Gomes ME. Metabolic Disease Epidemics: Emerging Challenges in Regenerative Medicine. Trends Endocrinol Metab 2019; 30:147-149. [PMID: 30704823 DOI: 10.1016/j.tem.2019.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/11/2018] [Accepted: 01/03/2019] [Indexed: 12/11/2022]
Abstract
The interplay between cell/tissue damage caused by metabolic dysfunction and regenerative potential remains elusive. The tissue engineering and regenerative medicine (TERM) field is now facing a worldwide epidemic of obesity. This Forum article uncovers prospective questions to be addressed in TERM toward the development of effective regenerative therapies adjusted to these new demands.
Collapse
Affiliation(s)
- Raquel Costa-Almeida
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal
| | - Manuela E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal.
| |
Collapse
|
710
|
Berky AJ, Ryde IT, Feingold B, Ortiz EJ, Wyatt LH, Weinhouse C, Hsu-Kim H, Meyer JN, Pan WK. Predictors of mitochondrial DNA copy number and damage in a mercury-exposed rural Peruvian population near artisanal and small-scale gold mining: An exploratory study. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2019; 60:197-210. [PMID: 30289587 PMCID: PMC6452630 DOI: 10.1002/em.22244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/22/2018] [Accepted: 07/31/2018] [Indexed: 05/22/2023]
Abstract
Mitochondrial DNA (mtDNA) copy number (CN) and damage in circulating white blood cells have been proposed as effect biomarkers for pollutant exposures. Studies have shown that mercury accumulates in mitochondria and affects mitochondrial function and integrity; however, these data are derived largely from experiments in model systems, rather than human population studies that evaluate the potential utility of mitochondrial exposure biomarkers. We measured mtDNA CN and damage in white blood cells (WBCs) from 83 residents of nine communities in the Madre de Dios region of the Peruvian Amazon that vary in proximity to artisanal and small-scale gold mining. Prior research from this region reported high levels of mercury in fish and a significant association between food consumption and human total hair mercury level of residents. We observed that mtDNA CN and damage were both associated with consumption of fruit and vegetables, higher diversity of fruit consumed, residential location, and health characteristics, suggesting common environmental drivers. Surprisingly, we observed negative associations of mtDNA damage with both obesity and age. We did not observe any association between total hair mercury or, in contrast to previous results, age, with either mtDNA damage or CN. The results of this exploratory study highlight the importance of combining epidemiological and laboratory research in studying the effects of stressors on mitochondria, suggesting that future work should incorporate nutritional and social characteristics, and caution should be taken when applying conclusions from epidemiological studies conducted in the developed world to other regions, as results may not be easily translated. Environ. Mol. Mutagen. 60: 197-210, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Axel J. Berky
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
| | - Ian T. Ryde
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
| | - Beth Feingold
- University of Albany, School of Public Health, Rensselaer, NY 12144, USA
| | - Ernesto J. Ortiz
- Duke Global Health Institute, Duke University, 310 Trent Dr, Durham, NC 27710, USA
| | - Lauren H. Wyatt
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
| | - Caren Weinhouse
- Duke Global Health Institute, Duke University, 310 Trent Dr, Durham, NC 27710, USA
| | - Heileen Hsu-Kim
- Department of Civil and Engineering, Box 90287, Duke University, Durham, NC 27708, USA
| | - Joel N. Meyer
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
- Corresponding authors: William Pan, Duke Global Health Institute, 310 Trent Drive, Campus Box 90519, Durham, NC 27708, fax 681-7748, , Joel N. Meyer, Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA,
| | - William K. Pan
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
- Duke Global Health Institute, Duke University, 310 Trent Dr, Durham, NC 27710, USA
- Corresponding authors: William Pan, Duke Global Health Institute, 310 Trent Drive, Campus Box 90519, Durham, NC 27708, fax 681-7748, , Joel N. Meyer, Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA,
| |
Collapse
|
711
|
Townsend LK, Wright DC. Looking on the "brite" side exercise-induced browning of white adipose tissue. Pflugers Arch 2019; 471:455-465. [PMID: 29982948 DOI: 10.1007/s00424-018-2177-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/17/2022]
Abstract
The need for effective and convenient ways of combatting obesity has created great interest in brown adipose tissue (BAT). However, because adult humans have relatively little amounts of BAT, the possibility of browning white adipose tissue (WAT), i.e., switching the metabolism of WAT from an energy storing to energy burning organ, has gained considerable attention. Exercise has countless health benefits, and has consistently been shown to cause browning in rodent white adipose tissue. The purpose of this review is to provide an overview of recent studies examining the effects of exercise and other interventions on the browning of white adipose tissue. The role of various endocrine factors, including catecholamines, interleukin-6, irisin, and meteorin-like in addition to local re-esterification-mediated mechanisms in inducing the browning of WAT will be discussed. The physiological importance of browning will be discussed, as will discrepancies in the literature between human and rodent studies.
Collapse
Affiliation(s)
- Logan K Townsend
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Rd E, Guelph, Ontario, N1G 2W1, Canada
| | - David C Wright
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Rd E, Guelph, Ontario, N1G 2W1, Canada.
| |
Collapse
|
712
|
Lefere S, Tacke F. Macrophages in obesity and non-alcoholic fatty liver disease: Crosstalk with metabolism. JHEP Rep 2019; 1:30-43. [PMID: 32149275 PMCID: PMC7052781 DOI: 10.1016/j.jhepr.2019.02.004] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent liver disease worldwide, and a major cause of liver cirrhosis and hepatocellular carcinoma. NAFLD is intimately linked with other metabolic disorders characterized by insulin resistance. Metabolic diseases are driven by chronic inflammatory processes, in which macrophages perform essential roles. The polarization status of macrophages is itself influenced by metabolic stimuli such as fatty acids, which in turn affect the progression of metabolic dysfunction at multiple disease stages and in various tissues. For instance, adipose tissue macrophages respond to obesity, adipocyte stress and dietary factors by a specific metabolic and inflammatory programme that stimulates disease progression locally and in the liver. Kupffer cells and monocyte-derived macrophages represent ontologically distinct hepatic macrophage populations that perform a range of metabolic functions. These macrophages integrate signals from the gut-liver axis (related to dysbiosis, reduced intestinal barrier integrity, endotoxemia), from overnutrition, from systemic low-grade inflammation and from the local environment of a steatotic liver. This makes them central players in the progression of NAFLD to steatohepatitis (non-alcoholic steatohepatitis or NASH) and fibrosis. Moreover, the particular involvement of Kupffer cells in lipid metabolism, as well as the inflammatory activation of hepatic macrophages, may pathogenically link NAFLD/NASH and cardiovascular disease. In this review, we highlight the polarization, classification and function of macrophage subsets and their interaction with metabolic cues in the pathophysiology of obesity and NAFLD. Evidence from animal and clinical studies suggests that macrophage targeting may improve the course of NAFLD and related metabolic disorders.
Collapse
Affiliation(s)
- Sander Lefere
- Department of Gastroenterology and Hepatology, Hepatology Research Unit, Ghent University, Ghent, Belgium
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
- Department of Hepatology/Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Corresponding author. Address: Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany.
| |
Collapse
|
713
|
Xu H, Du X, Liu G, Huang S, Du W, Zou S, Tang D, Fan C, Xie Y, Wei Y, Tian Y, Fu X. The pseudokinase MLKL regulates hepatic insulin sensitivity independently of inflammation. Mol Metab 2019; 23:14-23. [PMID: 30837196 PMCID: PMC6480316 DOI: 10.1016/j.molmet.2019.02.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 02/05/2023] Open
Abstract
Objective The mixed lineage kinase domain like (MLKL) protein, receptor interacting protein (RIPK) 1, and RIPK3 are key regulators of necroptosis, a highly pro-inflammatory mode of cell death that has been implicated in various pathological processes and human diseases. However, the role of these necroptotic regulators in diabetes remains unknown. Here we sought to delineate the role of MLKL in insulin resistance and type 2 diabetes (T2D). Methods We first analyzed the expression of key necroptotic regulators in obese/diabetic mouse models. We then utilized MLKL knockout (MLKL−/−) mice to evaluate the effects of MLKL on obesity-induced metabolic complications. We further determined the consequences of MLKL inhibition on hepatic insulin signaling and explored the underlying mechanism. Finally, we assessed the potential therapeutic effects of necroptotic inhibitor, necrostatin-1 (Nec-1), in ob/ob mice. Results In wild-type or obese mice (ob/ob, db/db, or diet-induced obesity), MLKL was increased in certain obesity-associated tissues, particularly in the liver. Whole-body deficiency of MLKL prevented obesity-induced insulin resistance and glucose intolerance. Inhibition of MLKL or other key necroptotic regulators enhanced hepatic insulin sensitivity. MLKL modulated insulin-stimulated PI(3,4,5)P3 production in liver cells but did not affect the expression of inflammatory genes in vitro and in vivo. Nec-1 administration ameliorated insulin resistance and glucose intolerance in ob/ob mice. Conclusions These findings reveal MLKL as a regulator of insulin sensitivity and suggest necroptotic regulators might be potential therapeutic targets for insulin resistance and T2D. Hepatic MLKL is increased in diabetic mouse models. MLKL deficiency prevents obesity-induced metabolic complications. Inhibition of MLKL enhances insulin sensitivity independent of inflammation. Pharmacological inhibition of RIPK1 in ob/ob mice ameliorates insulin resistance.
Collapse
Affiliation(s)
- Haixia Xu
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, Sichuan, China
| | - Xiao Du
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Geng Liu
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, Sichuan, China
| | - Shuang Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Wenya Du
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, Sichuan, China
| | - Sailan Zou
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, Sichuan, China
| | - Dongmei Tang
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, Sichuan, China
| | - Chen Fan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yan Tian
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, Sichuan, China
| | - Xianghui Fu
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, Sichuan, China.
| |
Collapse
|
714
|
Shin CH, Choi DS. Essential Roles for the Non-Canonical IκB Kinases in Linking Inflammation to Cancer, Obesity, and Diabetes. Cells 2019; 8:cells8020178. [PMID: 30791439 PMCID: PMC6406369 DOI: 10.3390/cells8020178] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 12/17/2022] Open
Abstract
Non-canonical IκB kinases (IKKs) TBK1 and IKKε have essential roles as regulators of innate immunity and cancer. Recent work has also implicated these kinases in distinctively controlling glucose homeostasis and repressing adaptive thermogenic and mitochondrial biogenic response upon obesity-induced inflammation. Additionally, TBK1 and IKKε regulate pancreatic β-cell regeneration. In this review, we summarize current data on the functions and molecular mechanisms of TBK1 and IKKε in orchestrating inflammation to cancer, obesity, and diabetes.
Collapse
Affiliation(s)
- Chong Hyun Shin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
715
|
Lopez-Pastor AR, Gomez-Hernandez A, Diaz-Castroverde S, Gonzalez-Aseguinolaza G, Gonzalez-Rodriguez A, Garcia G, Fernandez S, Escribano O, Benito M. Liver-specific insulin receptor isoform A expression enhances hepatic glucose uptake and ameliorates liver steatosis in a mouse model of diet-induced obesity. Dis Model Mech 2019; 12:dmm.036186. [PMID: 30642871 PMCID: PMC6398497 DOI: 10.1242/dmm.036186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 01/03/2019] [Indexed: 12/12/2022] Open
Abstract
Among the main complications associated with obesity are insulin resistance and altered glucose and lipid metabolism within the liver. It has previously been described that insulin receptor isoform A (IRA) favors glucose uptake and glycogen storage in hepatocytes compared with isoform B (IRB), improving glucose homeostasis in mice lacking liver insulin receptor. Thus, we hypothesized that IRA could also improve glucose and lipid metabolism in a mouse model of high-fat-diet-induced obesity. We addressed the role of insulin receptor isoforms in glucose and lipid metabolism in vivo. We expressed IRA or IRB specifically in the liver by using adeno-associated viruses (AAVs) in a mouse model of diet-induced insulin resistance and obesity. IRA, but not IRB, expression induced increased glucose uptake in the liver and muscle, improving insulin tolerance. Regarding lipid metabolism, we found that AAV-mediated IRA expression also ameliorated hepatic steatosis by decreasing the expression of Fasn, Pgc1a, Acaca and Dgat2 and increasing Scd-1 expression. Taken together, our results further unravel the role of insulin receptor isoforms in hepatic glucose and lipid metabolism in an insulin-resistant scenario. Our data strongly suggest that IRA is more efficient than IRB at favoring hepatic glucose uptake, improving insulin tolerance and ameliorating hepatic steatosis. Therefore, we conclude that a gene therapy approach for hepatic IRA expression could be a safe and promising tool for the regulation of hepatic glucose consumption and lipid metabolism, two key processes in the development of non-alcoholic fatty liver disease associated with obesity. This article has an associated First Person interview with the first author of the paper. Summary: Adeno-associated-virus-mediated gene therapy for insulin receptor isoform A expression in the liver improves glucose disposal and alleviates lipid accumulation in wild-type mice under a high-fat diet.
Collapse
Affiliation(s)
- Andrea Raposo Lopez-Pastor
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Almudena Gomez-Hernandez
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain.,CIBER of Diabetes and Related Diseases (CIBERDEM), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain
| | - Sabela Diaz-Castroverde
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain.,CIBER of Diabetes and Related Diseases (CIBERDEM), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain
| | - Gloria Gonzalez-Aseguinolaza
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain
| | - Agueda Gonzalez-Rodriguez
- Liver Research Unit, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa, Amadeo Vives 2, 28009 Madrid, Spain.,CIBER of Hepatic and Digestive Diseases (CIBERehd), 28029 Madrid, Spain
| | - Gema Garcia
- CIBER of Diabetes and Related Diseases (CIBERDEM), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain
| | - Silvia Fernandez
- CIBER of Diabetes and Related Diseases (CIBERDEM), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain
| | - Oscar Escribano
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain .,CIBER of Diabetes and Related Diseases (CIBERDEM), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain.,CIBER of Diabetes and Related Diseases (CIBERDEM), Health Institute Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
716
|
Wang Q, Sharma VP, Shen H, Xiao Y, Zhu Q, Xiong X, Guo L, Jiang L, Ohta K, Li S, Shi H, Rui L, Lin JD. The hepatokine Tsukushi gates energy expenditure via brown fat sympathetic innervation. Nat Metab 2019; 1:251-260. [PMID: 31535079 PMCID: PMC6750233 DOI: 10.1038/s42255-018-0020-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/20/2018] [Indexed: 12/17/2022]
Abstract
Thermogenesis is an important contributor to whole body energy expenditure and metabolic homeostasis. Although circulating factors that promote energy expenditure are known, endocrine molecules that suppress energy expenditure have remained largely elusive. Here we show that Tsukushi (TSK) is a liver-enriched secreted factor that is highly inducible in response to increased energy expenditure. Hepatic Tsk expression and plasma TSK levels are elevated in obesity. TSK deficiency increases sympathetic innervation and norepinephrine release in adipose tissue, leading to enhanced adrenergic signaling and thermogenesis, attenuation of brown fat whitening and protection from diet-induced obesity in mice. Our work reveals TSK as part of a negative feedback mechanism that gates thermogenic energy expenditure and highlights TSK as a potential target for therapeutic intervention in metabolic disease.
Collapse
Affiliation(s)
- Qiuyu Wang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Vishal P Sharma
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Hong Shen
- Department of Molecular & Integrated Physiology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yuanyuan Xiao
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Qi Zhu
- Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, USA
| | - Xuelian Xiong
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Liang Guo
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Lin Jiang
- Department of Molecular & Integrated Physiology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Kunimasa Ohta
- Department of Developmental Neurobiology, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Siming Li
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Haifei Shi
- Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, USA
| | - Liangyou Rui
- Department of Molecular & Integrated Physiology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA.
| |
Collapse
|
717
|
Grzybek M, Palladini A, Alexaki VI, Surma MA, Simons K, Chavakis T, Klose C, Coskun Ü. Comprehensive and quantitative analysis of white and brown adipose tissue by shotgun lipidomics. Mol Metab 2019; 22:12-20. [PMID: 30777728 PMCID: PMC6437637 DOI: 10.1016/j.molmet.2019.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 12/12/2022] Open
Abstract
Objective Shotgun lipidomics enables an extensive analysis of lipids from tissues and fluids. Each specimen requires appropriate extraction and processing procedures to ensure good coverage and reproducible quantification of the lipidome. Adipose tissue (AT) has become a research focus with regard to its involvement in obesity-related pathologies. However, the quantification of the AT lipidome is particularly challenging due to the predominance of triacylglycerides, which elicit high ion suppression of the remaining lipid classes. Methods We present a new and validated method for shotgun lipidomics of AT, which tailors the lipid extraction procedure to the target specimen and features high reproducibility with a linear dynamic range of at least 4 orders of magnitude for all lipid classes. Results Utilizing this method, we observed tissue-specific and diet-related differences in three AT types (brown, gonadal, inguinal subcutaneous) from lean and obese mice. Brown AT exhibited a distinct lipidomic profile with the greatest lipid class diversity and responded to high-fat diet by altering its lipid composition, which shifted towards that of white AT. Moreover, diet-induced obesity promoted an overall remodeling of the lipidome, where all three AT types featured a significant increase in longer and more unsaturated triacylglyceride and phospholipid species. Conclusions The here presented method facilitates reproducible systematic lipidomic profiling of AT and could be integrated with further –omics approaches used in (pre-) clinical research, in order to advance the understanding of the molecular metabolic dynamics involved in the pathogenesis of obesity-associated disorders. Validated shotgun lipidomics method of AT covering 300 lipids of 20 classes and linear dynamic range of 4 orders of magnitude. Increase of longer and more unsaturated triacylglycerides and phospholipids in brown and white AT under high-fat diet. Differences in the lipidomes of gonadal, subcutaneous and brown AT.
Collapse
Affiliation(s)
- Michal Grzybek
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Alessandra Palladini
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Vasileia I Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, Dresden, Germany
| | | | | | - Triantafyllos Chavakis
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, Dresden, Germany
| | | | - Ünal Coskun
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
| |
Collapse
|
718
|
Petimar J, Tabung FK, Valeri L, Rosner B, Chan AT, Smith-Warner SA, Giovannucci EL. Mediation of associations between adiposity and colorectal cancer risk by inflammatory and metabolic biomarkers. Int J Cancer 2019; 144:2945-2953. [PMID: 30521066 DOI: 10.1002/ijc.32047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 11/01/2018] [Accepted: 11/22/2018] [Indexed: 01/20/2023]
Abstract
Inflammation and hyperinsulinemia may drive associations between adiposity and colorectal cancer (CRC) risk, but few studies have examined this hypothesis using mediation analysis. We used inverse odds ratio weighting and logistic regression to calculate odds ratios (OR) and 95% confidence intervals (CI) for estimated total effects (ORTE ) of body mass index, waist circumference, and adult weight gain on CRC risk, and estimated effects operating through seven inflammatory and metabolic biomarkers (natural indirect effect; ORNIE ) or through paths independent of these biomarkers (natural direct effect; ORNDE ) among 209 CRC cases and 382 matched controls nested within the Health Professionals Follow-up Study, a prospective cohort of male health professionals. A one-interquartile range (IQR) increase in body mass index (3.6 kg/m2 ) was associated with an ORTE of 1.40 (95% CI: 1.13, 1.73), which decomposed into an ORNIE of 1.26 (95% CI: 0.97, 1.52) and an ORNDE of 1.11 (0.87, 1.42), with possibly stronger mediation by these biomarkers for adult weight gain (IQR = 10.4 kg; ORTE = 1.32 [95% CI: 1.06, 1.64]; ORNIE = 1.47 [95% CI: 1.01, 1.81]; ORNDE = 0.89 [95% CI: 0.72, 1.11]), but no mediation for waist circumference. Mediation appeared to be stronger for the metabolic biomarkers than the inflammatory biomarkers. Inflammatory and metabolic mechanisms may mediate associations between both body mass index and adult weight gain with CRC risk.
Collapse
Affiliation(s)
- Joshua Petimar
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Fred K Tabung
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Division of Medical Oncology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH
| | - Linda Valeri
- Department of Psychiatry, Harvard Medical School, Boston, MA.,Psychiatric Biostatistics Laboratory, McLean Hospital, Belmont, MA.,Department of Biostatistics, Columbia University, New York, NY
| | - Bernard Rosner
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Andrew T Chan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Division of Gastroenterology, Massachusetts General Hospital, Boston, MA
| | - Stephanie A Smith-Warner
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
719
|
Castellano-Castillo D, Moreno-Indias I, Sanchez-Alcoholado L, Ramos-Molina B, Alcaide-Torres J, Morcillo S, Ocaña-Wilhelmi L, Tinahones F, Queipo-Ortuño MI, Cardona F. Altered Adipose Tissue DNA Methylation Status in Metabolic Syndrome: Relationships Between Global DNA Methylation and Specific Methylation at Adipogenic, Lipid Metabolism and Inflammatory Candidate Genes and Metabolic Variables. J Clin Med 2019; 8:jcm8010087. [PMID: 30642114 PMCID: PMC6352101 DOI: 10.3390/jcm8010087] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 02/07/2023] Open
Abstract
Metabolic syndrome (MetS) has been postulated to increase the risk for type 2 diabetes, cardiovascular disease and cancer. Adipose tissue (AT) plays an important role in metabolic homeostasis, and AT dysfunction has an active role in metabolic diseases. MetS is closely related to lifestyle and environmental factors. Epigenetics has emerged as an interesting landscape to evaluate the possible interconnection between AT and metabolic disease, since it can be modulated by environmental factors and metabolic status. The aim of this study was to determine whether MetS has an impact on the global DNA methylation pattern and the DNA methylation of several genes related to adipogenesis (PPARG, PPARA), lipid metabolism (RXRA, SREBF2, SREBF1, SCD, LPL, LXRb), and inflammation (LRP1 C3, LEP and TNF) in visceral adipose tissue. LPL and TNF DNA methylation values were significantly different in the control-case comparisons, with higher and lower methylation respectively in the MetS group. Negative correlations were found between global DNA methylation (measured by LINE-1 methylation levels) and the metabolic deterioration and glucose levels. There were associations among variables of MetS, BMI, and HOMA-IR with DNA methylation at several CpG positions for the studied genes. In particular, there was a strong positive association between serum triglyceride levels (TG) with PPARA and LPL methylation levels. TNF methylation was negatively associated with the metabolic worsening and could be an important factor in preventing MetS occurrence according to logistic regression analysis. Therefore, global DNA methylation and methylation at specific genes related to adipogenesis, lipid metabolism and inflammation are related to the etiology of MetS and might explain in part some of the features associated to metabolic disorders.
Collapse
Affiliation(s)
- Daniel Castellano-Castillo
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, 28029 Madrid, Spain.
| | - Isabel Moreno-Indias
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, 28029 Madrid, Spain.
| | - Lidia Sanchez-Alcoholado
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, 28029 Madrid, Spain.
| | - Bruno Ramos-Molina
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, 28029 Madrid, Spain.
| | - Juan Alcaide-Torres
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, 28029 Madrid, Spain.
| | - Sonsoles Morcillo
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, 28029 Madrid, Spain.
| | - Luis Ocaña-Wilhelmi
- Unidad de Cirugía Metabólica, Hospital Clínico Virgen de la Victoria, 29010 Málaga, Spain.
| | - Francisco Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, 28029 Madrid, Spain.
| | - María Isabel Queipo-Ortuño
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, 28029 Madrid, Spain.
- Unidad de Gestión Clínica de Oncología Médica del Hospital Virgen de la Victoria, 29010 Málaga, Spain.
| | - Fernando Cardona
- Unidad de Gestión Clínica de Endocrinología y Nutrición del Hospital Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, 29010 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, CIBERobn, 28029 Madrid, Spain.
| |
Collapse
|
720
|
Pîrsean C, Neguț C, Stefan-van Staden RI, Dinu-Pirvu CE, Armean P, Udeanu DI. The salivary levels of leptin and interleukin-6 as potential inflammatory markers in children obesity. PLoS One 2019; 14:e0210288. [PMID: 30605486 PMCID: PMC6317816 DOI: 10.1371/journal.pone.0210288] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/19/2018] [Indexed: 01/04/2023] Open
Abstract
Background Obesity among children is an alarming issue due to an increased incidence over the last years with devastating physiological and psychological consequences. Current available medical diagnostic tools use invasive methods to evaluate and monitor the lipid profile, glycaemia or liver status for determining the overweight/ obesity complications. The standard methods proposed for the assay of IL6 and leptin from saliva cannot detect these two biomarkers in children saliva; the levels of IL6 and leptin in children’s saliva are lower than the limit of determination of the standard methods. Therefore, we proposed a method based on utilization of stochastic sensors, able to simultaneously perform a qualitative and quantitative determination of these two biomarkers within minutes, in the range able to cover healthy and obese children. Methods Children from the urban area monitored for annual standard analyses and health status assessment at National Institute of Endocrinology C.I. Parhon within University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania were included in the study. In the same day, for all participants of the study, blood analyses were performed and saliva samples were collected for the determination of the IL-6 and leptin levels. Findings/ Results The children diagnosed with overweight/ obesity presented not significantly different blood lipid profile and glycaemia comparing to the control group. Only few cases of the children presented high levels of cholesterol, low level of HDL-cholesterol, a slightly increased level of triglycerides and transaminases. No correlation with the body mass index could be established with the blood analyses results. In case of the overweight/obese children, the salivary level of the proinflammatory citokynes IL-6 (41ng/mL±21) and leptin (40.4ng/mL±28.8), were significantly increased comparing to normal weight children (IL-6 8.1±4.6, leptin 9.58±3.1). Moreover, the saliva level of the IL-6 was positively correlated with the body mass index. Salivary leptin level was highly variable in case of obese children, 6 patients presenting similar levels with the control group. Conclusions Increased levels of salivary IL-6 and leptin sustain a systemic inflammation status despite normal range of standard blood analyses. The results were positively correlated in case of IL-6 with the body mass index the general accepted method used for the assessment of the obesity or overweight degree The determination of these markers in saliva samples by a stochastic method proved the utility within the medical examination for a better evaluation of the health status in obesity. The method has some advantages like: easy collection of the biological sample, fast determination of low concentrations and could be promising in case of no associated oral cavity infections or inflammations which could interfere the results.
Collapse
Affiliation(s)
- Corina Pîrsean
- School of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Cătălina Neguț
- Laboratory of Electrochemistry and PATLAB Bucharest, National Institute of Research for Electrochemistry and Condensed Matter, Bucharest, Romania
| | - Raluca-Ioana Stefan-van Staden
- Laboratory of Electrochemistry and PATLAB Bucharest, National Institute of Research for Electrochemistry and Condensed Matter, Bucharest, Romania
- * E-mail:
| | - Cristina Elena Dinu-Pirvu
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Petru Armean
- School of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Denisa Ioana Udeanu
- Department of Clinical Laboratory. Food Safety, Faculty of Pharmacy, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| |
Collapse
|
721
|
Perugini J, Di Mercurio E, Tossetta G, Severi I, Monaco F, Reguzzoni M, Tomasetti M, Dani C, Cinti S, Giordano A. Biological Effects of Ciliary Neurotrophic Factor on hMADS Adipocytes. Front Endocrinol (Lausanne) 2019; 10:768. [PMID: 31781039 PMCID: PMC6861295 DOI: 10.3389/fendo.2019.00768] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022] Open
Abstract
Administration of ciliary neurotrophic factor (CNTF) to experimental animals exerts anti-obesity effects by acting on multiple targets. In white adipose tissue CNTF reduces lipid content, promotes fatty acid (FA) oxidation and improves insulin sensitivity. This study was performed to establish whether CNTF exerts similar effects on human white adipocytes. To this end, adipose differentiation was induced in vitro in human multipotent adipose-derived stem (hMADS) cells. CNTF receptor α (CNTFRα) expression was assessed in hMADS cells and adipocytes by qRT-PCR, Western blotting, and immunocytochemistry. After administration of human recombinant CNTF, signaling pathways and gene expression were evaluated by Western blotting and qRT-PCR. Glucose uptake was assessed by measuring 2-nitrobenzodeoxyglucose uptake with a fluorescence plate reader. Lastly, CNTF-induced anti-inflammatory responses were evaluated in hMADS adipocytes stressed with tumor necrosis factor α (TNFα) for 24 h. Results showed that CNTFRα protein expression was higher in undifferentiated hMADS cells than in hMADS adipocytes, where it was however clearly detectable. In hMADS adipocytes, 1 nM CNTF strongly activated the JAK-STAT3 (Janus kinase-signaling transducer and activator of transcription 3) pathway and acutely and transiently activated the AMPK (AMP-activated protein kinase) and AKT (protein kinase B) pathways. Acute CNTF treatment for 20 min significantly increased basal glucose uptake and was associated with increased AKT phosphorylation. Longer-term (24 and 48 h) treatment reduced the expression of lipogenic markers (FA synthase and sterol regulatory element-binding protein-1) and increased the expression of lipolytic [hormone-sensitive lipase (HSL) and adipose triglyceride lipase (ATGL)] and mitochondrial (peroxisome proliferator-activated receptor γ coactivator-1α and carnitine palmitoyltransferase 1) markers. In TNFα-treated hMADS adipocytes, CNTF significantly reduced the expression of monocyte chemoattractant protein 1 and TNFα-induced AKT inhibition. Collectively, these findings demonstrate for the first time that CNTF plays a role also in human adipocytes, driving their metabolism toward a less lipid-storing and more energy-consuming phenotype.
Collapse
Affiliation(s)
- Jessica Perugini
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Eleonora Di Mercurio
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Ilenia Severi
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Federica Monaco
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Marcella Reguzzoni
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Marco Tomasetti
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Christian Dani
- Université Côte d'Azur, CNRS, INSERM, iBV, Faculté de Médecine, Nice, France
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
- Center of Obesity, United Hospitals, Marche Polytechnic University, Ancona, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
- *Correspondence: Antonio Giordano
| |
Collapse
|
722
|
Meydan C. Population-based data registries suggest novel insight into malignancy and metabolism. Cancer 2019; 125:15-17. [PMID: 30417327 DOI: 10.1002/cncr.31762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/07/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Chanan Meydan
- Department of Internal Medicine, Mayanei Hayeshua Medical Center, Bnei Brak, Israel
| |
Collapse
|
723
|
Gonzalez-Gil AM, Peschard-Franco M, Castillo EC, Gutierrez-DelBosque G, Treviño V, Silva-Platas C, Perez-Villarreal L, Garcia-Rivas G, Elizondo-Montemayor L. Myokine-adipokine cross-talk: potential mechanisms for the association between plasma irisin and adipokines and cardiometabolic risk factors in Mexican children with obesity and the metabolic syndrome. Diabetol Metab Syndr 2019; 11:63. [PMID: 31404407 PMCID: PMC6683550 DOI: 10.1186/s13098-019-0458-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/25/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Adipokines and the myokine irisin, involved in mechanisms associated with obesity and metabolic syndrome (MS), are understudied in the pediatric population. OBJECTIVE To investigate the relationship between irisin, and leptin, resistin, adiponectin, adipsin, anthropometric and cardiovascular risk factors in Mexican children. METHODS A cross-sample of 126 Mexican children aged 6-12 years old were classified as normal weight (n = 46), obese (n = 40), and MS (n = 40) according to CDC's and Cook's age-modified criteria for obesity and MS. Anthropometric parameters and blood pressure were determined and percentiles calculated for age and gender. Irisin, leptin, adiponectin, adipsin, resistin, triglycerides, glucose, high-density lipoprotein cholesterol (HDL-c) levels, and physical activity were determined. Statistical tests for differences between groups, correlation, and multiple regression analyses were performed. RESULTS Irisin plasma levels were significantly lower in the obese (6.08 [4.68-6.65]) and MS groups (6.46 [5.74-7.02]) compared with the normal-weight group (8.05 [7.24-8.94]) (p < 0.001). Irisin levels were not influenced by age or gender, but significant dispersion was observed in obese girls (95% CI median [2.29-6.30]). Leptin, resistin, and adipsin levels were significantly increased in the obese and MS groups. Lean-fat ratio was significantly higher in the NW group. Irisin correlated negatively with leptin (- 0.310), resistin (- 0.389), adipsin (- 0.362), BMI% (-0.472), WC% (- 0.453), BMI z-score (- 0.496), fat free mass (- 0.257), fat percentage (- 0.532), fat mass (- 0.515), triglycerides (- 0.291), the number of cardiometabolic risk factors (- 0.443) (p < 0.001); positively with lean-fat ratio (0.489) and HDL-c (0.328) (p < 0.001) and none with physical activity (p < 0.001). Following stepwise multiple linear regression analysis, the lean-fat ratio was the only determinant of irisin levels (B = 1.168, p < 0.001). CONCLUSIONS Lean-fat ratio, more than the absolute amount of muscle or fat mass, as well as potential myokine-adipokine cross-talk mechanisms may explain the lower irisin levels in children with obesity and MS, through blunted compensatory responses interfering with tissue-dependent irisin secretion, contributing to a continuous deleterious effect cycle.
Collapse
Affiliation(s)
- Adrian M. Gonzalez-Gil
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Monterrey, N.L. Mexico
- Center for Research in Clinical Nutrition and Obesity, Tecnologico de Monterrey, Ave. Morones Prieto 300, 64710 Monterrey, N.L. Mexico
| | - Mariana Peschard-Franco
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Monterrey, N.L. Mexico
- Center for Research in Clinical Nutrition and Obesity, Tecnologico de Monterrey, Ave. Morones Prieto 300, 64710 Monterrey, N.L. Mexico
| | - Elena C. Castillo
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Monterrey, N.L. Mexico
- Cardiovascular and Metabolomics Research Group, Hospital Zambrano Hellion, Tecnologico de Monterrey, 66278 San Pedro Garza Garcia, P.C. Mexico
| | - Gustavo Gutierrez-DelBosque
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Monterrey, N.L. Mexico
- Center for Research in Clinical Nutrition and Obesity, Tecnologico de Monterrey, Ave. Morones Prieto 300, 64710 Monterrey, N.L. Mexico
| | - Victor Treviño
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Monterrey, N.L. Mexico
| | - Christian Silva-Platas
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Monterrey, N.L. Mexico
- Cardiovascular and Metabolomics Research Group, Hospital Zambrano Hellion, Tecnologico de Monterrey, 66278 San Pedro Garza Garcia, P.C. Mexico
| | - Luisa Perez-Villarreal
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Monterrey, N.L. Mexico
- Center for Research in Clinical Nutrition and Obesity, Tecnologico de Monterrey, Ave. Morones Prieto 300, 64710 Monterrey, N.L. Mexico
| | - Gerardo Garcia-Rivas
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Monterrey, N.L. Mexico
- Cardiovascular and Metabolomics Research Group, Hospital Zambrano Hellion, Tecnologico de Monterrey, 66278 San Pedro Garza Garcia, P.C. Mexico
| | - Leticia Elizondo-Montemayor
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Ave. Morones Prieto 3000, 64710 Monterrey, N.L. Mexico
- Center for Research in Clinical Nutrition and Obesity, Tecnologico de Monterrey, Ave. Morones Prieto 300, 64710 Monterrey, N.L. Mexico
- Cardiovascular and Metabolomics Research Group, Hospital Zambrano Hellion, Tecnologico de Monterrey, 66278 San Pedro Garza Garcia, P.C. Mexico
| |
Collapse
|
724
|
Palhinha L, Liechocki S, Hottz ED, Pereira JADS, de Almeida CJ, Moraes-Vieira PMM, Bozza PT, Maya-Monteiro CM. Leptin Induces Proadipogenic and Proinflammatory Signaling in Adipocytes. Front Endocrinol (Lausanne) 2019; 10:841. [PMID: 31920961 PMCID: PMC6923660 DOI: 10.3389/fendo.2019.00841] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Leptin is an adipokine with well-known effects on the central nervous system including the induction of energy expenditure and satiety. Leptin also has major relevance when activating immune cells and modulating inflammatory response. In obesity, increases in white adipose tissue accumulation and leptin levels are accompanied by hypothalamic resistance to leptin. Even though the adipose tissue is a leptin-rich environment, the local actions of leptin regarding adipogenesis were not thoroughly investigated until now. Here we evaluate the contributions of leptins direct signaling in preadipocytes and adipose tissue-derived stromal cells (ASCs) for adipogenesis. Methods: Adipocytes were differentiated from the murine lineage of preadipocytes 3T3-L1 or ASCs from subcutaneous and visceral (retroperitoneal) fat depots from C57Bl/6J mice. Differentiating cells were treated with leptin in addition to or in replacement of insulin. The advance of adipogenesis was assessed by the expression and secretion of adipogenesis- and lipogenesis-related proteins by Western blot and immunoenzimatic assays, and the accumulation of lipid droplets by fluorescence microscopy. Results: Leptin treatment in 3T3-L1 preadipocytes or ASCs increased the production of the adipogenesis- and lipogenesis-related proteins PLIN1, CAV-1, PPARγ, SREBP1C, and/or adiponectin at earlier stages of differentiation. In 3T3-L1 preadipocytes, we found that leptin induced lipid droplets' formation in an mTOR-dependent manner. Also, leptin induced a proinflammatory cytokine profile in 3T3-L1 and ASCs, modulating the production of TNF-α, IL-10, and IL-6. Since insulin is considered an essential factor for preadipocyte differentiation, we asked whether leptin would support adipogenesis in the absence of insulin. Importantly, leptin induced the formation of lipid droplets and the expression of adipogenesis-related proteins independently of insulin during the differentiation of 3T3-L1 cells and ASCs. Conclusions: Our results demonstrate that leptin induces intracellular signaling in preadipocytes and adipocytes promoting adipogenesis and modulating the secretion of inflammatory mediators. Also, leptin restores adipogenesis in the absence of insulin. These findings contribute to the understanding of the local signaling of leptin in precursor and mature adipose cells. The proadipogenic role of leptin unraveled here may be of especial relevance during obesity, when its central signaling is defective.
Collapse
Affiliation(s)
- Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Sally Liechocki
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Eugenio D. Hottz
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratory of Glycoconjugates Analysis, Department of Biochemistry, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Brazil
| | - Jéssica Aparecida da Silva Pereira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Post-Graduate Program in Immunology, Institute of Biological Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Cecília J. de Almeida
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Pedro Manoel M. Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Post-Graduate Program in Immunology, Institute of Biological Sciences, University of Sao Paulo, São Paulo, Brazil
- Experimental Medicine Research Cluster, EMRC, University of Cammpinas, Campinas, Brazil
| | - Patrícia T. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Clarissa Menezes Maya-Monteiro
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- *Correspondence: Clarissa Menezes Maya-Monteiro ;
| |
Collapse
|
725
|
Cho YL, Park JG, Kang HJ, Kim W, Cho MJ, Jang JH, Kwon MG, Kim S, Lee SH, Lee J, Kim YG, Park YJ, Kim WK, Bae KH, Kwon BM, Chung SJ, Min JK. Ginkgetin, a biflavone from Ginkgo biloba leaves, prevents adipogenesis through STAT5-mediated PPARγ and C/EBPα regulation. Pharmacol Res 2019; 139:325-336. [DOI: 10.1016/j.phrs.2018.11.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/28/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022]
|
726
|
Oliveira CT, Lacerda DR, Zicker MC, Martins LB, Teixeira MM, de Araujo RLB, Ferreira AVM. Ginger (Zingiber officinale Rosc.) Ameliorated Metabolic and Inflammatory Dysfunction Induced by High-Refined Carbohydrate-Containing Diet in Mice. J Med Food 2019; 22:38-45. [PMID: 30362875 DOI: 10.1089/jmf.2018.0062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This study aimed to evaluate the effects and the mechanisms of ginger extract intake in the adiposity gain, metabolic and inflammatory disturbances induced by a high-refined carbohydrate (HC) diet in mice. Ginger extract at doses of 200, 600, and 1800 mg/kg was supplemented in the daily food of obese Balb/c mice during an 8-week experiment. Our findings indicate that consumption of high doses of ginger extracts prevents the increase of adiposity induced by HC diet, improves lipid profile, and promotes decrease of inflammatory markers in mice. We showed that ginger addition to HC diet leads to decrease in the recruitment of cells visualized in vivo in the microvasculature of adipose tissue, decrease of inflammatory cytokines, and increase of adiponectin serum levels. These results indicate that the consumption of ginger decreases the negative metabolic consequences induced by HC diet.
Collapse
Affiliation(s)
- Cíntia Tarabal Oliveira
- 1 Department of Food Science, Faculty of Pharmacy, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Débora Romualdo Lacerda
- 1 Department of Food Science, Faculty of Pharmacy, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Marina Campos Zicker
- 1 Department of Food Science, Faculty of Pharmacy, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Laís Bhering Martins
- 1 Department of Food Science, Faculty of Pharmacy, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- 2 Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | | |
Collapse
|
727
|
Mantelmacher FD, Zvibel I, Cohen K, Epshtein A, Pasmanik-Chor M, Vogl T, Kuperman Y, Weiss S, Drucker DJ, Varol C, Fishman S. GIP regulates inflammation and body weight by restraining myeloid-cell-derived S100A8/A9. Nat Metab 2019; 1:58-69. [PMID: 32694806 DOI: 10.1038/s42255-018-0001-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/01/2018] [Indexed: 12/25/2022]
Abstract
Enteroendocrine cells relay energy-derived signals to immune cells to signal states of nutrient abundance and control immunometabolism. Emerging data suggest that the gut-derived nutrient-induced incretin glucose-dependent insulinotropic polypeptide (GIP) operates at the interface of metabolism and inflammation. Here we show that high-fat diet (HFD)-fed mice with immune cell-targeted GIP receptor (GIPR) deficiency exhibit greater weight gain, insulin resistance, hepatic steatosis and significant myelopoiesis concomitantly with impaired energy expenditure and inguinal white adipose tissue (WAT) beiging. Expression of the S100 calcium-binding protein S100A8 was increased in the WAT of mice with immune cell-targeted GIPR deficiency and co-deletion of GIPR and the heterodimer S100A8/A9 in immune cells ameliorated the aggravated metabolic and inflammatory phenotype following a HFD. Specific GIPR deletion in myeloid cells identified this lineage as the target of GIP effects. Furthermore, GIP directly downregulated S100A8 expression in adipose tissue macrophages. Collectively, our results identify a myeloid-GIPR-S100A8/A9 signalling axis coupling nutrient signals to the control of inflammation and adaptive thermogenesis.
Collapse
Affiliation(s)
- Fernanda Dana Mantelmacher
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Isabel Zvibel
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Keren Cohen
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Alona Epshtein
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Shai Weiss
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Sigal Fishman
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
728
|
Abstract
Obesity is a risk factor for all major gastrointestinal cancers. With the rapid increase in the prevalence of obesity worldwide, this link could lead to an elevated burden of cancers of the digestive system. Currently, three main mechanisms explaining the link between excess adiposity and gastrointestinal cancer risk are being considered, including altered insulin signaling, obesity-associated chronic low-grade inflammation, and altered sex hormone metabolism, although new potential mechanisms emerge. This review is aimed to present our current knowledge on biological mechanisms involved in adiposity-related gastrointestinal carcinogenesis supported by results collected in epidemiological studies.
Collapse
|
729
|
Zhao C, Sun Q, Tang L, Cao Y, Nourse JL, Pathak MM, Lu X, Yang Q. Mechanosensitive Ion Channel Piezo1 Regulates Diet-Induced Adipose Inflammation and Systemic Insulin Resistance. Front Endocrinol (Lausanne) 2019; 10:373. [PMID: 31263454 PMCID: PMC6584899 DOI: 10.3389/fendo.2019.00373] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022] Open
Abstract
Adipocytes function as an energy buffer and undergo significant size and volume changes in response to nutritional cues. This adipocyte plasticity is important for systemic lipid metabolism and insulin sensitivity. Accompanying the adipocyte size and volume changes, the mechanical pressure against cell membrane also changes. However, the role that mechanical pressure plays in lipid metabolism and insulin sensitivity remains to be elucidated. Here we show that Piezo1, a mechanically-activated cation channel stimulated by membrane tension and stretch, was highly expressed in adipocytes. Adipose Piezo1 expression was increased in obese mice. Adipose-specific piezo1 knockout mice (adipose-Piezo1-/-) developed insulin resistance, especially when challenged with a high-fat diet (HFD). Perigonadal white adipose tissue (pgWAT) weight was reduced while pro-inflammatory and lipolysis genes were increased in the pgWAT of HFD-fed adipose-Piezo1-/- mice. The adipose-Piezo1-/- mice also developed hepatic steatosis with elevated expression of fatty acid synthesis genes. In cultured adipocytes, Piezo1 activation decreased, while Piezo1 inhibition elevated pro-inflammatory gene expression. TLR4 antagonist TAK-242 abolished adipocyte inflammation induced by Piezo1 inhibition. Thus, adipose Piezo1 may serve as an adaptive mechanism for adipocyte plasticity restraining pro-inflammatory response in obesity.
Collapse
Affiliation(s)
- Can Zhao
- Department of Geriatrics, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
- Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, China
| | - Qiushi Sun
- Department of Geriatrics, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
| | - Lingyi Tang
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
- Department of Cardiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Cao
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
| | - Jamison L. Nourse
- Department of Physiology and Biophysics, Sue and Bill Gross Stem Cell Research Center, Center for Complex Systems Biology, University of California at Irvine, Irvine, CA, United States
| | - Medha M. Pathak
- Department of Physiology and Biophysics, Sue and Bill Gross Stem Cell Research Center, Center for Complex Systems Biology, University of California at Irvine, Irvine, CA, United States
| | - Xiang Lu
- Department of Geriatrics, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
- Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, China
- *Correspondence: Xiang Lu
| | - Qin Yang
- Department of Medicine, Physiology and Biophysics, UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA, United States
- Qin Yang
| |
Collapse
|
730
|
Vukovic R, Dos Santos TJ, Ybarra M, Atar M. Children With Metabolically Healthy Obesity: A Review. Front Endocrinol (Lausanne) 2019; 10:865. [PMID: 31920976 PMCID: PMC6914809 DOI: 10.3389/fendo.2019.00865] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023] Open
Abstract
Children with "metabolically healthy obesity" (MHO) are a distinct subgroup of youth with obesity, who are less prone to the clustering of cardiometabolic risk factors. Although this phenotype, frequently defined by the absence of metabolic syndrome components or insulin resistance, was first described during the early 1980s, a consensus-based definition of pediatric MHO was introduced only recently, in 2018. The purpose of this review was to concisely summarize current knowledge regarding the MHO phenomenon in youth. The prevalence of MHO in children varies from 3 to 87%, depending on the definition used and the parameters evaluated, as well as the ethnicity and the pubertal status of the sample. The most consistent predictors of MHO in youth include younger age, lower body mass index, lower waist circumference, and lower body fat measurements. Various hypotheses have been proposed to elucidate the underlying factors maintaining the favorable MHO phenotype. While preserved insulin sensitivity and lack of inflammation were previously considered to be the main etiological factors, the most recent findings have implicated adipokine levels, the number of inflammatory immune cells in the adipose tissue, and the reduction of visceral adiposity due to adipose tissue expandability. Physical activity and genetic factors also contribute to the MHO phenotype. Obesity constitutes a continuum-increased risk for cardiometabolic complications, which is less evident in children with MHO. However, some findings have highlighted the emergence of hepatic steatosis, increased carotid intima-media thickness and inflammatory biomarkers in the MHO group compared to peers without obesity. Screening should be directed at those more likely to develop clustering of cardiometabolic risk factors. Lifestyle modifications should include behavioral changes focusing on sleep duration, screen time, diet, physical activity, and tobacco smoke exposure. Weight loss has also been associated with the improvement of insulin sensitivity and inflammation. Further investigative efforts are needed in order to elucidate the mechanisms which protect against the clustering of cardiometabolic risk factors in pediatric obesity, to provide more efficient, targeted treatment approaches for children with obesity, and to identify the protective factors preserving the MHO profile, avoiding the crossover of MHO to the phenotype with metabolically unhealthy obesity.
Collapse
Affiliation(s)
- Rade Vukovic
- Department of Pediatric Endocrinology, Mother and Child Healthcare Institute of Serbia “Dr Vukan Cupic”, Belgrade, Serbia
- School of Medicine, University of Belgrade, Belgrade, Serbia
- *Correspondence: Rade Vukovic
| | | | - Marina Ybarra
- Research Center of Sainte Justine University Hospital, Université de Montréal, Montreal, QC, Canada
- Centre Armand-Frappier, Institut National de la Recherche Scientifique, Université du Québec, Laval, QC, Canada
| | - Muge Atar
- Department of Pediatric Endocrinology, School of Medicine, Demirel University, Isparta, Turkey
| |
Collapse
|
731
|
Bastin M, Andreelli F. Dual GIP-GLP1-Receptor Agonists In The Treatment Of Type 2 Diabetes: A Short Review On Emerging Data And Therapeutic Potential. Diabetes Metab Syndr Obes 2019; 12:1973-1985. [PMID: 31686879 PMCID: PMC6777434 DOI: 10.2147/dmso.s191438] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/17/2019] [Indexed: 01/11/2023] Open
Abstract
The need for efficient and safe therapy to improve such metabolic diseases as obesity and type 2 diabetes mellitus is currently unmet. The development of dual GIPR-GLP1R coagonists that bind to either one or the other receptor (sequence-mixed dual agonists) has emerged as an innovative therapeutic strategy for obesity and type 2 diabetes. Combined activation of both receptors may act synergistically providing additive effects on glucose and body weight in comparison of GLP1 analogues alone. Preclinical studies have confirmed that GIPR-GLP1R coagonists improve several hallmarks of metabolic syndrome, such as obesity, hyperglycemia, and dyslipidemia. These metabolic benefits have been translated from mice to nonhuman primates and humans. Recent clinical trials have shown that coagonists induce significant benefits on body weight, fasting, and postprandial glucose levels, insulin sensitivity, and total cholesterol. Combined GIP- and GLP1R activators have the potential to become a treatment option for patients with type 2 diabetes.
Collapse
Affiliation(s)
- Marie Bastin
- Diabetology-Metabolism Department, Sorbonne Université, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, F-75013, France
| | - Fabrizio Andreelli
- Diabetology-Metabolism Department, Sorbonne Université, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, F-75013, France
- Correspondence: Fabrizio Andreelli Diabetology-Metabolism Department, Pitié-Salpêtrière Hospital, 47-83 Boulevard de l’Hôpital, Paris cedex 13, 75951, FranceEmail
| |
Collapse
|
732
|
Reiche ME, den Toom M, Willemsen L, van Os B, Gijbels MJJ, Gerdes N, Aarts SABM, Lutgens E. Deficiency of T cell CD40L has minor beneficial effects on obesity-induced metabolic dysfunction. BMJ Open Diabetes Res Care 2019; 7:e000829. [PMID: 31908798 PMCID: PMC6936585 DOI: 10.1136/bmjdrc-2019-000829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/30/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Obesity-associated metabolic dysfunction increases the risk of multiple diseases such as type 2 diabetes and cardiovascular disease. The importance of the co-stimulatory CD40-CD40L dyad in diet-induced obesity (DIO), with opposing phenotypes arising when either the receptor (aggravating) or the ligand (protective) is deleted, has been described previously. The functions of CD40 and CD40L are cell type dependent. As co-stimulation via T cell-mediated CD40L is essential for driving inflammation, we here investigate the role of T cell CD40L in DIO. RESEARCH DESIGN AND METHODS CD4CreCD40Lfl/fl mice on a C57BL/6 background were generated and subjected to DIO by administration of 15 weeks of high fat diet (HFD). RESULTS HFD-fed CD4CreCD40Lfl/fl mice had similar weight gain, adipocyte sizes, plasma cholesterol and triglyceride levels as their wild-type (WT) counterparts. Insulin and glucose tolerance were comparable, although CD4CreCD40Lfl/fl mice did have a decreased plasma insulin concentration, suggesting a minor improvement of insulin resistance. Furthermore, although the degree of hepatosteatosis was similar in both genotypes, the gene expression of fatty acid synthase 1 and ATP-citrate lyase had decreased, whereas expression of peroxisome proliferator-activated receptor-α had increased in livers of CD4CreCD40Lfl/fl mice, suggesting decreased hepatic lipid uptake in absence of T cell CD40L.Moreover, CD4CreCD40Lfl/fl mice displayed significantly lower numbers of effector memory CD4+ T cells and regulatory T cells in blood and lymphoid organs compared with WT. However, immune cell composition and inflammatory status of the adipose tissue was similar in CD4CreCD40Lfl/fl and WT mice. CONCLUSIONS T cell CD40L deficiency results in a minor improvement of insulin sensitivity and hepatic steatosis in DIO, despite the strong decrease in effector T cells and regulatory T cells in blood and lymphoid organs. Our data indicate that other CD40L-expressing cell types are more relevant in the pathogenesis of obesity-associated metabolic dysfunction.
Collapse
Affiliation(s)
- Myrthe E Reiche
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Myrthe den Toom
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Lisa Willemsen
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Bram van Os
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Marion J J Gijbels
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
- Pathology, CARIM, Maastricht, The Netherlands
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Suzanne A B M Aarts
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
| | - Esther Lutgens
- Medical Biochemistry, Amsterdam UMC—Location AMC, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Munich, Germany
| |
Collapse
|
733
|
Hu JX, Helleberg M, Jensen AB, Brunak S, Lundgren J. A Large-Cohort, Longitudinal Study Determines Precancer Disease Routes across Different Cancer Types. Cancer Res 2018; 79:864-872. [PMID: 30591553 DOI: 10.1158/0008-5472.can-18-1677] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/29/2018] [Accepted: 12/18/2018] [Indexed: 11/16/2022]
Abstract
Although many diseases are associated with cancer, the full spectrum of temporal disease correlations across cancer types has not yet been characterized. A population-wide study of longitudinal disease trajectories is needed to interrogate the general medical histories of patients with cancer. Here we performed a retrospective study covering a 20-year period, using 6.9 million patients from the Danish National Patient Registry linked to 0.7 million patients with cancer from the Danish Cancer Registry. Statistical analysis identified all significant disease associations occurring prior to cancer diagnoses. These associations were used to build frequently occurring, longitudinal disease trajectories. Across 17 cancer types, a total of 648 significant diagnoses correlated directly with a cancer, while 168 diagnosis trajectories of time-ordered steps were identified for seven cancer types. The most common diseases across cancer types involved cardiovascular, obesity, and genitourinary diseases. A comprehensive, publicly available web tool of interactive illustrations for all cancer disease associations is provided. By exploring the precancer landscape using this large dataset, we identify disease associations that can be used to derive mechanistic hypotheses for future cancer research. SIGNIFICANCE: This study offers an innovative approach to examine prediagnostic disease and cancer development in a large national population-based setting and provides a publicly available tool to foster additional cancer surveillance research.
Collapse
Affiliation(s)
- Jessica X Hu
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Helleberg
- Department of Infectious Diseases (PERSIMUNE @ CHIP), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anders B Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark. .,Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jens Lundgren
- Department of Infectious Diseases (PERSIMUNE @ CHIP), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
734
|
The biochemical basis of disease. Essays Biochem 2018; 62:619-642. [PMID: 30509933 PMCID: PMC6279435 DOI: 10.1042/ebc20170054] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 12/19/2022]
Abstract
This article gives the reader an insight into the role of biochemistry in some of the current global health and disease problems. It surveys the biochemical causes of disease in an accessible and succinct form while also bringing in aspects of pharmacology, cell biology, pathology and physiology which are closely aligned with biochemistry. The discussion of the selected diseases highlights exciting new developments and illuminates key biochemical pathways and commonalities. The article includes coverage of diabetes, atherosclerosis, cancer, microorganisms and disease, nutrition, liver disease and Alzheimer’s disease, but does not attempt to be comprehensive in its coverage of disease, since this is beyond its remit and scope. Consequently there are many fascinating biochemical aspects of diseases, both common and rare, that are not addressed here that can be explored in the further reading cited. Techniques and biochemical procedures for studying disease are not covered in detail here, but these can be found readily in a range of biochemical methods sources.
Collapse
|
735
|
circARF3 Alleviates Mitophagy-Mediated Inflammation by Targeting miR-103/TRAF3 in Mouse Adipose Tissue. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 14:192-203. [PMID: 30623853 PMCID: PMC6325073 DOI: 10.1016/j.omtn.2018.11.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 11/20/2018] [Accepted: 11/20/2018] [Indexed: 12/29/2022]
Abstract
Adipose inflammation is an important cause for obesity-associated metabolic disorders, including insulin resistance and hypertension. Here we investigated that a circular RNA (circRNA), which we termed circARF3 (ADP-ribosylation factor 3), acts as an endogenous miR-103 sponge to alleviate adipose inflammation by promoting mitophagy. On the other hand, miR-103 aggravated inflammation by inhibiting mitophagy, revealing that miR-103 acts as a positive regulator of adipose inflammation. Furthermore, we found that tumor necrosis factor receptor-associated factor 3 (TRAF3), as a miR-103 downstream target, mediates the functions of miR-103 in adipose inflammation. Overexpressing TRAF3 attenuated miR-103-induced inflammation by accelerating mitophagy. Moreover, we identified that circARF3 blocked miR-103 effects, which resulted in an increase in TRAF3 expression. TRAF3 restrained the nuclear factor κB (NF-κB)-signaling pathway, heightened mitophagy, and suppressed NLRP3 inflammasome activation ultimately. Our data showed that circARF3 acts as an endogenous miR-103 sponge to inhibit mitophagy-mediated adipose inflammation both in vitro and in vivo. These findings disclose a new regulatory pathway for adipose inflammation, which consists of circARF3, miR-103, and TRAF3. This study can be a useful addition to our knowledge, as it provides a new strategy for the prevention of adipose inflammation in obesity disorder.
Collapse
|
736
|
Abstract
Hypoxia-inducible factors (HIFs), a family of transcription factors activated by hypoxia, consist of three α-subunits (HIF1α, HIF2α and HIF3α) and one β-subunit (HIF1β), which serves as a heterodimerization partner of the HIFα subunits. HIFα subunits are stabilized from constitutive degradation by hypoxia largely through lowering the activity of the oxygen-dependent prolyl hydroxylases that hydroxylate HIFα, leading to their proteolysis. HIF1α and HIF2α are expressed in different tissues and regulate target genes involved in angiogenesis, cell proliferation and inflammation, and their expression is associated with different disease states. HIFs have been widely studied because of their involvement in cancer, and HIF2α-specific inhibitors are being investigated in clinical trials for the treatment of kidney cancer. Although cancer has been the major focus of research on HIF, evidence has emerged that this pathway has a major role in the control of metabolism and influences metabolic diseases such as obesity, type 2 diabetes mellitus and non-alcoholic fatty liver disease. Notably increased HIF1α and HIF2α signalling in adipose tissue and small intestine, respectively, promotes metabolic diseases in diet-induced disease models. Inhibition of HIF1α and HIF2α decreases the adverse diet-induced metabolic phenotypes, suggesting that they could be drug targets for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| |
Collapse
|
737
|
Keinänen J, Suvisaari J, Reinikainen J, Kieseppä T, Lindgren M, Mäntylä T, Rikandi E, Sundvall J, Torniainen-Holm M, Mantere O. Low-grade inflammation in first-episode psychosis is determined by increased waist circumference. Psychiatry Res 2018; 270:547-553. [PMID: 30343240 DOI: 10.1016/j.psychres.2018.10.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/26/2018] [Accepted: 10/08/2018] [Indexed: 10/28/2022]
Abstract
Psychosis is associated with low-grade inflammation as measured by high-sensitivity C-reactive protein (hs-CRP), a risk factor for cardiovascular events and mortality in the general population. We investigated the relationship between hs-CRP and anthropometric and metabolic changes in first-episode psychosis (FEP) during the first treatment year. We recruited 95 FEP patients and 62 controls, and measured longitudinal changes in hs-CRP, weight, waist circumference, insulin resistance, and lipids. We used linear mixed models to analyze the longitudinal relationship between hs-CRP and clinical, anthropometric and metabolic measures. At baseline, patients with FEP had higher levels of insulin resistance, total and low-density lipoprotein cholesterol, apolipoprotein B, and triglycerides. Baseline weight, waist circumference, hs-CRP, fasting glucose, and high-density lipoprotein cholesterol were similar between patients and controls. Marked increases in anthropometric measures and hs-CRP were observed in FEP during the 12-month follow-up. However, glucose and lipid parameters did not change significantly. In the mixed models, waist circumference and female sex were significant predictors of hs-CRP levels in FEP. Prevention of the early development of abdominal obesity in FEP is crucial, as abdominal obesity is accompanied by chronic low-grade inflammation, which increases further the cardiovascular risk in this vulnerable population.
Collapse
Affiliation(s)
- Jaakko Keinänen
- Department of Public Health Solutions, Mental Health Unit, National Institute for Health and Welfare, P.O. Box 30, FIN-00271 Helsinki, Finland; Faculty of Medicine, Department of Psychiatry, University of Helsinki, P.O. Box 590, FIN-00029 Helsinki, Finland.
| | - Jaana Suvisaari
- Department of Public Health Solutions, Mental Health Unit, National Institute for Health and Welfare, P.O. Box 30, FIN-00271 Helsinki, Finland
| | - Jaakko Reinikainen
- Department of Public Health Solutions, Public Health Evaluation and Projection Unit, National Institute for Health and Welfare, P.O. Box 30, FIN-00271 Helsinki, Finland
| | - Tuula Kieseppä
- Psychiatry, University of Helsinki and Helsinki University Hospital, P.O. Box 590, FIN-00029 Helsinki, Finland
| | - Maija Lindgren
- Department of Public Health Solutions, Mental Health Unit, National Institute for Health and Welfare, P.O. Box 30, FIN-00271 Helsinki, Finland
| | - Teemu Mäntylä
- Department of Public Health Solutions, Mental Health Unit, National Institute for Health and Welfare, P.O. Box 30, FIN-00271 Helsinki, Finland; Faculty of Medicine, Department of Psychology and Logopedics, University of Helsinki, P.O. Box 63, FIN-00014 Helsinki, Finland
| | - Eva Rikandi
- Department of Public Health Solutions, Mental Health Unit, National Institute for Health and Welfare, P.O. Box 30, FIN-00271 Helsinki, Finland; Faculty of Medicine, Department of Psychology and Logopedics, University of Helsinki, P.O. Box 63, FIN-00014 Helsinki, Finland
| | - Jouko Sundvall
- Department of Public Health Solutions, Genomics and Biomarkers Unit, National Institute for Health and Welfare, P.O. Box 30, FIN-00271 Helsinki, Finland
| | - Minna Torniainen-Holm
- Department of Public Health Solutions, Mental Health Unit, National Institute for Health and Welfare, P.O. Box 30, FIN-00271 Helsinki, Finland
| | - Outi Mantere
- Psychiatry, University of Helsinki and Helsinki University Hospital, P.O. Box 590, FIN-00029 Helsinki, Finland; Department of Psychiatry, McGill University, 1033 Pine Avenue West, QC, H3A 1A1 Montréal, Canada; Bipolar Disorders Clinic, Douglas Mental Health University Institute, 6875 LaSalle Boulevard, QC, H4H 1R3 Montréal, Canada
| |
Collapse
|
738
|
Nox2 Activity Is Required in Obesity-Mediated Alteration of Bone Remodeling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6054361. [PMID: 30533174 PMCID: PMC6250007 DOI: 10.1155/2018/6054361] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023]
Abstract
Despite increasing evidence suggesting a role for NADPH oxidases (Nox) in bone pathophysiology, whether Nox enzymes contribute to obesity-mediated bone remodeling remains to be clearly elucidated. Nox2 is one of the predominant Nox enzymes expressed in the bone marrow microenvironment and is a major source of ROS generation during inflammatory processes. It is also well recognized that a high-fat diet (HFD) induces obesity, which negatively impacts bone remodeling. In this work, we investigated the effect of Nox2 loss of function on obesity-mediated alteration of bone remodeling using wild-type (WT) and Nox2-knockout (KO) mice fed with a standard lab chow diet (SD) as a control or a HFD as an obesity model. Bone mineral density (BMD) of mice was assessed at the beginning and after 3 months of feeding with SD or HFD. Our results show that HFD increased bone mineral density to a greater extent in KO mice than in WT mice without affecting the total body weight and fat mass. HFD also significantly increased the number of adipocytes in the bone marrow microenvironment of WT mice as compared to KO mice. The bone levels of proinflammatory cytokines and proosteoclastogenic factors were also significantly elevated in WT-HFD mice as compared to KO-HFD mice. Furthermore, the in vitro differentiation of bone marrow cells into osteoclasts was significantly increased when using bone marrow cells from WT-HFD mice as compared to KO-HFD mice. Our data collectively suggest that Nox2 is implicated in HFD-induced deleterious bone remodeling by enhancing bone marrow adipogenesis and osteoclastogenesis.
Collapse
|
739
|
Yuan X, Zhang P, Wang Y, Liu Y, Li X, Kumar BU, Hei G, Lv L, Huang XF, Fan X, Song X. Changes in metabolism and microbiota after 24-week risperidone treatment in drug naïve, normal weight patients with first episode schizophrenia. Schizophr Res 2018; 201:299-306. [PMID: 29859859 DOI: 10.1016/j.schres.2018.05.017] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/01/2018] [Accepted: 05/12/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE This study was to examine the alterations in metabolic parameters, anti-oxidant superoxide dismutase (SOD), inflammatory marker high-sensitivity C-reactive protein (hs-CRP) and microbiota after 24-week risperidone treatment in drug naïve, normal weight, first episode schizophrenia patients; the study further examined the relationship between metabolic changes and changes in microbiota. METHODS Forty-one patients completed the 24-week study and 41 controls were enrolled in this study. Metabolic parameters, SOD, hs-CRP and the copy numbers of 5 fecal bacteria were measured at baseline (both groups) and at different time points (patients only). RESULTS Patients had significantly lower numbers of fecal Bifidobacterium spp., Escherichia coli, Lactobacillus spp. compared with healthy controls (HC) (ps < 0.001); in contrast, the numbers of fecal Clostridium coccoides group were significantly higher in the patient group compared with HC (p < 0.001). After 24-week risperidone treatment, there were significant increases in body weight, BMI, fasting blood-glucose, triglycerides, LDL, hs-CRP, SOD and HOMA-IR (p < 0.001), significant increases in the numbers of fecal Bifidobacterium spp. and E. coli (ps < 0.001), and significant decreases in the numbers of fecal Clostridium coccoides group and Lactobacillus spp. (ps < 0.001). Hierarchical multiple linear regression analysis shows that after controlling for potential confounding variables, only the changes in fecal Bifidobacterium spp., among 4 types of fecal bacteria, entered into the model and significantly correlated with the changes in weight (unstandardized coefficient B = 4.413, R2 change = 0.167, p = 0.009) and BMI (B = 1.639, R2 change = 0.172, p = 0.008) after 24-week treatment. CONCLUSION Drug naïve, first episode schizophrenia patients show abnormalities in microbiota composition. Risperidone treatment causes significant changes in certain fecal bacteria, which are likely associated with antipsychotic medication induced metabolic changes.
Collapse
Affiliation(s)
- Xiuxia Yuan
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Peifen Zhang
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Yaping Wang
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Yafei Liu
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Xue Li
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Bachoo Upshant Kumar
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Gangrui Hei
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Luxian Lv
- Henan Province Mental Hospital, The Second Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW 2522, Australia
| | - Xiaoduo Fan
- Psychotic Disorders Program, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, United States.
| | - Xueqin Song
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China; Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China; Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
740
|
Khalyfa A, Kheirandish-Gozal L, Gozal D. Exosome and Macrophage Crosstalk in Sleep-Disordered Breathing-Induced Metabolic Dysfunction. Int J Mol Sci 2018; 19:ijms19113383. [PMID: 30380647 PMCID: PMC6274857 DOI: 10.3390/ijms19113383] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/22/2018] [Accepted: 10/25/2018] [Indexed: 12/12/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a highly prevalent worldwide public health problem that is characterized by repetitive upper airway collapse leading to intermittent hypoxia, pronounced negative intrathoracic pressures, and recurrent arousals resulting in sleep fragmentation. Obesity is a major risk factor of OSA and both of these two closely intertwined conditions result in increased sympathetic activity, oxidative stress, and chronic low-grade inflammation, which ultimately contribute, among other morbidities, to metabolic dysfunction, as reflected by visceral white adipose tissue (VWAT) insulin resistance (IR). Circulating extracellular vesicles (EVs), including exosomes, are released by most cell types and their cargos vary greatly and reflect underlying changes in cellular homeostasis. Thus, exosomes can provide insights into how cells and systems cope with physiological perturbations by virtue of the identity and abundance of miRNAs, mRNAs, proteins, and lipids that are packaged in the EVs cargo, and are secreted from the cells into bodily fluids under normal as well as diseased states. Accordingly, exosomes represent a novel pathway via which a cohort of biomolecules can travel long distances and result in the modulation of gene expression in selected and targeted recipient cells. For example, exosomes secreted from macrophages play a critical role in innate immunity and also initiate the adaptive immune response within specific metabolic tissues such as VWAT. Under normal conditions, phagocyte-derived exosomes represent a large portion of circulating EVs in blood, and carry a protective signature against IR that is altered when secreting cells are exposed to altered physiological conditions such as those elicited by OSA, leading to emergence of IR within VWAT compartment. Consequently, increased understanding of exosome biogenesis and biology should lead to development of new diagnostic biomarker assays and personalized therapeutic approaches. Here, the evidence on the major biological functions of macrophages and exosomes as pathophysiological effectors of OSA-induced metabolic dysfunction is discussed.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Sections of Pediatric Sleep Medicine and Pediatric Pulmonology, Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA.
| | - Leila Kheirandish-Gozal
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA.
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA.
| |
Collapse
|
741
|
Ottino-González J, Jurado MA, García-García I, Segura B, Marqués-Iturria I, Sender-Palacios MJ, Tor E, Prats-Soteras X, Caldú X, Junqué C, Pasternak O, Garolera M. Allostatic load and disordered white matter microstructure in overweight adults. Sci Rep 2018; 8:15898. [PMID: 30367110 PMCID: PMC6203765 DOI: 10.1038/s41598-018-34219-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 10/12/2018] [Indexed: 12/29/2022] Open
Abstract
Overweight and stress are both related to brain structural abnormalities. The allostatic load model states that frequent disruption of homeostasis is inherently linked to oxidative stress and inflammatory responses that in turn can damage the brain. However, the effects of the allostatic load on the central nervous system remain largely unknown. The current study aimed to assess the relationship between the allostatic load and the composition of whole-brain white matter tracts in overweight subjects. Additionally, we have also tested for grey matter changes regarding allostatic load increase. Thirty-one overweight-to-obese adults and 21 lean controls participated in the study. Our results showed that overweight participants presented higher allostatic load indexes. Such increases correlated with lower fractional anisotropy in the inferior fronto-occipital fasciculi and the right anterior corona radiata, as well as with grey matter reductions in the left precentral gyrus, the left lateral occipital gyrus, and the right pars opercularis. These results suggest that an otherwise healthy overweight status is linked to long-term biological changes potentially harmful to the brain.
Collapse
Affiliation(s)
- J Ottino-González
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - M A Jurado
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain.
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain.
| | - I García-García
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - B Segura
- Departament de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - I Marqués-Iturria
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain
| | - M J Sender-Palacios
- CAP Terrassa Nord, Consorci Sanitari de Terrassa, Barcelona, Spain
- Brain, Cognition and Behavior Clinical Research Group, Consorci Sanitari de Terrassa, Barcelona, Spain
| | - E Tor
- CAP Terrassa Nord, Consorci Sanitari de Terrassa, Barcelona, Spain
- Brain, Cognition and Behavior Clinical Research Group, Consorci Sanitari de Terrassa, Barcelona, Spain
| | - X Prats-Soteras
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - X Caldú
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - C Junqué
- Departament de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - O Pasternak
- Departments of Psychiatry and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - M Garolera
- Unitat de Neuropsicologia, Hospital de Terrassa, Consorci Sanitari de Terrassa, Barcelona, Spain
- Brain, Cognition and Behavior Clinical Research Group, Consorci Sanitari de Terrassa, Barcelona, Spain
| |
Collapse
|
742
|
Gil-Iturbe E, Castilla-Madrigal R, Barrenetxe J, Villaro AC, Lostao MP. GLUT12 expression and regulation in murine small intestine and human Caco-2 cells. J Cell Physiol 2018; 234:4396-4408. [PMID: 30352123 DOI: 10.1002/jcp.27231] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/17/2018] [Indexed: 12/17/2022]
Abstract
GLUT12 was cloned from the mammary cancer cell line MCF-7, but its physiological role still needs to be elucidated. To gain more knowledge of GLUT12 function in the intestine, we investigated GLUT12 subcellular localization in the small intestine and its regulation by sugars, hormones, and intracellular mediators in Caco-2 cells and mice. Immunohistochemical methods were used to determine GLUT12 subcellular localization in human and murine small intestine. Brush border membrane vesicles were isolated for western blot analyses. Functional studies were performed in Caco-2 cells by measuring α-methyl-d-glucose (αMG) uptake in the absence of sodium. GLUT12 is located in the apical cytoplasm, below the brush border membrane, and in the perinuclear region of murine and human enterocytes. In Caco-2 cells, GLUT12 translocation to the apical membrane and α-methyl- d-glucose uptake by the transporter are stimulated by protons, glucose, insulin, tumor necrosis factor-α (TNF-α), protein kinase C, and AMP-activated protein kinase. In contrast, hypoxia decreases GLUT12 expression in the apical membrane. Upregulation of TNF-α and hypoxia-inducible factor-1α ( HIF-1α) genes is found in the jejunal mucosa of diet-induced obese mice. In these animals, GLUT12 expression in the brush border membrane is slightly decreased compared with lean animals. Moreover, an intraperitoneal injection of insulin does not induce GLUT12 translocation to the membrane, as it occurs in lean animals. GLUT12 rapid translocation to the enterocytes' apical membrane in response to glucose and insulin could be related to GLUT12 participation in sugar absorption during postprandial periods. In obesity, in which insulin sensitivity is reduced, the contribution of GLUT12 to sugar absorption is affected.
Collapse
Affiliation(s)
- Eva Gil-Iturbe
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Nutrition Research Centre, University of Navarra, Pamplona, Spain
| | - Rosa Castilla-Madrigal
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Nutrition Research Centre, University of Navarra, Pamplona, Spain
| | - Jaione Barrenetxe
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain
| | - Ana Cristina Villaro
- Department of Histology and Pathological Anatomy, University of Navarra, Pamplona, Spain
| | - María Pilar Lostao
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Nutrition Research Centre, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| |
Collapse
|
743
|
Komakula SSB, Tumova J, Kumaraswamy D, Burchat N, Vartanian V, Ye H, Dobrzyn A, Lloyd RS, Sampath H. The DNA Repair Protein OGG1 Protects Against Obesity by Altering Mitochondrial Energetics in White Adipose Tissue. Sci Rep 2018; 8:14886. [PMID: 30291284 PMCID: PMC6173743 DOI: 10.1038/s41598-018-33151-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 09/21/2018] [Indexed: 12/15/2022] Open
Abstract
Obesity and related metabolic pathologies represent a significant public health concern. Obesity is associated with increased oxidative stress that damages genomic and mitochondrial DNA. Oxidatively-induced lesions in both DNA pools are repaired via the base-excision repair pathway, initiated by DNA glycosylases such as 8-oxoguanine DNA glycosylase (OGG1). Global deletion of OGG1 and common OGG1 polymorphisms render mice and humans susceptible to metabolic disease. However, the relative contribution of mitochondrial OGG1 to this metabolic phenotype is unknown. Here, we demonstrate that transgenic targeting of OGG1 to mitochondria confers significant protection from diet-induced obesity, insulin resistance, and adipose tissue inflammation. These favorable metabolic phenotypes are mediated by an increase in whole body energy expenditure driven by specific metabolic adaptations, including increased mitochondrial respiration in white adipose tissue of OGG1 transgenic (Ogg1Tg) animals. These data demonstrate a critical role for a DNA repair protein in modulating mitochondrial energetics and whole-body energy balance.
Collapse
Affiliation(s)
- Sai Santosh Babu Komakula
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, 08901, USA.,Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Jana Tumova
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Deeptha Kumaraswamy
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Natalie Burchat
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Vladimir Vartanian
- Oregon Institute of Occupational Health Sciences, Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Hong Ye
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Agnieszka Dobrzyn
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - R Stephen Lloyd
- Oregon Institute of Occupational Health Sciences, Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Harini Sampath
- Department of Nutritional Sciences and Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
744
|
Abstract
At the simplest level, obesity is the manifestation of an imbalance between caloric intake and expenditure; however, the pathophysiological mechanisms that govern the development of obesity and associated complications are enormously complex. Fibrosis within the adipose tissue compartment is one such factor that may influence the development of obesity and/or obesity-related comorbidities. Furthermore, the functional consequences of adipose tissue fibrosis are a matter of considerable debate, with evidence that fibrosis serves both adaptive and maladaptive roles. Tissue fibrosis itself is incompletely understood, and multiple cellular and molecular pathways are involved in the development, maintenance, and resolution of the fibrotic state. Within the context of obesity, fibrosis influences molecular and cellular events that relate to adipocytes, inflammatory cells, inflammatory mediators, and supporting adipose stromal tissue. In this Review, we explore what is known about the interplay between the development of adipose tissue fibrosis and obesity, with a view toward future investigative and therapeutic avenues.
Collapse
Affiliation(s)
| | - Michael J Podolsky
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Kamran Atabai
- Cardiovascular Research Institute.,Lung Biology Center, and.,Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
745
|
Pardo R, Vilà M, Cervela L, de Marco M, Gama-Pérez P, González-Franquesa A, Statuto L, Vilallonga R, Simó R, Garcia-Roves PM, Villena JA. Calorie restriction prevents diet-induced insulin resistance independently of PGC-1-driven mitochondrial biogenesis in white adipose tissue. FASEB J 2018; 33:2343-2358. [PMID: 30277821 DOI: 10.1096/fj.201800310r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Calorie restriction (CR) exerts remarkable, beneficial effects on glucose homeostasis by mechanisms that are not fully understood. Given the relevance of white adipose tissue (WAT) in glucose homeostasis, we aimed at identifying the main cellular processes regulated in WAT in response to CR in a pathologic context of obesity. For this, a gene-expression profiling study was first conducted in mice fed ad libitum or subjected to 40% CR. We found that the gene network related to mitochondria was the most highly upregulated in WAT by CR. To study the role that increased mitochondrial biogenesis plays on glucose homeostasis following CR, we generated a mouse model devoid of the coactivators peroxisome proliferator-activated receptor γ coactivator 1 (PGC-1)α and PGC-1β specifically in adipocytes. Our results show that mice lacking PGC-1s in adipocytes are unable to increase mitochondrial biogenesis in WAT upon CR. Despite a blunted induction of mitochondrial biogenesis in response to calorie deprivation, mice lacking adipose PGC-1s still respond to CR by improving their glucose homeostasis. Our study demonstrates that PGC-1 coactivators are major regulators of CR-induced mitochondrial biogenesis in WAT and that increased mitochondrial biogenesis and oxidative function in adipose tissue are not required for the improvement of glucose homeostasis mediated by CR.-Pardo, R., Vilà, M., Cervela, L., de Marco, M., Gama-Pérez, P., González-Franquesa, A., Statuto, L., Vilallonga, R., Simó, R., Garcia-Roves, P. M., Villena, J. A. Calorie restriction prevents diet-induced insulin resistance independently of PGC-1-driven mitochondrial biogenesis in white adipose tissue.
Collapse
Affiliation(s)
- Rosario Pardo
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Vilà
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luis Cervela
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marina de Marco
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pau Gama-Pérez
- Department of Physiological Sciences, Institut d'Investigació Biomèdica de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alba González-Franquesa
- Department of Physiological Sciences, Institut d'Investigació Biomèdica de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lucia Statuto
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ramon Vilallonga
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain.,Metabolic and Bariatric Surgery Unit, European Accreditation Council for Bariatric Surgery Center of Excellence, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Rafael Simó
- Unit of Diabetes and Metabolism, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; and.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain
| | - Pablo M Garcia-Roves
- Department of Physiological Sciences, Institut d'Investigació Biomèdica de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Josep A Villena
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
746
|
Igarashi Y, Nawaz A, Kado T, Bilal M, Kuwano T, Yamamoto S, Sasahara M, Jiuxiang X, Inujima A, Koizumi K, Imura J, Shibahara N, Usui I, Fujisaka S, Tobe K. Partial depletion of CD206-positive M2-like macrophages induces proliferation of beige progenitors and enhances browning after cold stimulation. Sci Rep 2018; 8:14567. [PMID: 30275453 PMCID: PMC6167387 DOI: 10.1038/s41598-018-32803-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/14/2018] [Indexed: 12/30/2022] Open
Abstract
Beige adipocytes are an inducible form of thermogenic adipocytes that become interspersed within white adipose tissue (WAT) depots in response to cold exposure. Previous studies have shown that type 2 cytokines and M2 macrophages induce cold-induced browning in inguinal WAT (ingWAT) by producing catecholamines. Exactly how the conditional and partial depletion of CD206+ M2-like macrophages regulates the cold-induced browning of ingWAT, however, remains unknown. We examined the role of CD206+ M2-like macrophages in the cold-induced browning of WAT using genetically engineered CD206DTR mice, in which CD206+ M2-like macrophages were conditionally depleted. The partial depletion of CD206+ M2-like enhanced UCP1 expression in ingWAT, as shown by immunostaining, and also upregulated the expression of Ucp1 and other browning-related marker genes in ingWAT after cold exposure. A flow cytometry analysis showed that the partial depletion of CD206+ M2-like macrophages caused an increase in the number of beige progenitors in ingWAT in response to cold. Thus, we concluded that CD206+ M2-like macrophages inhibit the proliferation of beige progenitors and that the partial depletion of CD206+ M2-like macrophages releases this inhibition, thereby enhancing browning and insulin sensitivity.
Collapse
Affiliation(s)
- Yoshiko Igarashi
- First Department of Internal Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Allah Nawaz
- First Department of Internal Medicine, University of Toyama, Toyama, 930-0194, Japan.
- Department of Metabolism and Nutrition, University of Toyama, Toyama, 930-0194, Japan.
- JSPS International Research Fellow, Department of Metabolism and Nutrition, University of Toyama, Toyama, 930-0194, Japan.
| | - Tomonobu Kado
- First Department of Internal Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Muhammad Bilal
- First Department of Internal Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Takahide Kuwano
- First Department of Internal Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Seiji Yamamoto
- Department of Pathology, University of Toyama, Toyama, 930-0194, Japan
| | - Masakiyo Sasahara
- Department of Pathology, University of Toyama, Toyama, 930-0194, Japan
| | - Xu Jiuxiang
- Division of Kampo Diagnostics, Institute of Natural Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Akiko Inujima
- Division of Kampo Diagnostics, Institute of Natural Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Keiichi Koizumi
- Division of Kampo Diagnostics, Institute of Natural Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Johji Imura
- Department of Diagnostic Pathology, University of Toyama, Toyama, 930-0194, Japan
| | - Naotoshi Shibahara
- Division of Kampo Diagnostics, Institute of Natural Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Isao Usui
- First Department of Internal Medicine, University of Toyama, Toyama, 930-0194, Japan
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi, 321-0293, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
747
|
Renner S, Blutke A, Dobenecker B, Dhom G, Müller TD, Finan B, Clemmensen C, Bernau M, Novak I, Rathkolb B, Senf S, Zöls S, Roth M, Götz A, Hofmann SM, Hrabĕ de Angelis M, Wanke R, Kienzle E, Scholz AM, DiMarchi R, Ritzmann M, Tschöp MH, Wolf E. Metabolic syndrome and extensive adipose tissue inflammation in morbidly obese Göttingen minipigs. Mol Metab 2018; 16:180-190. [PMID: 30017782 PMCID: PMC6157610 DOI: 10.1016/j.molmet.2018.06.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/16/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The worldwide prevalence of obesity has increased to 10% in men and 15% in women and is associated with severe comorbidities such as diabetes, cancer, and cardiovascular disease. Animal models of obesity are central to experimental studies of disease mechanisms and therapeutic strategies. Diet-induced obesity (DIO) models in rodents have provided important insights into the pathophysiology of obesity and, in most instances, are the first in line for exploratory pharmacology studies. To deepen the relevance towards translation to human patients, we established a corresponding DIO model in Göttingen minipigs (GM). METHODS Young adult female ovariectomized GM were fed a high-fat/high-energy diet for a period of 70 weeks. The ration was calculated to meet the requirements and maintain body weight (BW) of lean adult minipigs (L-GM group) or increased stepwise to achieve an obese state (DIO-GM group). Body composition, blood parameters and intravenous glucose tolerance were determined at regular intervals. A pilot chronic treatment trial with a GLP1 receptor agonist was conducted in DIO-GM. At the end of the study, the animals were necropsied and a biobank of selected tissues was established. RESULTS DIO-GM developed severe subcutaneous and visceral adiposity (body fat >50% of body mass vs. 22% in L-GM), increased plasma cholesterol, triglyceride, and free fatty acid levels, insulin resistance (HOMA-IR >5 vs. 2 in L-GM), impaired glucose tolerance and increased heart rate when resting and active. However, fasting glucose concentrations stayed within normal range throughout the study. Treatment with a long-acting GLP1 receptor agonist revealed substantial reduction of food intake and body weight within four weeks, with increased drug sensitivity relative to observations in other DIO animal models. Extensive adipose tissue inflammation and adipocyte necrosis was observed in visceral, but not subcutaneous, adipose tissue of DIO-GM. CONCLUSIONS The Munich DIO-GM model resembles hallmarks of the human metabolic syndrome with extensive adipose tissue inflammation and adipocyte necrosis reported for the first time. DIO-GM may be used for evaluating novel treatments of obesity and associated comorbidities. They may help to identify triggers and mechanisms of fat tissue inflammation and mechanisms preventing complete metabolic decompensation despite morbid obesity.
Collapse
Affiliation(s)
- Simone Renner
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany; Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Hackerstr. 27, 85764, Oberschleißheim, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.
| | - Andreas Blutke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Veterinärstr. 13, 80539, Munich, Germany
| | - Britta Dobenecker
- Chair of Animal Nutrition and Dietetics, Department of Veterinary Sciences, LMU Munich, Schönleutnerstr. 8, 85764, Oberschleißheim, Germany
| | - Georg Dhom
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany; Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Hackerstr. 27, 85764, Oberschleißheim, Germany
| | - Timo D Müller
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Ismaninger Str. 22, 81675, Munich, Germany
| | - Brian Finan
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Ismaninger Str. 22, 81675, Munich, Germany
| | - Christoffer Clemmensen
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Ismaninger Str. 22, 81675, Munich, Germany
| | - Maren Bernau
- Livestock Center of the Veterinary Faculty, LMU Munich, St.-Hubertus-Str. 12, 85764, Oberschleißheim, Germany
| | - Istvan Novak
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany; Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Hackerstr. 27, 85764, Oberschleißheim, Germany
| | - Birgit Rathkolb
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany; Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Hackerstr. 27, 85764, Oberschleißheim, Germany; German Mouse Clinic (GMC), Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Steffanie Senf
- Clinic for Swine, Center for Clinical Veterinary Medicine, LMU Munich, Sonnenstr. 16, 85764, Oberschleißheim, Germany
| | - Susanne Zöls
- Clinic for Swine, Center for Clinical Veterinary Medicine, LMU Munich, Sonnenstr. 16, 85764, Oberschleißheim, Germany
| | - Mirjam Roth
- Animal aspects, 88400, Biberach an der Riss, Germany
| | - Anna Götz
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Department of Internal Medicine I, University Hospital RWTH Aachen, Pauwelstr. 30, 52074, Aachen, Germany
| | - Susanna M Hofmann
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Institute of Diabetes and Regeneration Research (IDR), Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der LMU, Ziemssenstr, 180336, Munich, Germany
| | - Martin Hrabĕ de Angelis
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; German Mouse Clinic (GMC), Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Genome Analysis Center (GAC), Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health and Chair of Experimental Genetics, Technische Universität, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Veterinärstr. 13, 80539, Munich, Germany
| | - Ellen Kienzle
- Chair of Animal Nutrition and Dietetics, Department of Veterinary Sciences, LMU Munich, Schönleutnerstr. 8, 85764, Oberschleißheim, Germany
| | - Armin M Scholz
- Livestock Center of the Veterinary Faculty, LMU Munich, St.-Hubertus-Str. 12, 85764, Oberschleißheim, Germany
| | - Richard DiMarchi
- Novo Nordisk Research Center Indianapolis, 5225 Exploration Drive, Indianapolis, IN, 46241, USA; Department of Chemistry, Indiana University, 800 E. Kirkwood Ave., Bloomington, IN, 47405-7102, USA
| | - Mathias Ritzmann
- Clinic for Swine, Center for Clinical Veterinary Medicine, LMU Munich, Sonnenstr. 16, 85764, Oberschleißheim, Germany
| | - Matthias H Tschöp
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Ismaninger Str. 22, 81675, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany; Center for Innovative Medical Models (CiMM), Department of Veterinary Sciences, LMU Munich, Hackerstr. 27, 85764, Oberschleißheim, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany; Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| |
Collapse
|
748
|
Ritter A, Louwen F, Yuan J. Deficient primary cilia in obese adipose-derived mesenchymal stem cells: obesity, a secondary ciliopathy? Obes Rev 2018; 19:1317-1328. [PMID: 30015415 DOI: 10.1111/obr.12716] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/24/2018] [Accepted: 05/09/2018] [Indexed: 12/14/2022]
Abstract
Obesity alters the composition, structure and function of adipose tissue, characterized by chronic inflammation, insulin resistance and metabolic dysfunction. Adipose-derived mesenchymal stem cells (ASCs) are responsible for cell renewal, spontaneous repair and immunomodulation in adipose tissue. Increasing evidence highlights that ASCs are deficient in obesity, and the underlying mechanisms are not well understood. We have recently shown that obese ASCs have defective primary cilia, which are shortened and unable to properly respond to stimuli. Impaired cilia compromise ASC functions. This work suggests an intertwined connection of obesity, defective cilia and dysfunctional ASCs. We have here discussed the current data regarding defective cilia in various cell types in obesity. Based on these observations, we hypothesize that obesity, a systemic chronic metainflammation, could impair cilia in diverse ciliated cells, like pancreatic islet cells, stem cells and hypothalamic neurons, making these critical cells dysfunctional by shutting down their signal sensors and transducers. In this context, obesity may represent a secondary form of ciliopathy induced by obesity-related inflammation and metabolic dysfunction. Reactivation of ciliated cells might be an alternative strategy to combat obesity and its associated diseases.
Collapse
Affiliation(s)
- A Ritter
- Department of Gynecology and Obstetrics, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - F Louwen
- Department of Gynecology and Obstetrics, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - J Yuan
- Department of Gynecology and Obstetrics, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
749
|
Hu L, He F, Huang M, Peng M, Zhou Z, Liu F, Dai YS. NFATc3 deficiency reduces the classical activation of adipose tissue macrophages. J Mol Endocrinol 2018; 61:79-89. [PMID: 30307161 DOI: 10.1530/jme-18-0070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nuclear factors of activated T cells (NFAT) c3 have a prominent role in the regulation of proinflammatory factors in immune cells. The classically activated M1 macrophages are key players in the initiation and maintenance of adipose tissue (AT) inflammation. The role of NFATc3 in obesity and AT inflammation is unknown. We set out to determine how deficiency of NFATc3 effected macrophage polarization, inflammation and insulin resistance in visceral AT of high-fat diet (HFD)-fed mice. Nfatc3−/− and WT mice were fed a HFD for 8–17 weeks. Epididymal white AT (eWAT) F4/80(+) cells were characterized by fluorescence-activated cell sorting and quantitative RT-PCR. Results showed that Nfatc3−/− mice developed HFD-induced obesity similar to WT mice, but insulin sensitivity and glucose tolerance were improved, and liver fat accumulation was reduced in Nfatc3−/− mice compared to WT control mice. Moreover, M1 macrophage content and proinflammatory factors were reduced, whereas the alternatively activated M2 macrophage content was increased in eWAT of HFD-fed Nfatc3−/− mice compared to that of WT mice. In addition, eWAT insulin signaling was improved in HFD-fed Nfatc3−/− mice. Importantly, after bone-marrow-derived macrophages had been isolated from Nfatc3−/− mice and cultured in vitro, treatment of these cells with interferon-γ and lipopolysaccharide resulted in reduction of M1 inflammatory markers, suggesting that NFATc3 promoted M1 polarization by a cell-autonomous mechanism. The results demonstrated that NFATc3 played an important role in M1 macrophage polarization, AT inflammation and insulin resistance in response to obesity through transcriptional activation of proinflammatory genes.
Collapse
Affiliation(s)
- Li Hu
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fengli He
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Melfeng Huang
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Melhua Peng
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Liu
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Yan-Shan Dai
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
750
|
Graja A, Garcia-Carrizo F, Jank AM, Gohlke S, Ambrosi TH, Jonas W, Ussar S, Kern M, Schürmann A, Aleksandrova K, Blüher M, Schulz TJ. Loss of periostin occurs in aging adipose tissue of mice and its genetic ablation impairs adipose tissue lipid metabolism. Aging Cell 2018; 17:e12810. [PMID: 30088333 PMCID: PMC6156450 DOI: 10.1111/acel.12810] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 06/01/2018] [Accepted: 06/10/2018] [Indexed: 12/22/2022] Open
Abstract
Remodeling of the extracellular matrix is a key component of the metabolic adaptations of adipose tissue in response to dietary and physiological challenges. Disruption of its integrity is a well-known aspect of adipose tissue dysfunction, for instance, during aging and obesity. Adipocyte regeneration from a tissue-resident pool of mesenchymal stem cells is part of normal tissue homeostasis. Among the pathophysiological consequences of adipogenic stem cell aging, characteristic changes in the secretory phenotype, which includes matrix-modifying proteins, have been described. Here, we show that the expression of the matricellular protein periostin, a component of the extracellular matrix produced and secreted by adipose tissue-resident interstitial cells, is markedly decreased in aged brown and white adipose tissue depots. Using a mouse model, we demonstrate that the adaptation of adipose tissue to adrenergic stimulation and high-fat diet feeding is impaired in animals with systemic ablation of the gene encoding for periostin. Our data suggest that loss of periostin attenuates lipid metabolism in adipose tissue, thus recapitulating one aspect of age-related metabolic dysfunction. In human white adipose tissue, periostin expression showed an unexpected positive correlation with age of study participants. This correlation, however, was no longer evident after adjusting for BMI or plasma lipid and liver function biomarkers. These findings taken together suggest that age-related alterations of the adipose tissue extracellular matrix may contribute to the development of metabolic disease by negatively affecting nutrient homeostasis.
Collapse
Affiliation(s)
- Antonia Graja
- Department of Adipocyte Development and Nutrition; German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
- University of Potsdam, Institute of Nutritional Science; Potsdam-Rehbrücke Germany
| | - Francisco Garcia-Carrizo
- Department of Adipocyte Development and Nutrition; German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
| | - Anne-Marie Jank
- Department of Adipocyte Development and Nutrition; German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
| | - Sabrina Gohlke
- Department of Adipocyte Development and Nutrition; German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
| | - Thomas H. Ambrosi
- Department of Adipocyte Development and Nutrition; German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
| | - Wenke Jonas
- Department of Experimental Diabetology; German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
- German Center for Diabetes Research (DZD); Munich-Neuherberg Germany
| | - Siegfried Ussar
- German Center for Diabetes Research (DZD); Munich-Neuherberg Germany
- JRG Adipocytes and Metabolism; Institute for Diabetes and Obesity; Helmholtz Center Munich; Garching Germany
| | - Matthias Kern
- Department of Medicine; University of Leipzig; Leipzig Germany
| | - Annette Schürmann
- University of Potsdam, Institute of Nutritional Science; Potsdam-Rehbrücke Germany
- Department of Experimental Diabetology; German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
- German Center for Diabetes Research (DZD); Munich-Neuherberg Germany
| | - Krasimira Aleksandrova
- University of Potsdam, Institute of Nutritional Science; Potsdam-Rehbrücke Germany
- Nutrition, Immunity and Metabolism Senior Scientist Group; German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
| | - Matthias Blüher
- Department of Medicine; University of Leipzig; Leipzig Germany
| | - Tim J. Schulz
- Department of Adipocyte Development and Nutrition; German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
- University of Potsdam, Institute of Nutritional Science; Potsdam-Rehbrücke Germany
- German Center for Diabetes Research (DZD); Munich-Neuherberg Germany
| |
Collapse
|