101
|
Li Q, Zhang P, Yu X, Zhao Y, Li Q, Zhang Y, Yang Z, Xie Y, Xue P, Sun S, Jia X, Zhou Z, He M, Zhang Y. Lead Transiently Promotes Granulocyte-Macrophage Progenitor Differentiation and Subsequently Suppresses Common Myeloid Progenitor Differentiation. Toxicol Sci 2017; 160:268-283. [DOI: 10.1093/toxsci/kfx176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Qian Li
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Peng Zhang
- Huzhou Center for Disease Control and Prevention, Zhejiang 313000, China
| | - Xinchun Yu
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Yifan Zhao
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Qiang Li
- Putuo District Center for Disease Control and Prevention, Shanghai 200062, China
| | - Yandong Zhang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Zhengli Yang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Yunli Xie
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Peng Xue
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Shuhui Sun
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaodong Jia
- Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, China
| | - Zhijun Zhou
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Miao He
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yubin Zhang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| |
Collapse
|
102
|
Liu P, Huang G, Cao Z, Xie Q, Wei T, Huang C, Li Q, Sun M, Shen W, Gao P. Haematopoietic TLR4 deletion attenuates perivascular brown adipose tissue inflammation in atherosclerotic mice. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:946-957. [PMID: 28579235 DOI: 10.1016/j.bbalip.2017.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 05/20/2017] [Accepted: 05/29/2017] [Indexed: 01/07/2023]
Abstract
AIMS To investigate whether haematopoietic TLR4 deletion attenuates perivascular brown adipose tissue inflammation in atherosclerotic mice. METHODS AND RESULTS Experiments were performed using irradiated LDL receptor-deficient (LDLR-/-) mice with marrow from either TLR4-deficient (TLR4-/-) or age-matched wild-type (WT) mice. After 12 weeks of being fed a high-cholesterol diet, TLR4-/-→LDLR-/- mice developed fewer atherosclerotic lesions in the aorta compared to WT→LDLR-/- mice. This effect was associated with an increase in multilocular lipid droplets and mitochondria in perivascular adipose tissue (PVAT). Immunofluorescence analysis confirmed that there was an increase in capillary density and M2 macrophage infiltration, accompanied by a decrease in tumour necrosis factor (TNF)-α expression in the localized PVAT of TLR4-/-→LDLR-/- mice. In vitro studies indicated that bone marrow-derived macrophages (BMDMs) from WT mice demonstrated an M1-like phenotype and expression of inflammatory cytokines induced by palmitate. These effects were attenuated in BMDMs isolated from TLR4-/- mice. Furthermore, brown adipocytes incubated with conditioned medium (CM) derived from palmitate-treated BMDMs, exhibited larger and more unilocular lipid droplets, and reduced expression of brown adipocyte-specific markers and perilipin-1 compared to those observed in brown adipocytes exposed to CM from palmitate-treated BMDMs of TLR4-/- mice. This decreased potency was primarily due to TNF-α, as demonstrated by the capacity of the TNF-α neutralizing antibody to reverse these effects. CONCLUSIONS These results suggest that haematopoietic-specific deletion of TLR4 promotes PVAT homeostasis, which is involved in reducing macrophage-induced TNF-α secretion and increasing mitochondrial biogenesis in brown adipocytes.
Collapse
Affiliation(s)
- Penghao Liu
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China; Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Gaojian Huang
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Zhiyong Cao
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China; Department of General Internal Medicine, Branch of 411 Hospital of People's Liberation Army, Shanghai 200433, China
| | - Qihai Xie
- Department of Cardiology, Shanghai Jia Ding District Central Hospital, Shanghai 201800, China
| | - Tong Wei
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Chenglin Huang
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Qun Li
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Mengwei Sun
- Key Laboratory of State General Administration of Sport, Shanghai Research Institute of Sports Science, Shanghai 200030, China
| | - Weili Shen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| | - Pingjin Gao
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
103
|
Subramanian S, Pallati PK, Sharma P, Agrawal DK, Nandipati KC. TREM-1 associated macrophage polarization plays a significant role in inducing insulin resistance in obese population. J Transl Med 2017; 15:85. [PMID: 28454543 PMCID: PMC5408415 DOI: 10.1186/s12967-017-1187-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/19/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND TREM-1 acts as an amplifier of inflammation expressed on macrophages. The objective of this study was to evaluate the relationship between TREM-1 and macrophage polarization, and association of TREM-1 and M1 macrophage polarization with insulin resistance (IR) in obese population compared to non-obese population. METHODS We enrolled 38 patients after obtaining IRB approval for this study. We evaluated the mRNA and protein expression levels of general macrophage marker (CD68), M1 marker (CD86, CCR7, iNOS, IFNγ, TNF-α and IL-6,), M2 marker (CD206, CD163, IL-10, IL-4) and chemokine axis (MCP-1, CCR2 and CCR5) along with TREM-1 and TREM-2 in omentum fat, subcutaneous fat, and liver biopsy tissues of non-obese (N = 5), obese non-diabetics, (N = 16) and obese diabetics (N = 17). RESULTS The results of our study showed over-expression of TREM-1, M1 markers and down-regulation of TREM-2 and M2 markers in the omentum, subcutaneous and liver biopsies of obese patients (diabetics and non-diabetics) compared to non-obese patients. Overall, the obese diabetic group showed a significant (p < 0.05) higher number of patients with over expression of M1 markers (TREM-1, CD68, CD86, CCR-7, iNOS, IFN-γ, TNF-α, IL-6, MCP-1, CCR-2 and CCR-5) and down-regulation of M2 markers (CD206, CD163 and IL-4) in liver biopsy compared to obese non-diabetics. CONCLUSIONS TREM-1 expression is significantly increased along with the M1 markers in liver biopsy of obese diabetic (17/17) and obese non-diabetic patients (9/16). Our data suggests that TREM-1 overexpression and M1 macrophage polarization are associated with obesity-induced IR.
Collapse
Affiliation(s)
- Saravanan Subramanian
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE USA
| | - Pradeep K. Pallati
- Department of Surgery, Creighton University School of Medicine, 601 N. 30th Street, Suite # 3700, Omaha, NE 68131 USA
| | - Poonam Sharma
- Department of Pathology, Creighton University School of Medicine, Omaha, NE USA
| | - Devendra K. Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE USA
| | - Kalyana C. Nandipati
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE USA
- Department of Surgery, Creighton University School of Medicine, 601 N. 30th Street, Suite # 3700, Omaha, NE 68131 USA
| |
Collapse
|
104
|
Allen JN, Dey A, Nissly R, Fraser J, Yu S, Balandaram G, Peters JM, Hankey-Giblin PA. Isolation, Characterization, and Purification of Macrophages from Tissues Affected by Obesity-related Inflammation. J Vis Exp 2017:55445. [PMID: 28447981 PMCID: PMC5564464 DOI: 10.3791/55445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Obesity promotes a chronic inflammatory state that is largely mediated by tissue-resident macrophages as well as monocyte-derived macrophages. Diet-induced obesity (DIO) is a valuable model in studying the role of macrophage heterogeneity; however, adequate macrophage isolations are difficult to acquire from inflamed tissues. In this protocol, we outline the isolation steps and necessary troubleshooting guidelines derived from our studies for obtaining a suitable population of tissue-resident macrophages from mice following 18 weeks of high-fat (HFD) or high-fat/high-cholesterol (HFHCD) diet intervention. This protocol focuses on three hallmark tissues studied in obesity and atherosclerosis including the liver, white adipose tissues (WAT), and the aorta. We highlight how dualistic usage of flow cytometry can achieve a new dimension of isolation and characterization of tissue-resident macrophages. A fundamental section of this protocol addresses the intricacies underlying tissue-specific enzymatic digestions and macrophage isolation, and subsequent cell-surface antibody staining for flow cytometric analysis. This protocol addresses existing complexities underlying fluorescent-activated cell sorting (FACS) and presents clarifications to these complexities so as to obtain broad range characterization from adequately sorted cell populations. Alternate enrichment methods are included for sorting cells, such as the dense liver, allowing for flexibility and time management when working with FACS. In brief, this protocol aids the researcher to evaluate macrophage heterogeneity from a multitude of inflamed tissues in a given study and provides insightful troubleshooting tips that have been successful for favorable cellular isolation and characterization of immune cells in DIO-mediated inflammation.
Collapse
Affiliation(s)
- Joselyn N Allen
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University;
| | - Adwitia Dey
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University
| | - Ruth Nissly
- Microscopy and Cytometry Facility, The Huck Institutes of Life Sciences, Pennsylvania State University
| | - James Fraser
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University
| | - Shan Yu
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University
| | - Gayathri Balandaram
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University
| | | |
Collapse
|
105
|
Impairment of systemic DHA synthesis affects macrophage plasticity and polarization: implications for DHA supplementation during inflammation. Cell Mol Life Sci 2017; 74:2815-2826. [PMID: 28299384 PMCID: PMC5491590 DOI: 10.1007/s00018-017-2498-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/15/2017] [Accepted: 02/28/2017] [Indexed: 02/06/2023]
Abstract
Docosahexaenoic acid (DHA) is an omega-3 fatty acid obtained from the diet or synthesized from alpha-linolenic acid through the action of fatty acid elongases (ELOVL) and desaturases. DHA plays important roles in the central nervous system as well as in peripheral organs and is the precursor of several molecules that regulate resolution of inflammation. In the present study, we questioned whether impaired synthesis of DHA affected macrophage plasticity and polarization both in vitro and in vivo models. For this we investigated the activation status and inflammatory response of bone marrow-derived M1 and M2 macrophages obtained from mice deficient of Elovl2 (Elovl2−/−), a key enzyme for DHA synthesis in mammals. Although both wild type and Elovl2−/− mice were able to generate efficient M1 and M2 macrophages, M1 cells derived from Elovl2−/− mice showed an increased expression of key markers (iNOS, CD86 and MARCO) and cytokines (IL-6, IL-12 and IL-23). However, M2 macrophages exhibited upregulated M1-like markers like CD80, CD86 and IL-6, concomitantly with a downregulation of their signature marker CD206. These effects were counteracted in cells obtained from DHA-supplemented animals. Finally, white adipose tissue of Elovl2−/− mice presented an M1-like pro-inflammatory phenotype. Hence, impairment of systemic DHA synthesis delineates an alteration of M1/M2 macrophages both in vitro and in vivo, with M1 being hyperactive and more pro-inflammatory while M2 less protective, supporting the view that DHA has a key role in controlling the balance between pro- and anti-inflammatory processes.
Collapse
|
106
|
Zhan W, Lu F. Activated macrophages as key mediators of capsule formation on adipose constructs in tissue engineering chamber models. Cell Biol Int 2017; 41:354-360. [DOI: 10.1002/cbin.10731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 01/15/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Weiqing Zhan
- Department of Plastic and Cosmetic Surgery; Nanfang Hospital, Southern Medical University, Guang Zhou; Guang Dong People's Republic of China
- O'Brien Institute Department; St Vincent's Institute of Medical Research; Victoria Australia
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery; Nanfang Hospital, Southern Medical University, Guang Zhou; Guang Dong People's Republic of China
| |
Collapse
|
107
|
Schmatz R, Bitencourt MR, Patias LD, Beck M, da C. Alvarez G, Zanini D, Gutierres JM, Diehl LN, Pereira LB, Leal CA, Duarte MF, Schetinger MR, Morsch VM. Evaluation of the biochemical, inflammatory and oxidative profile of obese patients given clinical treatment and bariatric surgery. Clin Chim Acta 2017; 465:72-79. [DOI: 10.1016/j.cca.2016.12.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 11/09/2016] [Accepted: 12/12/2016] [Indexed: 10/20/2022]
|
108
|
Winn NC, Vieira-Potter VJ, Gastecki ML, Welly RJ, Scroggins RJ, Zidon TM, Gaines TL, Woodford ML, Karasseva NG, Kanaley JA, Sacks HS, Padilla J. Loss of UCP1 exacerbates Western diet-induced glycemic dysregulation independent of changes in body weight in female mice. Am J Physiol Regul Integr Comp Physiol 2016; 312:R74-R84. [PMID: 27881400 DOI: 10.1152/ajpregu.00425.2016] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/15/2016] [Accepted: 11/17/2016] [Indexed: 01/23/2023]
Abstract
We tested the hypothesis that female mice null for uncoupling protein 1 (UCP1) would have increased susceptibility to Western diet-induced "whitening" of brown adipose tissue (AT) and glucose intolerance. Six-week-old C57BL/6J wild-type (WT) and UCP1 knockout (UCP1-/-) mice, housed at 25°C, were randomized to either a control diet (10% kcal from fat) or Western diet (45% kcal from fat and 1% cholesterol) for 28 wk. Loss of UCP1 had no effect on energy intake, energy expenditure, spontaneous physical activity, weight gain, or visceral white AT mass. Despite similar susceptibility to weight gain compared with WT, UCP1-/- exhibited whitening of brown AT evidenced by a striking ~500% increase in mass and appearance of large unilocular adipocytes, increased expression of genes related to inflammation, immune cell infiltration, and endoplasmic reticulum/oxidative stress (P < 0.05), and decreased mitochondrial subunit protein (COX I, II, III, and IV, P < 0.05), all of which were exacerbated by Western diet (P < 0.05). UCP1-/- mice also developed liver steatosis and glucose intolerance, which was worsened by Western diet. Collectively, these findings demonstrate that loss of UCP1 exacerbates Western diet-induced whitening of brown AT, glucose intolerance, and induces liver steatosis. Notably, the adverse metabolic manifestations of UCP1-/- were independent of changes in body weight, visceral adiposity, and energy expenditure. These novel findings uncover a previously unrecognized metabolic protective role of UCP1 that is independent of its already established role in energy homeostasis.
Collapse
Affiliation(s)
- Nathan C Winn
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | | | - Michelle L Gastecki
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Rebecca J Welly
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Rebecca J Scroggins
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Terese M Zidon
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - T'Keaya L Gaines
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Makenzie L Woodford
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | | | - Jill A Kanaley
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Harold S Sacks
- Endocrine and Diabetes Division, Veterans Greater Los Angeles Healthcare System and Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Jaume Padilla
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri; .,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; and.,Child Health, University of Missouri, Columbia, Missouri
| |
Collapse
|
109
|
Li Z, Xu F, Wang Z, Dai T, Ma C, Liu B, Liu Y. Macrophages Undergo M1-to-M2 Transition in Adipose Tissue Regeneration in a Rat Tissue Engineering Model. Artif Organs 2016; 40:E167-E178. [DOI: 10.1111/aor.12756] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/08/2016] [Accepted: 03/21/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Zhijin Li
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery; School of Stomatology, Fourth Military Medical University; Xi'an P.R. China
- Department of Stomatology; Wuhan General Hospital of Guangzhou Command; Wuhan P.R. China
| | - Fangfang Xu
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery; School of Stomatology, Fourth Military Medical University; Xi'an P.R. China
| | - Zhifa Wang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery; School of Stomatology, Fourth Military Medical University; Xi'an P.R. China
| | - Taiqiang Dai
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery; School of Stomatology, Fourth Military Medical University; Xi'an P.R. China
| | - Chao Ma
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery; School of Stomatology, Fourth Military Medical University; Xi'an P.R. China
| | - Bin Liu
- State Key Laboratory of Military Stomatology; Laboratory Animal Center, School of Stomatology, Fourth Military Medical University; Xi'an P.R. China
| | - Yanpu Liu
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery; School of Stomatology, Fourth Military Medical University; Xi'an P.R. China
| |
Collapse
|
110
|
Vergoni B, Cornejo PJ, Gilleron J, Djedaini M, Ceppo F, Jacquel A, Bouget G, Ginet C, Gonzalez T, Maillet J, Dhennin V, Verbanck M, Auberger P, Froguel P, Tanti JF, Cormont M. DNA Damage and the Activation of the p53 Pathway Mediate Alterations in Metabolic and Secretory Functions of Adipocytes. Diabetes 2016; 65:3062-74. [PMID: 27388216 DOI: 10.2337/db16-0014] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 07/01/2016] [Indexed: 11/13/2022]
Abstract
Activation of the p53 pathway in adipose tissue contributes to insulin resistance associated with obesity. However, the mechanisms of p53 activation and the effect on adipocyte functions are still elusive. Here we found a higher level of DNA oxidation and a reduction in telomere length in adipose tissue of mice fed a high-fat diet and an increase in DNA damage and activation of the p53 pathway in adipocytes. Interestingly, hallmarks of chronic DNA damage are visible at the onset of obesity. Furthermore, injection of lean mice with doxorubicin, a DNA damage-inducing drug, increased the expression of chemokines in adipose tissue and promoted its infiltration by proinflammatory macrophages and neutrophils together with adipocyte insulin resistance. In vitro, DNA damage in adipocytes increased the expression of chemokines and triggered the production of chemotactic factors for macrophages and neutrophils. Insulin signaling and effect on glucose uptake and Glut4 translocation were decreased, and lipolysis was increased. These events were prevented by p53 inhibition, whereas its activation by nutlin-3 reproduced the DNA damage-induced adverse effects. This study reveals that DNA damage in obese adipocytes could trigger p53-dependent signals involved in alteration of adipocyte metabolism and secretory function leading to adipose tissue inflammation, adipocyte dysfunction, and insulin resistance.
Collapse
Affiliation(s)
- Bastien Vergoni
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France
| | - Pierre-Jean Cornejo
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France
| | - Jérôme Gilleron
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France
| | - Mansour Djedaini
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France
| | - Franck Ceppo
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France
| | - Arnaud Jacquel
- Université Côte d'Azur, C3M, Nice, France INSERM, UMR 1065, C3M, Team 2 Cell Death, Differentiation and Cancer, Nice, France
| | - Gwennaelle Bouget
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France
| | - Clémence Ginet
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France
| | - Teresa Gonzalez
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France INSERM, UMR 1062, Nutrition, Obesity and Risk of Thrombosis, Marseille, France
| | - Julie Maillet
- CNRS, UMR 8199, Lille Pasteur Institute, Lille, France Lille University, Lille, France European Genomic Institute for Diabetes, Lille, France
| | - Véronique Dhennin
- CNRS, UMR 8199, Lille Pasteur Institute, Lille, France Lille University, Lille, France European Genomic Institute for Diabetes, Lille, France
| | - Marie Verbanck
- CNRS, UMR 8199, Lille Pasteur Institute, Lille, France Lille University, Lille, France European Genomic Institute for Diabetes, Lille, France
| | - Patrick Auberger
- Université Côte d'Azur, C3M, Nice, France INSERM, UMR 1065, C3M, Team 2 Cell Death, Differentiation and Cancer, Nice, France
| | - Philippe Froguel
- CNRS, UMR 8199, Lille Pasteur Institute, Lille, France Lille University, Lille, France European Genomic Institute for Diabetes, Lille, France Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London, U.K
| | - Jean-François Tanti
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France
| | - Mireille Cormont
- INSERM, UMR 1065, C3M, Team 7 Molecular and Cellular Physiopathology of Obesity and Diabetes, Nice, France Université Côte d'Azur, C3M, Nice, France
| |
Collapse
|
111
|
Ringling RE, Gastecki ML, Woodford ML, Lum-Naihe KJ, Grant RW, Pulakat L, Vieira-Potter VJ, Padilla J. Loss of Nlrp3 Does Not Protect Mice from Western Diet-Induced Adipose Tissue Inflammation and Glucose Intolerance. PLoS One 2016; 11:e0161939. [PMID: 27583382 PMCID: PMC5008778 DOI: 10.1371/journal.pone.0161939] [Citation(s) in RCA: 20] [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: 01/13/2016] [Accepted: 08/15/2016] [Indexed: 02/03/2023] Open
Abstract
We tested the hypothesis that loss of Nlrp3 would protect mice from Western diet-induced adipose tissue (AT) inflammation and associated glucose intolerance and cardiovascular complications. Five-week old C57BL6J wild-type (WT) and Nlrp3 knockout (Nlrp3-/-) mice were randomized to either a control diet (10% kcal from fat) or Western diet (45% kcal from fat and 1% cholesterol) for 24 weeks (n = 8/group). Contrary to our hypothesis that obesity-mediated white AT inflammation is Nlrp3-dependent, we found that Western diet-induced expression of AT inflammatory markers (i.e., Cd68, Cd11c, Emr1, Itgam, Lgals, Il18, Mcp1, Tnf, Ccr2, Ccl5 mRNAs, and Mac-2 protein) were not accompanied by increased caspase-1 cleavage, a hallmark feature of NLRP3 inflammasome activation. Furthermore, Nlrp3 null mice were not protected from Western diet-induced white or brown AT inflammation. Although Western diet promoted glucose intolerance in both WT and Nlrp3-/- mice, Nlrp3-/- mice were protected from Western diet-induced aortic stiffening. Additionally, Nlrp3-/- mice exhibited smaller cardiomyocytes and reduced cardiac fibrosis, independent of diet. Collectively, these findings suggest that presence of the Nlrp3 gene is not required for Western diet-induced AT inflammation and/or glucose intolerance; yet Nlrp3 appears to play a role in potentiating arterial stiffening, cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Rebecca E. Ringling
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Michelle L. Gastecki
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Makenzie L. Woodford
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Kelly J. Lum-Naihe
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Ryan W. Grant
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, United States of America
| | - Lakshmi Pulakat
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States of America
- Department of Medicine, University of Missouri, Columbia, Missouri, United States of America
- Research Service, Harry S Truman Memorial Veterans Affairs Hospital, Columbia, Missouri, United States of America
| | - Victoria J. Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States of America
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Child Health, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
112
|
Seki M, LaCanna R, Powers JC, Vrakas C, Liu F, Berretta R, Chacko G, Holten J, Jadiya P, Wang T, Arkles JS, Copper JM, Houser SR, Huang J, Patel VV, Recchia FA. Class I Histone Deacetylase Inhibition for the Treatment of Sustained Atrial Fibrillation. J Pharmacol Exp Ther 2016; 358:441-9. [PMID: 27353074 PMCID: PMC4998670 DOI: 10.1124/jpet.116.234591] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/22/2016] [Indexed: 01/07/2023] Open
Abstract
Current therapies are less effective for treating sustained/permanent versus paroxysmal atrial fibrillation (AF). We and others have previously shown that histone deacetylase (HDAC) inhibition reverses structural and electrical atrial remodeling in mice with inducible, paroxysmal-like AF. Here, we hypothesize an important, specific role for class I HDACs in determining structural atrial alterations during sustained AF. The class I HDAC inhibitor N-acetyldinaline [4-(acetylamino)-N-(2-amino-phenyl) benzamide] (CI-994) was administered for 2 weeks (1 mg/kg/day) to Hopx transgenic mice with atrial remodeling and inducible AF and to dogs with atrial tachypacing-induced sustained AF. Class I HDAC inhibition prevented atrial fibrosis and arrhythmia inducibility in mice. Dogs were divided into three groups: 1) sinus rhythm, 2) sustained AF plus vehicle, and 3) sustained AF plus CI-994. In group 3, the time in AF over 2 weeks was reduced by 30% compared with group 2, along with attenuated atrial fibrosis and intra-atrial adipocyte infiltration. Moreover, group 2 dogs had higher atrial and serum inflammatory cytokines, adipokines, and atrial immune cells and adipocytes compared with groups 1 and 3. On the other hand, groups 2 and 3 displayed similar left atrial size, ventricular function, and mitral regurgitation. Importantly, the same histologic alterations found in dogs with sustained AF and reversed by CI-994 were also present in atrial tissue from transplanted patients with chronic AF. This is the first evidence that, in sustained AF, class I HDAC inhibition can reduce the total time of fibrillation, atrial fibrosis, intra-atrial adipocytes, and immune cell infiltration without significant effects on cardiac function.
Collapse
Affiliation(s)
- Mitsuru Seki
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Ryan LaCanna
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Jeffery C Powers
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Christine Vrakas
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Fang Liu
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Remus Berretta
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Geena Chacko
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - John Holten
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Pooja Jadiya
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Tao Wang
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Jeffery S Arkles
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Joshua M Copper
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Steven R Houser
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Jianhe Huang
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Vickas V Patel
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| | - Fabio A Recchia
- Cardiovascular Research Center (M.S., R.L.C., J.C.P., C.V., R.B., G.C., Jo.H., P.J., T.W., S.R.H., Ji.H., V.V.P., F.A.R.), and Section of Clinical Cardiac Electrophysiology (J.S.A., J.M.C., V.V.P.), Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy (F.A.R.); and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania (F.L.)
| |
Collapse
|
113
|
Novel Role of Endogenous Catalase in Macrophage Polarization in Adipose Tissue. Mediators Inflamm 2016; 2016:8675905. [PMID: 27597806 PMCID: PMC5002490 DOI: 10.1155/2016/8675905] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/11/2016] [Accepted: 06/29/2016] [Indexed: 12/19/2022] Open
Abstract
Macrophages are important components of adipose tissue inflammation, which results in metabolic diseases such as insulin resistance. Notably, obesity induces a proinflammatory phenotypic switch in adipose tissue macrophages, and oxidative stress facilitates this switch. Thus, we examined the role of endogenous catalase, a key regulator of oxidative stress, in the activity of adipose tissue macrophages in obese mice. Catalase knockout (CKO) exacerbated insulin resistance, amplified oxidative stress, and accelerated macrophage infiltration into epididymal white adipose tissue in mice on normal or high-fat diet. Interestingly, catalase deficiency also enhanced classical macrophage activation (M1) and inflammation but suppressed alternative activation (M2) regardless of diet. Similarly, pharmacological inhibition of catalase activity using 3-aminotriazole induced the same phenotypic switch and inflammatory response in RAW264.7 macrophages. Finally, the same phenotypic switch and inflammatory responses were observed in primary bone marrow-derived macrophages from CKO mice. Taken together, the data indicate that endogenous catalase regulates the polarization of adipose tissue macrophages and thereby inhibits inflammation and insulin resistance.
Collapse
|
114
|
Bjørklund G, Chirumbolo S. Role of oxidative stress and antioxidants in daily nutrition and human health. Nutrition 2016; 33:311-321. [PMID: 27746034 DOI: 10.1016/j.nut.2016.07.018] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/14/2016] [Accepted: 07/24/2016] [Indexed: 12/20/2022]
Abstract
Diet may be defined as a complex process that should involve a deeper comprehension of metabolism, energy balance, and the molecular pathways involved in cellular stress response and survival, gut microflora genetics, enzymatic polymorphism within the human population, and the role of plant-derived polyphenols in this context. Metabolic syndrome, encompassing pathologies with a relatively high morbidity, such as type 2 diabetes, obesity, and cardiovascular disease, is a bullet point of the big concern about how daily dietary habits should promote health and prevent metabolic impairments to prevent hospitalization and the need for health care. From a clinical point of view, very few papers deal with this concern, whereas most of the evidence reported focuses on in vitro and animal models, which study the activity of phytochemicals contained in the daily diet. A fundamental issue addressed by dietitians deals with the role exerted by redox-derived reactive species. Most plant polyphenols act as antioxidants, but recent evidence supports the idea that these compounds primarily activate a mild oxidative stress to elicit a positive, beneficial response from cells. How these compounds may act upon the detoxifying system exerting a scavenging role from reactive oxygen or nitrogen species is still a matter of debate; however, it can be argued that their role is even more complex than expected, acting as signaling molecules in the cross-talk mitochondria-endoplasmic reticulum and in enzymatic pathways involved in the energetic balance. In this relationship, a fundamental role is played by the brain-adipose tissue-gut axis. The aim of this review was to elucidate this topic and the state of art about the role of reactive species in cell signaling and the function of metabolism and survival to reappraise the role of plant-derived chemicals.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy.
| |
Collapse
|
115
|
Female sex and obesity increase photophobic behavior in mice. Neuroscience 2016; 331:99-108. [PMID: 27328418 DOI: 10.1016/j.neuroscience.2016.06.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/31/2016] [Accepted: 06/12/2016] [Indexed: 12/16/2022]
Abstract
Migraine affects predominantly women. Furthermore, epidemiological studies suggest that obesity is a risk factor for migraine and this association is influenced by sex. However, the biological basis for this bias is unclear. To address this issue, we assessed light avoidant behavior, a surrogate of photophobia, in female C57BL/6J mice fed regular diet (RD) or high-fat diet (HFD, 60% kcal from fat). We first assessed sex differences in basal photophobia in 20-25-week-old mice and found that both obese and lean females spent significantly less time in light than their male counterparts. Next, we assessed photophobia evoked by trigeminal stimulation with intradermal capsaicin. Females at 20-25weeks of age did not display capsaicin-evoked photophobic behavior unless they had diet-induced obesity. When we tested 8-11-week-old females to determine if the diet alone could be responsible for this effect, we found that both HFD and RD 8-11-week-old females exhibit capsaicin-evoked photophobic behavior. This is in contrast to what we have previously shown in males and indicates a sex difference in the photophobic behavior of mice. Comparison of 20-25-week-old RD mice with 8-11-week-old RD mice suggests that age or age-related weight gain may contribute to capsaicin-evoked photophobic behavior in males, but not in females. These findings suggest that obesity exacerbates photophobia in both sexes, but additional work is needed to understand the sex- and age-specific mechanisms that may contribute to photophobia and trigeminal pain.
Collapse
|
116
|
Differentiation of preadipocytes and mature adipocytes requires PSMB8. Sci Rep 2016; 6:26791. [PMID: 27225296 PMCID: PMC4880908 DOI: 10.1038/srep26791] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 05/10/2016] [Indexed: 01/17/2023] Open
Abstract
The differentiation of adipocytes is tightly regulated by a variety of intrinsic molecules and also by extrinsic molecules produced by adjacent cells. Dysfunction of adipocyte differentiation causes lipodystrophy, which impairs glucose and lipid homeostasis. Although dysfunction of immunoproteasomes causes partial lipodystrophy, the detailed molecular mechanisms remain to be determined. Here, we demonstrate that Psmb8, a catalytic subunit for immunoproteasomes, directly regulates the differentiation of preadipocytes and additionally the differentiation of preadipocytes to mature adipocytes. Psmb8(-/-) mice exhibited slower weight gain than wild-type mice, and this was accompanied by reduced adipose tissue volume and smaller size of mature adipocytes compared with controls. Blockade of Psmb8 activity in 3T3-L1 cells disturbed the differentiation to mature adipocytes. Psmb8(-/-) mice had fewer preadipocyte precursors, fewer preadipocytes and a reduced ability to differentiate preadipocytes toward mature adipocytes. Our data demonstrate that Psmb8-mediated immunoproteasome activity is a direct regulator of the differentiation of preadipocytes and their ultimate maturation.
Collapse
|
117
|
Ciszek BP, O'Buckley SC, Nackley. AG. Persistent Catechol-O-methyltransferase-dependent Pain Is Initiated by Peripheral β-Adrenergic Receptors. Anesthesiology 2016; 124:1122-35. [PMID: 26950706 PMCID: PMC5015695 DOI: 10.1097/aln.0000000000001070] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Patients with chronic pain disorders exhibit increased levels of catecholamines alongside diminished activity of catechol-O-methyltransferase (COMT), an enzyme that metabolizes catecholamines. The authors found that acute pharmacologic inhibition of COMT in rodents produces hypersensitivity to mechanical and thermal stimuli via β-adrenergic receptor (βAR) activation. The contribution of distinct βAR populations to the development of persistent pain linked to abnormalities in catecholamine signaling requires further investigation. METHODS Here, the authors sought to determine the contribution of peripheral, spinal, and supraspinal βARs to persistent COMT-dependent pain. They implanted osmotic pumps to deliver the COMT inhibitor OR486 (Tocris, USA) for 2 weeks. Behavioral responses to mechanical and thermal stimuli were evaluated before and every other day after pump implantation. The site of action was evaluated in adrenalectomized rats receiving sustained OR486 or in intact rats receiving sustained βAR antagonists peripherally, spinally, or supraspinally alongside OR486. RESULTS The authors found that male (N = 6) and female (N = 6) rats receiving sustained OR486 exhibited decreased paw withdrawal thresholds (control 5.74 ± 0.24 vs. OR486 1.54 ± 0.08, mean ± SEM) and increased paw withdrawal frequency to mechanical stimuli (control 4.80 ± 0.22 vs. OR486 8.10 ± 0.13) and decreased paw withdrawal latency to thermal heat (control 9.69 ± 0.23 vs. OR486 5.91 ± 0.11). In contrast, adrenalectomized rats (N = 12) failed to develop OR486-induced hypersensitivity. Furthermore, peripheral (N = 9), but not spinal (N = 4) or supraspinal (N = 4), administration of the nonselective βAR antagonist propranolol, the β2AR antagonist ICI-118,511, or the β3AR antagonist SR59230A blocked the development of OR486-induced hypersensitivity. CONCLUSIONS Peripheral adrenergic input is necessary for the development of persistent COMT-dependent pain, and peripherally-acting βAR antagonists may benefit chronic pain patients.
Collapse
Affiliation(s)
- Brittney P. Ciszek
- Center for Pain Research and Innovation, Koury Oral Health Sciences Building, University of North Carolina, Chapel Hill NC 27599-7455
| | - Sandra C. O'Buckley
- Center for Pain Research and Innovation, Koury Oral Health Sciences Building, University of North Carolina, Chapel Hill NC 27599-7455
| | - Andrea G. Nackley.
- Center for Pain Research and Innovation, Koury Oral Health Sciences Building, University of North Carolina, Chapel Hill NC 27599-7455
| |
Collapse
|
118
|
Hersoug LG, Møller P, Loft S. Gut microbiota-derived lipopolysaccharide uptake and trafficking to adipose tissue: implications for inflammation and obesity. Obes Rev 2016; 17:297-312. [PMID: 26712364 DOI: 10.1111/obr.12370] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 12/12/2022]
Abstract
The composition of the gut microbiota and excessive ingestion of high-fat diets (HFD) are considered to be important factors for development of obesity. In this review we describe a coherent mechanism of action for the development of obesity, which involves the composition of gut microbiota, HFD, low-grade inflammation, expression of fat translocase and scavenger receptor CD36, and the scavenger receptor class B type 1 (SR-BI). SR-BI binds to both lipids and lipopolysaccharide (LPS) from Gram-negative bacteria, which may promote incorporation of LPS in chylomicrons (CMs). These CMs are transported via lymph to the circulation, where LPS is transferred to other lipoproteins by translocases, preferentially to HDL. LPS increases the SR-BI binding, transcytosis of lipoproteins over the endothelial barrier,and endocytosis in adipocytes. Especially large size adipocytes with high metabolic activity absorb LPS-rich lipoproteins. In addition, macrophages in adipose tissue internalize LPS-lipoproteins. This may contribute to the polarization from M2 to M1 phenotype, which is a consequence of increased LPS delivery into the tissue during hypertrophy. In conclusion, evidence suggests that LPS is involved in the development of obesity as a direct targeting molecule for lipid delivery and storage in adipose tissue.
Collapse
Affiliation(s)
- L-G Hersoug
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - P Møller
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - S Loft
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
119
|
Böhm A, Weigert C, Staiger H, Häring HU. Exercise and diabetes: relevance and causes for response variability. Endocrine 2016; 51:390-401. [PMID: 26643313 PMCID: PMC4762932 DOI: 10.1007/s12020-015-0792-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/28/2015] [Indexed: 12/31/2022]
Abstract
Exercise as a key prevention strategy for diabetes and obesity is commonly accepted and recommended throughout the world. Unfortunately, not all individuals profit to the same extent, some exhibit exercise resistance. This phenomenon of non-response to exercise is found for several endpoints, including glucose tolerance and insulin sensitivity. Since these non-responders are of notable quantity, there is the need to understand the underlying mechanisms and to identify predictors of response. This displays the basis to develop personalized training intervention regimes. In this review, we summarize the current knowledge on response variability, with focus on human studies and improvement of glucose homeostasis as outcome.
Collapse
Affiliation(s)
- Anja Böhm
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany
| | - Cora Weigert
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany
| | - Harald Staiger
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, Tübingen, Germany.
- German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany.
| |
Collapse
|
120
|
Vianello E, Dozio E, Arnaboldi F, Marazzi MG, Martinelli C, Lamont J, Tacchini L, Sigrüner A, Schmitz G, Corsi Romanelli MM. Epicardial adipocyte hypertrophy: Association with M1-polarization and toll-like receptor pathways in coronary artery disease patients. Nutr Metab Cardiovasc Dis 2016; 26:246-253. [PMID: 26841679 DOI: 10.1016/j.numecd.2015.12.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 11/02/2015] [Accepted: 12/07/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS In coronary artery disease (CAD) epicardial adipose tissue (EAT) shows an elevated inflammatory infiltrate. Toll-like receptors (TLRs) are important mediators of adipose tissue inflammation and they are able to recognize endogenous products released by damaged cells. Because adipocyte death may be driven by hypertrophy, our aim was to investigate in CAD and non-CAD patients the association between EAT adipocyte size, macrophage infiltration/polarization and TLR-2 and TLR-4 expression. METHODS AND RESULTS EAT biopsies were collected from CAD and non-CAD patients. The adipocyte size was determined by morphometric analysis. Microarray technology was used for gene expression analysis; macrophage phenotype and TLRs expression were analyzed by immunofluorescence and immunohistochemical techniques. Inflammatory mediator levels were determined by immunoassays. EAT adipocytes were larger in CAD than non-CAD patients and do not express perilipin A, a marker of lipid droplet integrity. In CAD, EAT is more infiltrated by CD68-positive cells which are polarized toward an M1 state (CD11c positive) and presents an increased pro-inflammatory profile. Both TLR-2 and TLR-4 expression is higher in EAT from CAD and observed on all the CD68-positive cells. CONCLUSIONS Our findings suggested that EAT hypertrophy in CAD promotes adipocyte degeneration and drives local inflammation through increased infiltration of macrophages which are mainly polarized towards an M1 state and express both TLR-2 and TLR-4.
Collapse
Affiliation(s)
- E Vianello
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy.
| | - E Dozio
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - F Arnaboldi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - M G Marazzi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - C Martinelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - J Lamont
- Randox Laboratories LTD, R&D, Crumlin-Antrim, Belfast, Northern Ireland, UK
| | - L Tacchini
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - A Sigrüner
- Institute for Clinical Chemistry and Laboratory Medicine, University of Regensburg, Germany
| | - G Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University of Regensburg, Germany
| | - M M Corsi Romanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy; SMEL-1 Clinical Pathology, I.R.C.C.S. Policlinico San Donato, Milan, Italy
| |
Collapse
|
121
|
|
122
|
Abstract
Adipose tissue inflammation links obesity and metabolic disease. Both exercise and estrogen improve metabolic health, enhance mitochondrial function, and have antiinflammatory effects. We hypothesize that there is an inverse relationship between mitochondrial function and inflammation in adipose tissue and that exercise acts as an estrogen "mimetic." Explicitly, exercise may improve adipose tissue "immunometabolism" by improving mitochondrial function and reducing inflammation.
Collapse
Affiliation(s)
- Victoria J Vieira-Potter
- 1Departments of Nutrition and Exercise Physiology, and 2Child Health, and 3Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
| | | | | |
Collapse
|
123
|
Vieira-Potter VJ, Lee S, Bayless DS, Scroggins RJ, Welly RJ, Fleming NJ, Smith TN, Meers GM, Hill MA, Rector RS, Padilla J. Disconnect between adipose tissue inflammation and cardiometabolic dysfunction in Ossabaw pigs. Obesity (Silver Spring) 2015; 23:2421-9. [PMID: 26524201 PMCID: PMC4701582 DOI: 10.1002/oby.21252] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 07/15/2015] [Accepted: 07/16/2015] [Indexed: 02/01/2023]
Abstract
OBJECTIVE The Ossabaw pig is emerging as an attractive model of human cardiometabolic disease because of its size and susceptibility to atherosclerosis, among other characteristics. The relationship between adipose tissue inflammation and metabolic dysfunction in this model was investigated here. METHODS Young female Ossabaw pigs were fed a Western-style high-fat diet (HFD) (n = 4) or control low-fat diet (LFD) (n = 4) for a period of 9 months and compared for cardiometabolic outcomes and adipose tissue inflammation. RESULTS The HFD-fed "OBESE" pigs were 2.5 times heavier (P < 0.001) than LFD-fed "LEAN" pigs and developed severe obesity. HFD feeding caused pronounced dyslipidemia, hypertension, and insulin resistance (systemic and adipose), as well as induction of inflammatory genes, impairments in vasomotor reactivity to insulin, and atherosclerosis in the coronary arteries. Remarkably, visceral, subcutaneous, and perivascular adipose tissue inflammation (via FACS analysis and RT-PCR) was not increased in OBESE pigs, nor were circulating inflammatory cytokines. CONCLUSIONS These findings reveal a disconnect between adipose tissue inflammation and cardiometabolic dysfunction induced by Western diet feeding in the Ossabaw pig model.
Collapse
Affiliation(s)
| | - Sewon Lee
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
- Division of Sport Science & Sport Science Institute, Incheon National University, Incheon, South Korea
| | - David S. Bayless
- Medical Pharmacology and Physiology, University of Missouri, Columbia, MO
| | | | - Rebecca J. Welly
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | | | - Thomas N. Smith
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | - Grace M. Meers
- Research Service, Harry S Truman Memorial VA Medical Center, Columbia, MO
| | - Michael A. Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
- Research Service, Harry S Truman Memorial VA Medical Center, Columbia, MO
| | - R. Scott Rector
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
- Research Service, Harry S Truman Memorial VA Medical Center, Columbia, MO
- Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO
| | - Jaume Padilla
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO
- Child Health, University of Missouri, Columbia, MO
| |
Collapse
|
124
|
Favennec M, Hennart B, Caiazzo R, Leloire A, Yengo L, Verbanck M, Arredouani A, Marre M, Pigeyre M, Bessede A, Guillemin GJ, Chinetti G, Staels B, Pattou F, Balkau B, Allorge D, Froguel P, Poulain-Godefroy O. The kynurenine pathway is activated in human obesity and shifted toward kynurenine monooxygenase activation. Obesity (Silver Spring) 2015; 23:2066-2074. [PMID: 26347385 DOI: 10.1002/oby.21199] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/20/2015] [Accepted: 06/01/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study characterized the kynurenine pathway (KP) in human obesity by evaluating circulating levels of kynurenines and the expression of KP enzymes in adipose tissue. METHODS Tryptophan and KP metabolite levels were measured in serum of individuals from the D.E.S.I.R. cohort (case-cohort study: 212 diabetic, 836 randomly sampled) and in women with obesity, diabetic or normoglycemic, from the ABOS cohort (n = 100). KP enzyme gene expressions were analyzed in omental and subcutaneous adipose tissue of women from the ABOS cohort, in human primary adipocytes and in monocyte-derived macrophages. RESULTS In the D.E.S.I.R. cohort, kynurenine levels were positively associated with body mass index (BMI) (P = 4.68 × 10(-19) ) and with a higher HOMA2-IR insulin resistance index (P = 6.23 × 10(-4) ). The levels of kynurenine, kynurenic acid, and quinolinic acid were associated with higher BMI (P < 0.05). The expression of several KP enzyme genes (indoleamine 2,3-dioxygenase 1 [IDO1], kynureninase [KYNU], kynurenine 3-monooxygenase [KMO], and kynurenine aminotransferase III [CCBL2]) was increased in the omental adipose tissue of women with obesity compared to lean (P < 0.05), and their expression was induced by proinflammatory cytokines in human primary adipocytes (P < 0.05), except for KMO that is not expressed in these cells. The expressions of IDO1, KYNU, KMO, and CCBL2 were higher in proinflammatory than in anti-inflammatory macrophages (P < 0.05). CONCLUSIONS In the context of obesity, the presence of macrophages in adipose tissue may contribute to diverting KP toward KMO activation.
Collapse
Affiliation(s)
- Marie Favennec
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
| | - Benjamin Hennart
- University of Lille, Lille, France
- CHRU De Lille, Lille, France
- EA4483, Lille, France
| | - Robert Caiazzo
- University of Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
- INSERM UMR 1190, Lille, France
| | - Audrey Leloire
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Loïc Yengo
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Marie Verbanck
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | | | | | - Marie Pigeyre
- University of Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
- INSERM UMR 1190, Lille, France
| | | | - Gilles J Guillemin
- Neuroinflammation Group, Macquarie University, Sydney, New South Wales, Australia
| | - Giulia Chinetti
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- INSERM UMR 1011, Lille, France
| | - Bart Staels
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- INSERM UMR 1011, Lille, France
| | - François Pattou
- University of Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
- INSERM UMR 1190, Lille, France
| | | | - Delphine Allorge
- University of Lille, Lille, France
- CHRU De Lille, Lille, France
- EA4483, Lille, France
| | - Philippe Froguel
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
- CHRU De Lille, Lille, France
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, UK
| | - Odile Poulain-Godefroy
- CNRS UMR 8199, Lille, France
- University of Lille, Lille, France
- Institut Pasteur De Lille, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| |
Collapse
|
125
|
Schultze JL, Schmieder A, Goerdt S. Macrophage activation in human diseases. Semin Immunol 2015; 27:249-56. [PMID: 26303100 DOI: 10.1016/j.smim.2015.07.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 12/24/2022]
Abstract
It is becoming increasingly accepted that macrophages play a crucial role in many diseases associated with chronic inflammation, including atherosclerosis, obesity, diabetes, cancer, skin diseases, and even neurodegenerative diseases. It is therefore not surprising that macrophages in human diseases have gained significant interest during the last years. Molecular analysis combined with more sophisticated murine disease models and the application of genome-wide technologies has resulted in a much better understanding of the role of macrophages in human disease. We highlight important gain of knowledge during the last years for tumor-associated macrophages, and for macrophages in atherosclerosis, obesity and wound healing. Albeit these exciting findings certainly pave the way to novel diagnostics and therapeutics, several hurdles still need to be overcome. We propose a general outline for future research and development in disease-related macrophage biology based on integrating (1) genome-wide technologies, (2) direct human sampling, and (3) a dedicated use of in vivo model systems.
Collapse
Affiliation(s)
- Joachim L Schultze
- Genomics & Immunoregulation, LIMES-Institute, University of Bonn, Carl-Troll-Str. 31, D-53115 Bonn, Germany.
| | - Astrid Schmieder
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - S Goerdt
- Department of Dermatology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
126
|
Smitka K, Marešová D. Adipose Tissue as an Endocrine Organ: An Update on Pro-inflammatory and Anti-inflammatory Microenvironment. Prague Med Rep 2015; 116:87-111. [PMID: 26093665 DOI: 10.14712/23362936.2015.49] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Adipose tissue is recognized as an active endocrine organ that produces a number of endocrine substances referred to as "adipokines" including leptin, adiponectin, adipolin, visfatin, omentin, tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), resistin, pigment epithelium-derived factor (PEDF), and progranulin (PGRN) which play an important role in the food intake regulation and significantly influence insulin sensitivity and in some cases directly affect insulin resistance in skeletal muscle, liver, and adipose tissue. The review summarizes current knowledge about adipose tissue-derived hormones and their influence on energy homeostasis regulation. The possible therapeutic potential of these adipokines in the treatment of insulin resistance, endothelial dysfunction, a pro-inflammatory response, obesity, eating disorders, progression of atherosclerosis, type 1 diabetes, and type 2 diabetes is discussed.
Collapse
Affiliation(s)
- Kvido Smitka
- Institute of Physiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Dana Marešová
- Institute of Physiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| |
Collapse
|
127
|
Guo J, Liu Z, Sun H, Huang Y, Albrecht E, Zhao R, Yang X. Lipopolysaccharide challenge significantly influences lipid metabolism and proteome of white adipose tissue in growing pigs. Lipids Health Dis 2015; 14:68. [PMID: 26152344 PMCID: PMC4493945 DOI: 10.1186/s12944-015-0067-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 06/26/2015] [Indexed: 12/11/2022] Open
Abstract
Background White adipose tissue is recognized as a highly active organ, which is closely related to a large number of physiological and metabolic processes besides storing triglycerides. However, little is known regarding the response of adipose tissue to acute inflammation. Therefore, in this study we employed growing pigs to investigate the changes of lipid metabolism and proteome in white adipose tissue after lipopolysaccharide (LPS) stimulation as a model for bacterial infection. Methods The expression of lipid metabolism and inflammation related genes was determined by quantitative real-time polymerase chain reaction. Label-free proteomics analysis was used to investigate changes of the protein profile in white adipose tissue and western blot was used to verify changes of selected adipokines. Results The results indicated that LPS significantly increased the expression of toll-like receptor (TLR) 2/4 pathway-related genes and pro-inflammatory factors. Lipid metabolism related genes, including acetyl-CoA carboxylase 1 (ACACA), fatty acid synthase (FASN), stearoyl-CoA desaturase (SCD), uncoupling protein 2 (UCP2), and 11 β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), were down-regulated and the lipolytic enzyme activity was decreased after LPS injection. Proteome analysis revealed 47 distinct proteins with > 2-fold changes. The down-regulation of two proteins (cAMP-dependent protein kinase type II-alpha regulatory subunit and β-tubulin) has been verified by western blot analysis. In addition, the abundance of two adipokines (adiponectin and zinc-α2-glycoprotein) was significantly increased after LPS injection. Conclusion In conclusion, LPS challenge can cause acute inflammation in white adipose tissue. Concurrently, lipid metabolism was significantly suppressed and the abundance of several proteins changed in white adipose tissue. The results provide new clues to understand the adipose dysfunction during inflammation. Electronic supplementary material The online version of this article (doi:10.1186/s12944-015-0067-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jun Guo
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, PR China.
| | - Zhiqing Liu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, PR China.
| | - Hailin Sun
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, PR China.
| | - Yanping Huang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, PR China.
| | - Elke Albrecht
- Institute of Muscle Biology and Growth, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany.
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, PR China.
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
128
|
Ultra-structural morphology of long-term cultivated white adipose tissue-derived stem cells. Cell Tissue Bank 2015; 16:639-47. [PMID: 26093679 DOI: 10.1007/s10561-015-9513-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/29/2015] [Indexed: 12/11/2022]
Abstract
White adipose tissue was long perceived as a passive lipid storage depot but it is now considered as an active and important endocrine organ. It also harbours not only adipocytes and vascular cells but also a wide array of immunologically active cells, including macrophages and lymphocytes, which may induce obesity-related inflammation. Recently, adipose tissue has been reported as a source of adult mesenchymal stem cells with wide use in regenerative medicine and tissue engineering. Their relatively non-complicated procurement and collection (often performed as liposuction during aesthetic surgery) and grand plasticity support this idea even more. We focused our research on exploring the issues of isolation and long-term cultivation of mesenchymal stem cells obtained from adipose tissue. Ultra-structural morphology of the cells cultivated in vitro has been studied and analysed in several cultivation time periods and following serial passages--up to 30 passages. In the first passages they had ultra-structural characteristics of cells with high proteosynthetic activity. Within the cytoplasm, big number of small lipid droplets and between them, sparsely placed, small and inconspicuous, electron-dense, lamellar bodies, which resembled myelin figures were observed. The cells from the later passages contained high number of lamellar electron-dense structures, which filled out almost the entire cytoplasm. In between, mitochondria were often found. These bodies were sometimes small and resembled myelin figures, but several of them reached huge dimensions (more than 1 µm) and their lamellar structure was not distinguishable. We did not have an answer to the question about their function, but they probably represented the evidence of active metabolism of lipids present in the cytoplasm of these cells or represented residual bodies, which arise after the breakdown of cellular organelles, notably mitochondria during long-term cultivation.
Collapse
|
129
|
Wakana N, Irie D, Kikai M, Terada K, Yamamoto K, Kawahito H, Kato T, Ogata T, Ueyama T, Matoba S, Yamada H. Maternal High-Fat Diet Exaggerates Atherosclerosis in Adult Offspring by Augmenting Periaortic Adipose Tissue-Specific Proinflammatory Response. Arterioscler Thromb Vasc Biol 2015; 35:558-69. [DOI: 10.1161/atvbaha.114.305122] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objective—
Maternal obesity elicits offspring’s metabolic disorders via developmental modifications of visceral adipose tissue; however, its effect on atherogenesis remains undefined. Perivascular adipose tissue has recently been implicated in vascular remodeling and vasoreactivity. We hypothesize that developmental modifications of perivascular adipose tissue by maternal high-fat diet (HFD) exposure promotes atherosclerosis in adult offspring.
Approach and Results—
Eight-week-old female apolipoprotein E-deficient mice were fed an HFD or normal diet (ND) during gestation and lactation. Offspring were fed a high-cholesterol diet from 8 weeks of age. Twenty-week-old male offspring of HFD-fed dams (O-HFD) showed a 2.1-fold increase in atherosclerotic lesion of the entire aorta compared with those of ND-fed dams (O-ND). Although mRNA expressions of interleukin-6, tumor necrosis factor, and monocyte chemotactic protein-1 and accumulation of macrophages in epididymal white adipose tissue were less in O-HFD than in O-ND, thoracic periaortic adipose tissue (tPAT) showed an exaggerated inflammatory response in O-HFD. Intra-abdominal transplantation of tPAT from 8-week-old O-HFD alongside the distal abdominal aorta exaggerated atherosclerosis development of the infrarenal aorta in recipient apolipoprotein E-deficient mice compared with tPAT from O-ND (210%,
P
<0.01). Although macrophage accumulation was rarely detected in tPAT of 8-week-old offspring, mRNA expression and protein levels of macrophage colony–stimulating factor were markedly elevated in O-HFD (2.3-fold, 3.3-fold, respectively,
P
<0.05), suggesting that increased macrophage colony–stimulating factor expression contributes to the augmented accumulation of macrophages, followed by the enhanced proinflammatory response.
Conclusions—
Our findings demonstrate that maternal HFD exaggerates atherosclerosis development in offspring by augmenting tPAT-specific inflammatory response proceeded by an increased expression of macrophage colony–stimulating factor.
Collapse
Affiliation(s)
- Noriyuki Wakana
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Irie
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masakazu Kikai
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kensuke Terada
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keita Yamamoto
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Kawahito
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taku Kato
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takehiro Ogata
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomomi Ueyama
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Yamada
- From the Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
130
|
Carreras A, Zhang SXL, Almendros I, Wang Y, Peris E, Qiao Z, Gozal D. Resveratrol attenuates intermittent hypoxia-induced macrophage migration to visceral white adipose tissue and insulin resistance in male mice. Endocrinology 2015; 156:437-43. [PMID: 25406018 PMCID: PMC5393321 DOI: 10.1210/en.2014-1706] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Chronic intermittent hypoxia during sleep (IH), as occurs in sleep apnea, promotes systemic insulin resistance. Resveratrol (Resv) has been reported to ameliorate high-fat diet-induced obesity, inflammation, and insulin resistance. To examine the effect of Resv on IH-induced metabolic dysfunction, male mice were subjected to IH or room air conditions for 8 weeks and treated with either Resv or vehicle (Veh). Fasting plasma levels of glucose, insulin, and leptin were obtained, homeostatic model assessment of insulin resistance index levels were calculated, and insulin sensitivity tests (phosphorylated AKT [also known as protein kinase B]/total AKT) were performed in 2 visceral white adipose tissue (VWAT) depots (epididymal [Epi] and mesenteric [Mes]) along with flow cytometry assessments for VWAT macrophages and phenotypes (M1 and M2). IH-Veh and IH-Resv mice showed initial reductions in food intake with later recovery, with resultant lower body weights after 8 weeks but with IH-Resv showing better increases in body weight vs IH-Veh. IH-Veh and IH-Resv mice exhibited lower fasting glucose levels, but only IH-Veh had increased homeostatic model assessment of insulin resistance index vs all 3 other groups. Leptin levels were preserved in IH-Veh but were significantly lower in IH-Resv. Reduced VWAT phosphorylated-AKT/AKT responses to insulin emerged in both Mes and Epi in IH-Veh but normalized in IH-Resv. Increases total macrophage counts and in M1 to M2 ratios occurred in IH-Veh Mes and Epi compared all other 3 groups. Thus, Resv ameliorates food intake and weight gain during IH exposures and markedly attenuates VWAT inflammation and insulin resistance, thereby providing a potentially useful adjunctive therapy for metabolic morbidity in the context of sleep apnea.
Collapse
Affiliation(s)
- Alba Carreras
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois 60637
| | | | | | | | | | | | | |
Collapse
|
131
|
Castoldi A, Naffah de Souza C, Câmara NOS, Moraes-Vieira PM. The Macrophage Switch in Obesity Development. Front Immunol 2015; 6:637. [PMID: 26779183 PMCID: PMC4700258 DOI: 10.3389/fimmu.2015.00637] [Citation(s) in RCA: 294] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/04/2015] [Indexed: 12/11/2022] Open
Abstract
Immune cell infiltration in (white) adipose tissue (AT) during obesity is associated with the development of insulin resistance. In AT, the main population of leukocytes are macrophages. Macrophages can be classified into two major populations: M1, classically activated macrophages, and M2, alternatively activated macrophages, although recent studies have identified a broad range of macrophage subsets. During obesity, AT M1 macrophage numbers increase and correlate with AT inflammation and insulin resistance. Upon activation, pro-inflammatory M1 macrophages induce aerobic glycolysis. By contrast, in lean humans and mice, the number of M2 macrophages predominates. M2 macrophages secrete anti-inflammatory cytokines and utilize oxidative metabolism to maintain AT homeostasis. Here, we review the immunologic and metabolic functions of AT macrophages and their different facets in obesity and the metabolic syndrome.
Collapse
Affiliation(s)
- Angela Castoldi
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Cristiane Naffah de Souza
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
- Division of Nephrology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
- Laboratory of Renal Physiology (LIM 16), Department of Medicine, University of São Paulo, São Paulo, Brazil
| | - Pedro M. Moraes-Vieira
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- *Correspondence: Pedro M. Moraes-Vieira,
| |
Collapse
|