101
|
M1 macrophage subtypes activation and adipocyte dysfunction worsen during prolonged consumption of a fructose-rich diet. J Nutr Biochem 2018; 61:173-182. [PMID: 30245336 DOI: 10.1016/j.jnutbio.2018.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/12/2018] [Accepted: 08/15/2018] [Indexed: 12/12/2022]
Abstract
Fructose-rich diet (FRD) has been associated with obesity development, which is characterized by adipocytes hypertrophy and chronic low-grade inflammation. Interaction of adipocytes and immune cells plays a key role in adipose tissue (AT) alterations in obesity. We assessed the metabolic and immune impairments in AT in a murine obesity model induced by FRD at different periods. Adult Swiss mice were divided into groups of 6 and 10 weeks of fructose (FRD 6wk, FRD 10wk) or water intake (CTR 6wk, CTR 10wk). FRD induced increased in body weight, epidydimal AT mass, and plasmatic and liver Tg, and impaired insulin sensitivity. Also, hypertrophic adipocytes from FRD 6wk-10wk mice showed higher IL-6 when stimulated with LPS and leptin secretion. Several of these alterations worsened in FRD 10wk. Regarding AT inflammation, FRD mice have increased TNFα, IL-6 and IL1β, and decrease in IL-10 and CD206 mRNA levels. Using CD11b, LY6C, CD11c and CD206 as macrophages markers, we identified for first time in AT M1 (M1a: Ly6C+/-CD11c+CD206- and M1b: Ly6C+/-CD11c+CD206+) and M2 subtypes (Ly6C+/-CD11c-CD206+). M1a phenotype increased from 6 weeks onward, while Ly6C+/- M1b phenotype increased only after 10 weeks. Finally, co-culture of RAW264.7 (monocytes cell line) and CTR or FRD adipocytes showed that FRD 10wk adipocytes increased IL-6 expression in non- or LPS-stimulated monocytes. Our results showed that AT dysfunction got worse as the period of fructose consumption was longer. Inflammatory macrophage subtypes increased depending on the period of FRD intake, and hypertrophic adipocytes were able to create an environment that favored M1 phenotype in vitro.
Collapse
|
102
|
Jaisinghani N, Dawa S, Singh K, Nandy A, Menon D, Bhandari PD, Khare G, Tyagi A, Gandotra S. Necrosis Driven Triglyceride Synthesis Primes Macrophages for Inflammation During Mycobacterium tuberculosis Infection. Front Immunol 2018; 9:1490. [PMID: 30018616 PMCID: PMC6037689 DOI: 10.3389/fimmu.2018.01490] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/15/2018] [Indexed: 01/01/2023] Open
Abstract
Pulmonary tuberculosis (TB) exhibits granulomatous inflammation, a site of controlling bacterial dissemination at the cost of host tissue damage. Intrigued by the granuloma type-dependent expression of inflammatory markers in TB, we sought to investigate underlying metabolic changes that drive amplification of inflammation in TB. Here, we show an association of higher inflammation in necrotic granulomas with the presence of triglyceride (TG)-rich foamy macrophages. The conspicuous absence of these macrophages in solid granulomas identified a link between the ensuing pathology and the metabolic programming of foamy macrophages. Consistent with in vivo findings, in vitro infection of macrophages with Mycobacterium tuberculosis (Mtb) led to increase in TG synthesis only under conditions of ~60% necrosis. Genetic and pharmacologic intervention that reduced necrosis prevented this bystander response. We further demonstrate that necrosis independent of Mtb also elicits the same bystander response in human macrophages. We identified a role for the human enzyme involved in TG synthesis, diacylglycerol O-acyltransferase (DGAT1), in this phenomenon. The increased TG levels in necrosis-associated foamy macrophages promoted the pro-inflammatory state of macrophages to infection while silencing expression of diacylglycerol O-acyltransferase (DGAT1) suppressed expression of pro-inflammatory genes. Our data thus invoke a role for storage lipids in the heightened host inflammatory response during infection-associated necrosis. Our data provide a functional role to macrophage lipid droplets in host defense and open new avenues for developing host-directed therapies against TB.
Collapse
Affiliation(s)
- Neetika Jaisinghani
- Chemical and Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Stanzin Dawa
- Chemical and Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Kaurab Singh
- Chemical and Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Ananya Nandy
- Chemical and Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Dilip Menon
- Chemical and Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Purva Deepak Bhandari
- Chemical and Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Garima Khare
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Anil Tyagi
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India.,Guru Gobind Singh Indraprastha University, New Delhi, India
| | - Sheetal Gandotra
- Chemical and Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| |
Collapse
|
103
|
Zhu X, Tu Y, Chen H, Jackson AO, Patel V, Yin K. Micro-environment and intracellular metabolism modulation of adipose tissue macrophage polarization in relation to chronic inflammatory diseases. Diabetes Metab Res Rev 2018; 34:e2993. [PMID: 29475214 DOI: 10.1002/dmrr.2993] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 02/08/2018] [Accepted: 02/11/2018] [Indexed: 12/13/2022]
Abstract
The accumulation and pro-inflammatory polarization of immune cells, mainly macrophages, in adipose tissue (AT) are considered crucial factors for obesity-induced chronic inflammatory diseases. In this review, we highlighted the role of adipose tissue macrophage (ATM) polarization on AT function in the obese state and the effect of the micro-environment and intracellular metabolism on the dynamic switch of ATMs into their pro-inflammatory or anti-inflammatory phenotypes, which may have distinct influences on obesity-related chronic inflammatory diseases. Obesity-associated metabolic dysfunctions, including those of glucose, fatty acid, cholesterol, and other nutrient substrates such as vitamin D and iron in AT, promote the pro-inflammatory polarization of ATMs and AT inflammation via regulating the interaction between ATMs and adipocytes and intracellular metabolic pathways, including glycolysis, fatty acid oxidation, and reverse cholesterol transportation. Focusing on the regulation of ATM metabolism will provide a novel target for the treatment of obesity-related chronic inflammatory diseases, including insulin resistance, cardiovascular diseases, and cancers.
Collapse
Affiliation(s)
- Xiao Zhu
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, University of South China, Hengyang, China
| | - Yixuan Tu
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
| | - Hainan Chen
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, University of South China, Hengyang, China
| | - Ampadu O Jackson
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
| | - Vaibhav Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Kai Yin
- Research Laboratory of Translational Medicine, Medical School, University of South China, Hengyang, China
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, University of South China, Hengyang, China
| |
Collapse
|
104
|
Cardenas-Perez RE, Fuentes-Mera L, de la Garza AL, Torre-Villalvazo I, Reyes-Castro LA, Rodriguez-Rocha H, Garcia-Garcia A, Corona-Castillo JC, Tovar AR, Zambrano E, Ortiz-Lopez R, Saville J, Fuller M, Camacho A. Maternal overnutrition by hypercaloric diets programs hypothalamic mitochondrial fusion and metabolic dysfunction in rat male offspring. Nutr Metab (Lond) 2018; 15:38. [PMID: 29991958 PMCID: PMC5987395 DOI: 10.1186/s12986-018-0279-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Background Maternal overnutrition including pre-pregnancy, pregnancy and lactation promotes a lipotoxic insult leading to metabolic dysfunction in offspring. Diet-induced obesity models (DIO) show that changes in hypothalamic mitochondria fusion and fission dynamics modulate metabolic dysfunction. Using three selective diet formula including a High fat diet (HFD), Cafeteria (CAF) and High Sugar Diet (HSD), we hypothesized that maternal diets exposure program leads to selective changes in hypothalamic mitochondria fusion and fission dynamics in male offspring leading to metabolic dysfunction which is exacerbated by a second exposure after weaning. Methods We exposed female Wistar rats to nutritional programming including Chow, HFD, CAF, or HSD for 9 weeks (pre-mating, mating, pregnancy and lactation) or to the same diets to offspring after weaning. We determined body weight, food intake and metabolic parameters in the offspring from 21 to 60 days old. Hypothalamus was dissected at 60 days old to determine mitochondria-ER interaction markers by mRNA expression and western blot and morphology by transmission electron microscopy (TEM). Mitochondrial-ER function was analyzed by confocal microscopy using hypothalamic cell line mHypoA-CLU192. Results Maternal programming by HFD and CAF leads to failure in glucose, leptin and insulin sensitivity and fat accumulation. Additionally, HFD and CAF programming promote mitochondrial fusion by increasing the expression of MFN2 and decreasing DRP1, respectively. Further, TEM analysis confirms that CAF exposure after programing leads to an increase in mitochondria fusion and enhanced mitochondrial-ER interaction, which partially correlates with metabolic dysfunction and fat accumulation in the HFD and CAF groups. Finally, we identified that lipotoxic palmitic acid stimulus in hypothalamic cells increases Ca2+ overload into mitochondria matrix leading to mitochondrial dysfunction. Conclusions We concluded that maternal programming by HFD induces hypothalamic mitochondria fusion, metabolic dysfunction and fat accumulation in male offspring, which is exacerbated by HFD or CAF exposure after weaning, potentially due to mitochondria calcium overflux.
Collapse
Affiliation(s)
- Robbi E Cardenas-Perez
- 1Departmento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico.,2Unidad de Neurometabolismo, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autónoma de Nuevo Leon, Monterrey, Mexico
| | - Lizeth Fuentes-Mera
- 1Departmento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Ana Laura de la Garza
- 3Centro de Investigacion en Nutricion y Salud Publica, Facultad de Salud Publica y Nutricion, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Ivan Torre-Villalvazo
- 4Departamento Fisiología de la Nutrición, Instituto Nacional de Ciencias Medicas y Nutrición, Mexico City, Mexico
| | - Luis A Reyes-Castro
- 5Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Medicas y Nutrición Salvador Zubiran, México City, Mexico
| | - Humberto Rodriguez-Rocha
- 6Departmento de Histología, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Aracely Garcia-Garcia
- 6Departmento de Histología, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | | | - Armando R Tovar
- 4Departamento Fisiología de la Nutrición, Instituto Nacional de Ciencias Medicas y Nutrición, Mexico City, Mexico
| | - Elena Zambrano
- 5Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Medicas y Nutrición Salvador Zubiran, México City, Mexico
| | - Rocio Ortiz-Lopez
- 8Escuela de Medicina y Ciencias de la Salud, Instituto Tecnologico de Monterrey, Monterrey, Mexico
| | - Jennifer Saville
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, University of Adelaide, Adelaide, Australia
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, University of Adelaide, Adelaide, Australia
| | - Alberto Camacho
- 1Departmento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico.,2Unidad de Neurometabolismo, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autónoma de Nuevo Leon, Monterrey, Mexico.,10Departamento de Bioquimica y Medicina Molecular. Facultad de Medicina, Universidad Autónoma de Nuevo León, Ave. Francisco I Madero y Dr. Eduardo Aguirre Pequeño s/n. Colonia Mitras Centro, C.P. 64460 Monterrey, Nuevo Leon Mexico
| |
Collapse
|
105
|
Macrophage Populations in Visceral Adipose Tissue from Pregnant Women: Potential Role of Obesity in Maternal Inflammation. Int J Mol Sci 2018; 19:ijms19041074. [PMID: 29617296 PMCID: PMC5979476 DOI: 10.3390/ijms19041074] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/22/2018] [Accepted: 03/26/2018] [Indexed: 12/23/2022] Open
Abstract
Obesity is associated with inflammatory changes and accumulation and phenotype polarization of adipose tissue macrophages (ATMs). Obese pregnant women have alterations in adipose tissue composition, but a detailed description of macrophage population is not available. In this study, we characterized macrophage populations in visceral adipose tissue (VAT) from pregnant women with normal, overweight, and obese pregestational weight. Immunophenotyping of macrophages from VAT biopsies was performed by flow cytometry using CD45 and CD14 as markers of hematopoietic and monocyte linage, respectively, while HLA-DR, CD11c, CD163, and CD206 were used as pro- and anti-inflammatory markers. Adipocyte number and size were evaluated by light microscopy. The results show that pregnant women that were overweight and obese during the pregestational period had adipocyte hypertrophy. Two different macrophage populations in VAT were identified: recruited macrophages (CD45+CD14+), and a novel population lacking CD45, which was considered to be a resident macrophages subset (CD45−CD14+). The number of resident HLA−DRlow/− macrophages showed a negative correlation with body mass index (BMI). Both resident and recruited macrophages from obese women expressed higher CD206 levels. CD11c expression was higher in resident HLA-DR+ macrophages from obese women. A strong correlation between CD206 and CD11c markers and BMI was observed. Our findings show that being overweight and obese in the pregestational period is associated with adipocyte hypertrophy and specific ATMs populations in VAT.
Collapse
|
106
|
Verboven K, Wouters K, Gaens K, Hansen D, Bijnen M, Wetzels S, Stehouwer CD, Goossens GH, Schalkwijk CG, Blaak EE, Jocken JW. Abdominal subcutaneous and visceral adipocyte size, lipolysis and inflammation relate to insulin resistance in male obese humans. Sci Rep 2018; 8:4677. [PMID: 29549282 PMCID: PMC5856747 DOI: 10.1038/s41598-018-22962-x] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 03/05/2018] [Indexed: 12/16/2022] Open
Abstract
Obesity is associated with a disturbed adipose tissue (AT) function characterized by adipocyte hypertrophy, an impaired lipolysis and pro-inflammatory phenotype, which contributes to insulin resistance (IR). We investigated whether AT phenotype in different AT depots of obese individuals with and without type 2 diabetes mellitus (T2DM) is associated with whole-body IR. Subcutaneous (SC) and visceral (V) AT biopsies from 18 lean, 17 obese and 8 obese T2DM men were collected. AT phenotype was characterized by ex vivo measurement of basal and stimulated lipolysis (mature adipocytes), adipocyte size distribution (AT tissue sections) and AT immune cells (flow cytometry). In VAT, mean adipocyte size, CD45+ leukocytes and M1 macrophages were significantly increased in both obese groups compared to lean individuals. In SCAT, despite adipocyte hypertrophy, no significant differences in immune cell populations between groups were found. In SCAT, multiple linear regression analysis showed that none of the AT phenotype markers independently contributed to HOMA-IR while in VAT, mean adipocyte size was significantly related to HOMA-IR. In conclusion, beside adipocyte hypertrophy in VAT, M1 macrophage- or B-cell-mediated inflammation, may contribute to IR, while inflammation in hypertrophic SCAT does not seem to play a major role in IR.
Collapse
Affiliation(s)
- K Verboven
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands. .,Rehabilitation Research Center, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium.
| | - K Wouters
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - K Gaens
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - D Hansen
- Rehabilitation Research Center, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium.,Heart Centre Hasselt, Jessa Hospital, Hasselt, Belgium
| | - M Bijnen
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - S Wetzels
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - C D Stehouwer
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - G H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - C G Schalkwijk
- Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - E E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - J W Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
107
|
Song H, Lim DY, Jung JI, Cho HJ, Park SY, Kwon GT, Kang YH, Lee KW, Choi MS, Park JHY. Dietary oleuropein inhibits tumor angiogenesis and lymphangiogenesis in the B16F10 melanoma allograft model: a mechanism for the suppression of high-fat diet-induced solid tumor growth and lymph node metastasis. Oncotarget 2018; 8:32027-32042. [PMID: 28410190 PMCID: PMC5458266 DOI: 10.18632/oncotarget.16757] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/20/2017] [Indexed: 12/20/2022] Open
Abstract
Previously, we reported that high-fat-diet (HFD)-induced obesity stimulates melanoma progression in the B16F10 allograft model. In this study, we examined whether oleuropein (OL), the most abundant phenolic compound in olives, inhibits HFD-induced melanoma progression. Four-week-old male C57BL/6N mice were fed a HFD-diet with or without OL. After 16 weeks of feeding, B16F10-luc cells were subcutaneously injected and the primary tumor was resected 3 weeks later. OL suppressed HFD-induced solid tumor growth. In the tumor tissues, OL reduced HFD-induced expression of angiogenesis (CD31, VE-cadherin, VEGF-A, and VEGFR2), lymphangiogenesis (LYVE-1, VEGF-C, VEGF-D, and VEGFR3), and hypoxia (HIF-1α and GLUT-1) markers as well as HFD-induced increases in lipid vacuoles and M2 macrophages (MΦs). All animals were euthanized 2.5 weeks after tumor resection. OL suppressed HFD-induced increases in lymph node (LN) metastasis; expression of VEGF-A, VEGF-C, and VEGF-D in the LN; and M2-MΦs and the size of adipocytes in adipose tissues surrounding LNs. Co-culture results revealed that the crosstalk between B16F10s, M2-MΦs, and differentiated 3T3-L1 cells under hypoxic conditions increased the secretion of VEGF-A and -D, which stimulated tube formation and migration of endothelial cells (HUVECs) and lymphatic endothelial cells (LEC), respectively. Additionally, OL directly inhibited the differentiation of 3T3-L1 preadipocytes and tube formation by HUVECs and LECs. The overall results indicated that dietary OL inhibits lipid and M2-MΦ accumulation in HFD-fed mice, which contributes to decreases in VEGF secretion, thereby leading to inhibition of angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Hyerim Song
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Do Young Lim
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Jae In Jung
- Division of Bio-Imaging, Chuncheon Center, Korea Basic Science Institute, Chuncheon 24341, Republic of Korea
| | - Han Jin Cho
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.,WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea
| | - So Young Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea
| | - Gyoo Taik Kwon
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea.,Berry and Biofood Research Institute, Jeonbuk 56417, Republic of Korea
| | - Young-Hee Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Ki Won Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea.,Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Myung-Sook Choi
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jung Han Yoon Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea.,Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
108
|
Abstract
Adipose tissue is a special tissue environment due to its high lipid content. Adipose tissue macrophages (ATMs) help maintain adipose tissue homeostasis in steady state by clearing dead adipocytes. However, adipose tissue changes drastically during obesity, resulting in a state of chronic low grade inflammation and a shift in the adipose immune landscape. In this review we will discuss how these changes influence the polarization of ATMs.
Collapse
Affiliation(s)
- Leen Catrysse
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.
| |
Collapse
|
109
|
Muir LA, Kiridena S, Griffin C, DelProposto JB, Geletka L, Martinez-Santibañez G, Zamarron BF, Lucas H, Singer K, O' Rourke RW, Lumeng CN. Frontline Science: Rapid adipose tissue expansion triggers unique proliferation and lipid accumulation profiles in adipose tissue macrophages. J Leukoc Biol 2018; 103:615-628. [PMID: 29493813 DOI: 10.1002/jlb.3hi1017-422r] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/04/2018] [Accepted: 01/10/2018] [Indexed: 12/22/2022] Open
Abstract
Obesity-related changes in adipose tissue leukocytes, in particular adipose tissue macrophages (ATMs) and dendritic cells (ATDCs), are implicated in metabolic inflammation, insulin resistance, and altered regulation of adipocyte function. We evaluated stromal cell and white adipose tissue (WAT) expansion dynamics with high fat diet (HFD) feeding for 3-56 days, quantifying ATMs, ATDCs, endothelial cells (ECs), and preadipocytes (PAs) in visceral epididymal WAT and subcutaneous inguinal WAT. To better understand mechanisms of the early response to obesity, we evaluated ATM proliferation and lipid accumulation. ATMs, ATDCs, and ECs increased with rapid WAT expansion, with ATMs derived primarily from a CCR2-independent resident population. WAT expansion stimulated proliferation in resident ATMs and ECs, but not CD11c+ ATMs or ATDCs. ATM proliferation was unperturbed in Csf2- and Rag1-deficient mice with WAT expansion. Additionally, ATM apoptosis decreased with WAT expansion, and proliferation and apoptosis reverted to baseline with weight loss. Adipocytes reached maximal hypertrophy at 28 days of HFD, coinciding with a plateau in resident ATM accumulation and the appearance of lipid-laden CD11c+ ATMs in visceral epididymal WAT. ATM increases were proportional to tissue expansion and adipocyte hypertrophy, supporting adipocyte-mediated regulation of resident ATMs. The appearance of lipid-laden CD11c+ ATMs at peak adipocyte size supports a role in responding to ectopic lipid accumulation within adipose tissue. In contrast, ATDCs increase independently of proliferation and may be derived from circulating precursors. These changes precede and establish the setting in which large-scale adipose tissue infiltration of CD11c+ ATMs, inflammation, and adipose tissue dysfunction contributes to insulin resistance.
Collapse
Affiliation(s)
- Lindsey A Muir
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Samadhi Kiridena
- College of Literature Science and the Arts, University of Michigan, Ann Arbor, Michigan, USA
| | - Cameron Griffin
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jennifer B DelProposto
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lynn Geletka
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gabriel Martinez-Santibañez
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Brian F Zamarron
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Hannah Lucas
- College of Literature Science and the Arts, University of Michigan, Ann Arbor, Michigan, USA
| | - Kanakadurga Singer
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Robert W O' Rourke
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Surgery, Ann Arbor Veterans Administration Hospital, Ann Arbor, Michigan, USA
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
110
|
Ivanov S, Merlin J, Lee MKS, Murphy AJ, Guinamard RR. Biology and function of adipose tissue macrophages, dendritic cells and B cells. Atherosclerosis 2018; 271:102-110. [PMID: 29482037 DOI: 10.1016/j.atherosclerosis.2018.01.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/22/2017] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
The increasing incidence of obesity and its socio-economical impact is a global health issue due to its associated co-morbidities, namely diabetes and cardiovascular disease [1-5]. Obesity is characterized by an increase in adipose tissue, which promotes the recruitment of immune cells resulting in low-grade inflammation and dysfunctional metabolism. Macrophages are the most abundant immune cells in the adipose tissue of mice and humans. The adipose tissue also contains other myeloid cells (dendritic cells (DC) and neutrophils) and to a lesser extent lymphocyte populations, including T cells, B cells, Natural Killer (NK) and Natural Killer T (NKT) cells. While the majority of studies have linked adipose tissue macrophages (ATM) to the development of low-grade inflammation and co-morbidities associated with obesity, emerging evidence suggests for a role of other immune cells within the adipose tissue that may act in part by supporting macrophage homeostasis. In this review, we summarize the current knowledge of the functions ATMs, DCs and B cells possess during steady-state and obesity.
Collapse
Affiliation(s)
- Stoyan Ivanov
- INSERM U1065, Mediterranean Center of Molecular Medicine, University of Nice Sophia-Antipolis, Faculty of Medicine, Nice, France.
| | - Johanna Merlin
- INSERM U1065, Mediterranean Center of Molecular Medicine, University of Nice Sophia-Antipolis, Faculty of Medicine, Nice, France
| | - Man Kit Sam Lee
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Rodolphe R Guinamard
- INSERM U1065, Mediterranean Center of Molecular Medicine, University of Nice Sophia-Antipolis, Faculty of Medicine, Nice, France.
| |
Collapse
|
111
|
Kim SM, Neuendorff N, Alaniz RC, Sun Y, Chapkin RS, Earnest DJ. Shift work cycle-induced alterations of circadian rhythms potentiate the effects of high-fat diet on inflammation and metabolism. FASEB J 2018; 32:3085-3095. [PMID: 29405095 DOI: 10.1096/fj.201700784r] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Based on genetic models with mutation or deletion of core clock genes, circadian disruption has been implicated in the pathophysiology of metabolic disorders. Thus, we examined whether circadian desynchronization in response to shift work-type schedules is sufficient to compromise metabolic homeostasis and whether inflammatory mediators provide a key link in the mechanism by which alterations of circadian timekeeping contribute to diet-induced metabolic dysregulation. In high-fat diet (HFD)-fed mice, exposure to chronic shifts of the light-dark cycle (12 h advance every 5 d): 1) disrupts photoentrainment of circadian behavior and modulates the period of spleen and macrophage clock gene rhythms; 2) potentiates HFD-induced adipose tissue infiltration and activation of proinflammatory M1 macrophages; 3) amplifies macrophage proinflammatory cytokine expression in adipose tissue and bone marrow-derived macrophages; and 4) exacerbates diet-induced increases in body weight, insulin resistance, and glucose intolerance in the absence of changes in total daily food intake. Thus, complete disruption of circadian rhythmicity or clock gene function as transcription factors is not requisite to the link between circadian and metabolic phenotypes. These findings suggest that macrophage proinflammatory activation and inflammatory signaling are key processes in the physiologic cascade by which dysregulation of circadian rhythmicity exacerbates diet-induced systemic insulin resistance and glucose intolerance.-Kim, S.-M., Neuendorff, N., Alaniz, R. C., Sun, Y., Chapkin, R. S., Earnest, D. J. Shift work cycle-induced alterations of circadian rhythms potentiate the effects of high-fat diet on inflammation and metabolism.
Collapse
Affiliation(s)
- Sam-Moon Kim
- Department of Biology, Texas A&M University, College Station, Texas, USA.,Center for Biological Clocks Research, Texas A&M University, College Station, Texas, USA
| | - Nichole Neuendorff
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Robert C Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Yuxiang Sun
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA; and
| | - Robert S Chapkin
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA.,Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA; and.,Program in Integrative Nutrition and Complex Diseases, Center for Translational Environmental Health Research, Texas A&M University, College Station, Texas, USA
| | - David J Earnest
- Department of Biology, Texas A&M University, College Station, Texas, USA.,Center for Biological Clocks Research, Texas A&M University, College Station, Texas, USA.,Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| |
Collapse
|
112
|
Jing Y, Wu F, Li D, Yang L, Li Q, Li R. Metformin improves obesity-associated inflammation by altering macrophages polarization. Mol Cell Endocrinol 2018; 461:256-264. [PMID: 28935544 DOI: 10.1016/j.mce.2017.09.025] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 08/24/2017] [Accepted: 09/16/2017] [Indexed: 11/20/2022]
Abstract
Obesity is reported to be a chronic low-grade inflammatory state. Adipose tissue macrophages play a key role in obesity-related inflammation. Metformin, the most widely used anti-diabetic drug, has recently been reported to have an effect on inflammation, but the mechanism is poorly understood. This study aims to investigate how metformin works on chronic low-grade inflammation in obesity and whether the mechanism underlying it is associated with macrophage polarization. Metformin was administered for 7 weeks to high fat-fed C57/6J male mice in vivo. Metformin, compound C (an AMPK inhibitor) and AICAR (an AMPK activator) were used for the in vitro intervention. The gene expression of macrophages markers was examined. Pro-inflammatory cytokines IL-6 and TNF-α were tested by ELISA. The macrophage subsets were analyzed by flow cytometry. In vivo, we discovered that metformin not only decreased the serum level of the pro-inflammatory cytokines IL-6 and TNF-α but also lowered the expression of the M1 macrophage markers CD11c and MCP-1 in adipose tissue. In vitro, metformin reduced the secretion of IL-6 and TNF-α in palmitate-stimulated RAW264.7 macrophages, while compound C treatment blocked the effect of metformin. Moreover, treatment with metformin and AICAR decreased the proportion of M1 macrophages and increased the proportion of M2 macrophages, as analyzed by flow cytometry, in palmitate-stimulated BMDMs. In addition, the effect of AICAR on macrophage polarization was stronger than that of metformin. These results suggest that metformin improves low-grade inflammation in obesity and modulates macrophage polarization to an anti-inflammatory, M2 phenotype partly via the activation of AMPK.
Collapse
Affiliation(s)
- Yuanyuan Jing
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fan Wu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Dai Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lei Yang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qi Li
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rong Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
113
|
Kuda O, Rossmeisl M, Kopecky J. Omega-3 fatty acids and adipose tissue biology. Mol Aspects Med 2018; 64:147-160. [PMID: 29329795 DOI: 10.1016/j.mam.2018.01.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 12/16/2022]
Abstract
This review provides evidence for the importance of white and brown adipose tissue (i.e. WAT and BAT) function for the maintenance of healthy metabolic phenotype and its preservation in response to omega-3 polyunsaturated fatty acids (omega-3 PUFA), namely in the context of diseased states linked to aberrant accumulation of body fat, systemic low-grade inflammation, dyslipidemia and insulin resistance. More specifically, the review deals with (i) the concept of immunometabolism, i.e. how adipose-resident immune cells and adipocytes affect each other and define the immune-metabolic interface; and (ii) the characteristic features of "healthy adipocytes" in WAT, which are relatively small fat cells endowed with a high capacity for mitochondrial oxidative phosphorylation, triacylglycerol/fatty acid (TAG/FA) cycling and de novo lipogenesis (DNL). The intrinsic metabolic features of WAT and their flexible regulations, reflecting the presence of "healthy adipocytes", provide beneficial local and systemic effects, including (i) protection against in situ endoplasmic reticulum stress and related inflammatory response during activation of adipocyte lipolysis; (ii) prevention of ectopic fat accumulation and dyslipidemia caused by increased hepatic VLDL synthesis, as well as prevention of lipotoxic damage of insulin signaling in extra-adipose tissues; and also (iii) increased synthesis of anti-inflammatory and insulin-sensitizing lipid mediators with pro-resolving properties, including the branched fatty acid esters of hydroxy fatty acids (FAHFAs), also depending on the activity of DNL in WAT. The "healthy adipocytes" phenotype can be induced in WAT of obese mice in response to various stimuli including dietary omega-3 PUFA, especially when combined with moderate calorie restriction, and possibly also with other life style (e.g. physical activity) or pharmacological (e.g. thiazolidinediones) interventions. While omega-3 PUFA could exert beneficial systemic effects by improving immunometabolism of WAT without a concomitant induction of BAT, it is currently not clear whether the metabolic effects of the combined intervention using omega-3 PUFA and calorie restriction or thiazolidinediones depend also on the activation of BAT function and/or the induction of brite/beige adipocytes in WAT. It remains to be established why omega-3 PUFA intervention in type 2 diabetic subjects does not improve insulin sensitivity and glucose homeostasis despite inducing various anti-inflammatory mediators in WAT, including the recently discovered docosahexaenoyl esters of hydroxy linoleic acid, the lipokines from the FAHFA family, as well as several endocannabinoid-related anti-inflammatory lipids. To answer the question whether and to which extent omega-3 PUFA supplementation could promote the formation of "healthy adipocytes" in WAT of human subjects, namely in the obese insulin-resistant patients, represents a challenging task that is of great importance for the treatment of some serious non-communicable diseases.
Collapse
Affiliation(s)
- Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic
| | - Martin Rossmeisl
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic
| | - Jan Kopecky
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic.
| |
Collapse
|
114
|
Sun G, Yang S, Cao G, Wang Q, Hao J, Wen Q, Li Z, So KF, Liu Z, Zhou S, Zhao Y, Yang H, Zhou L, Yin Z. γδ T cells provide the early source of IFN-γ to aggravate lesions in spinal cord injury. J Exp Med 2017; 215:521-535. [PMID: 29282251 PMCID: PMC5789408 DOI: 10.1084/jem.20170686] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/22/2017] [Accepted: 11/28/2017] [Indexed: 02/05/2023] Open
Abstract
Immune responses and neuroinflammation are critically involved in spinal cord injury (SCI). γδ T cells, a small subset of T cells, regulate the inflammation process in many diseases, yet their function in SCI is still poorly understood. In this paper, we demonstrate that mice deficient in γδ T cells (TCRδ-/- ) showed improved functional recovery after SCI. γδ T cells are detected at the lesion sites within 24 hours after injury and are predominantly of the Vγ4 subtype and express the inflammatory cytokine IFN-γ. Inactivating IFN-γ signaling in macrophages results in a significantly reduced production of proinflammatory cytokines in the cerebrospinal fluid (CSF) of mice with SCIs and improves functional recovery. Furthermore, treatment of SCI with anti-Vγ4 antibodies has a beneficial effect, similar to that obtained with anti-TNF-α. In SCI patients, γδ T cells are detected in the CSF, and most of them are IFN-γ positive. In conclusion, manipulation of γδ T cell functions may be a potential approach for future SCI treatment.
Collapse
Affiliation(s)
- Guodong Sun
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Shuxian Yang
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Guangchao Cao
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Qianghua Wang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Jianlei Hao
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Qiong Wen
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Zhizhong Li
- The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Zonghua Liu
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China.,State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Sufang Zhou
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Yongxiang Zhao
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Hengwen Yang
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China.,State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Libing Zhou
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China .,Co-Innovation Center of Neuroregeneration, Nantong University, Jiangsu, China.,Key Laboratory of Neuroscience, School of Basic Medical Sciences, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhinan Yin
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China .,State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
115
|
Kumari M, Heeren J, Scheja L. Regulation of immunometabolism in adipose tissue. Semin Immunopathol 2017; 40:189-202. [DOI: 10.1007/s00281-017-0668-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/22/2017] [Indexed: 12/14/2022]
|
116
|
Morgan-Bathke M, Harteneck D, Jaeger P, Sondergaard E, Karwoski R, De Ycaza AE, Carranza-Leon BG, Faubion WA, Oliveira AM, Jensen MD. Comparison of Methods for Analyzing Human Adipose Tissue Macrophage Content. Obesity (Silver Spring) 2017; 25:2100-2107. [PMID: 28985040 PMCID: PMC5705319 DOI: 10.1002/oby.22012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 08/18/2017] [Accepted: 08/19/2017] [Indexed: 12/26/2022]
Abstract
OBJECTIVE The relationship between inflammation, obesity, and adverse metabolic conditions is associated with adipose tissue macrophages (ATM). This study compared the measurements of human ATM using flow cytometry, immunohistochemistry (IHC), and real-time polymerase chain reaction (RT-PCR) of ATM markers. METHODS A new software program (AMCounter) was evaluated to help measure ATM using IHC, and this was compared to flow cytometry and RT-PCR. RESULTS IHC had good intraindividual reproducibility for total (CD68), proinflammatory (CD14), and anti-inflammatory (CD206) ATM. The AMCounter improved interreader agreement and was more time efficient. Flow cytometry had acceptable intraindividual reproducibility for the percentage of CD68+ cells that were CD14+ or CD206+ , but not for ATMs per gram of tissue. ATMs per gram of tissue was much greater using IHC than flow cytometry. The flow cytometry and IHC measures of ATM from the same biopsies were not correlated. There were statistically significant correlations between RT-PCR CD68 and IHC CD68, CD14, and CD206 ATMs per 100 adipocytes. Also of interest were statistically significant correlations between RT-PCR CD68 and IHC CD68, CD14, and adipose flow cytometry measures of CD68+ , CD68+ /CD14+ , and CD68+ /CD206+ ATMs per gram of tissue. CONCLUSIONS The AMCounter software helps provide reproducible and efficient measures of IHC ATMs. Flow cytometry, IHC, and RT-PCR measures of adipose inflammation provide somewhat different information.
Collapse
Affiliation(s)
| | - Debra Harteneck
- Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota USA
| | - Philippa Jaeger
- Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota USA
| | - Esben Sondergaard
- Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota USA
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus DENMARK
| | - Ron Karwoski
- Biomedical Imaging Resources, Mayo Clinic, Rochester, Minnesota USA
| | | | | | | | | | - Michael D. Jensen
- Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota USA
- Corresponding Author: Michael D. Jensen, Mayo Clinic, Endocrine Research Unit, 200 1 St SW, Rm 5-194 Joseph, Rochester, MN 55905, 507-255-6515 (tel), 507-255-4828 (fax),
| |
Collapse
|
117
|
Integrated Immunomodulatory Mechanisms through which Long-Chain n-3 Polyunsaturated Fatty Acids Attenuate Obese Adipose Tissue Dysfunction. Nutrients 2017; 9:nu9121289. [PMID: 29186929 PMCID: PMC5748740 DOI: 10.3390/nu9121289] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is a global health concern with rising prevalence that increases the risk of developing other chronic diseases. A causal link connecting overnutrition, the development of obesity and obesity-associated co-morbidities is visceral adipose tissue (AT) dysfunction, characterized by changes in the cellularity of various immune cell populations, altered production of inflammatory adipokines that sustain a chronic state of low-grade inflammation and, ultimately, dysregulated AT metabolic function. Therefore, dietary intervention strategies aimed to halt the progression of obese AT dysfunction through any of the aforementioned processes represent an important active area of research. In this connection, fish oil-derived dietary long-chain n-3 polyunsaturated fatty acids (PUFA) in the form of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been demonstrated to attenuate obese AT dysfunction through multiple mechanisms, ultimately affecting AT immune cellularity and function, adipokine production, and metabolic signaling pathways, all of which will be discussed herein.
Collapse
|
118
|
Alsabeeh N, Chausse B, Kakimoto PA, Kowaltowski AJ, Shirihai O. Cell culture models of fatty acid overload: Problems and solutions. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:143-151. [PMID: 29155055 DOI: 10.1016/j.bbalip.2017.11.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022]
Abstract
High plasma levels of fatty acids occur in a variety of metabolic diseases. Cellular effects of fatty acid overload resulting in negative cellular responses (lipotoxicity) are often studied in vitro, in an attempt to understand mechanisms involved in these diseases. Fatty acids are poorly soluble, and thus usually studied when complexed to albumins such as bovine serum albumin (BSA). The conjugation of fatty acids to albumin requires care pertaining to preparation of the solutions, effective free fatty acid concentrations, use of different fatty acid species, types of BSA, appropriate controls and ensuring cellular fatty acid uptake. This review discusses lipotoxicity models, the potential problems encountered when using these cellular models, as well as practical solutions for difficulties encountered.
Collapse
Affiliation(s)
- Nour Alsabeeh
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA; Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA; Department of Physiology, Faculty of Medicine, Kuwait University, Kuwait
| | - Bruno Chausse
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Pamela A Kakimoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil.
| | - Orian Shirihai
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
119
|
Asperlin Inhibits LPS-Evoked Foam Cell Formation and Prevents Atherosclerosis in ApoE -/- Mice. Mar Drugs 2017; 15:md15110358. [PMID: 29135917 PMCID: PMC5706047 DOI: 10.3390/md15110358] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/02/2017] [Accepted: 11/07/2017] [Indexed: 12/14/2022] Open
Abstract
Asperlin is a marine-derived natural product with antifungal and anti-inflammatory activities in vitro. In the present study, we isolated asperlin from a marine Aspergillus versicolor LZD4403 fungus and investigated its anti-atherosclerotic effects in vitro and in vivo. Asperlin significantly inhibited lipopolysaccharides (LPS)- but not oxidated low-density lipoprotein (oxLDL)-evoked foam cell formation and promoted cholesterol efflux in RAW264.7 macrophages. Supplementation with asperlin also suppressed LPS-elicited production of pro-inflammatory factors in RAW264.7 macrophages, decreased the expression levels of iNOS, IL-1β and TNFα, and increased the expression of IL-10 and IL-4, indicating a remarkable shift in M1/M2 macrophages polarization. In vivo experiments in high-fat diet (HFD)-fed ApoE−/− mice showed that oral administration of asperlin for 12 weeks remarkably suppressed atherosclerotic plaque formation in the aorta, as revealed by the reduced aortic dilatation and decreased atherosclerotic lesion area. Asperlin also decreased serum levels of pro-inflammatory factors but showed little impact on blood lipids in ApoE−/− atherosclerotic mice. These results suggested that asperlin is adequate to prevent atherosclerosis in vivo. It may exert atheroprotective function through suppressing inflammation rather than ameliorating dyslipidemia.
Collapse
|
120
|
Yu X, Tang Y, Liu P, Xiao L, Liu L, Shen R, Deng Q, Yao P. Flaxseed Oil Alleviates Chronic HFD-Induced Insulin Resistance through Remodeling Lipid Homeostasis in Obese Adipose Tissue. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:9635-9646. [PMID: 28988484 DOI: 10.1021/acs.jafc.7b03325] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Emerging evidence suggests that higher circulating long-chain n-3 polyunsaturated fatty acids (n-3PUFA) levels were intimately associated with lower prevalence of obesity and insulin resistance. However, the understanding of bioactivity and potential mechanism of α-linolenic acid-rich flaxseed oil (ALA-FO) against insulin resistance was still limited. This study evaluated the effect of FO on high-fat diet (HFD)-induced insulin resistance in C57BL/6J mice focused on adipose tissue lipolysis. Mice after HFD feeding for 16 weeks (60% fat-derived calories) exhibited systemic insulin resistance, which was greatly attenuated by medium dose of FO (M-FO), paralleling with differential accumulation of ALA and its n-3 derivatives across serum lipid fractions. Moreover, M-FO was sufficient to effectively block the metabolic activation of adipose tissue macrophages (ATMs), thereby improving adipose tissue insulin signaling. Importantly, suppression of hypoxia-inducible factors HIF-1α and HIF-2α were involved in FO-mediated modulation of adipose tissue lipolysis, accompanied by specific reconstitution of n-3PUFA within adipose tissue lipid fractions.
Collapse
Affiliation(s)
- Xiao Yu
- College of Food and Biological Engineering, Zhengzhou University of Light Industry , Zhengzhou 450001, China
- Henan Collaborative Innovation Center for Food Production and Safety , Zhengzhou 450001, China
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene and MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430030, China
| | - Peiyi Liu
- Department of Nutrition and Food Hygiene and MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430030, China
| | - Lin Xiao
- Department of Nutrition and Food Hygiene and MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430030, China
| | - Liegang Liu
- Department of Nutrition and Food Hygiene and MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430030, China
| | - Ruiling Shen
- College of Food and Biological Engineering, Zhengzhou University of Light Industry , Zhengzhou 450001, China
- Henan Collaborative Innovation Center for Food Production and Safety , Zhengzhou 450001, China
| | - Qianchun Deng
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences , Wuhan 430062, China
- Hubei Key Laboratory of Lipid Chemistry and Nutrition , Wuhan 430062, China
- Key Laboratory of Oilseeds Processing, Ministry of Agriculture , Wuhan 430062, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene and MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology , Wuhan 430030, China
| |
Collapse
|
121
|
Litwinoff EMS, Gold MY, Singh K, Hu J, Li H, Cadwell K, Schmidt AM. Myeloid ATG16L1 does not affect adipose tissue inflammation or body mass in mice fed high fat diet. Obes Res Clin Pract 2017; 12:174-186. [PMID: 29103907 DOI: 10.1016/j.orcp.2017.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/04/2017] [Accepted: 10/17/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND An influx of lipid-loaded macrophages characterizes visceral adipose tissue (VAT) inflammation, which is an important factor in the development of insulin resistance (IR) in obesity. Depletion of macrophage lipids accompanies increased whole body insulin sensitivity, but the underlying mechanism is unknown. Deficiency of autophagy protein ATG16L1 is associated with increases in inflammatory diseases and lipid metabolism, but the connection between ATG16L1, IR, and obesity remains elusive. We hypothesize that myeloid ATG16L1 contributes to lipid loading in macrophages and to IR. METHODS Wild-type (WT) bone marrow derived macrophages (BMDMs) were treated with fatty acids and assessed for markers of autophagy. Myeloid-deficient Atg16l1 and littermate control male mice were fed high fat diet (HFD) or low fat diet (LFD) for 3 months starting at 8 weeks of age. Mice were assessed for body mass, fat and lean mass, glucose and insulin sensitivity, food consumption and adipose inflammation. Fluorescence-activated cell sorted VAT macrophages were assessed for lipid content and expression of autophagy related genes. RESULTS VAT and VAT macrophages from HFD-fed WT mice did not show differences in autophagy protein and gene expression compared to tissue from LFD-fed mice. Fatty acid-treated BMDMs increased neutral lipid content but did not change autophagy protein expression. HFD-fed Atg16l1 myeloid-deficient and littermate mice demonstrated no differences in body mass, glucose or insulin sensitivity, food consumption, fat or lean mass, macrophage lipid content, or adipose tissue inflammation. CONCLUSION ATG16L1 does not contribute to obesity, IR, adipose tissue inflammation or lipid loading in macrophages in mice fed HFD.
Collapse
Affiliation(s)
- Evelyn M S Litwinoff
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Merav Y Gold
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Karan Singh
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Jiyuan Hu
- Departments of Population Health (Biostatistics) and Environmental Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Huilin Li
- Departments of Population Health (Biostatistics) and Environmental Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, and the Department of Microbiology, NYU Langone Health, New York, NY 10016, USA
| | - Ann Marie Schmidt
- Kimmel Center for Biology and Medicine at the Skirball Institute, and the Department of Microbiology, NYU Langone Health, New York, NY 10016, USA.
| |
Collapse
|
122
|
Lee S, Norheim F, Langleite TM, Noreng HJ, Storås TH, Afman LA, Frost G, Bell JD, Thomas EL, Kolnes KJ, Tangen DS, Stadheim HK, Gilfillan GD, Gulseth HL, Birkeland KI, Jensen J, Drevon CA, Holen T. Effect of energy restriction and physical exercise intervention on phenotypic flexibility as examined by transcriptomics analyses of mRNA from adipose tissue and whole body magnetic resonance imaging. Physiol Rep 2017; 4:4/21/e13019. [PMID: 27821717 PMCID: PMC5112497 DOI: 10.14814/phy2.13019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/09/2016] [Accepted: 10/03/2016] [Indexed: 12/11/2022] Open
Abstract
Overweight and obesity lead to changes in adipose tissue such as inflammation and reduced insulin sensitivity. The aim of this study was to assess how altered energy balance by reduced food intake or enhanced physical activity affect these processes. We studied sedentary subjects with overweight/obesity in two intervention studies, each lasting 12 weeks affecting energy balance either by energy restriction (~20% reduced intake of energy from food) in one group, or by enhanced energy expenditure due to physical exercise (combined endurance‐ and strength‐training) in the other group. We monitored mRNA expression by microarray and mRNA sequencing from adipose tissue biopsies. We also measured several plasma parameters as well as fat distribution with magnetic resonance imaging and spectroscopy. Comparison of microarray and mRNA sequencing showed strong correlations, which were also confirmed using RT‐PCR. In the energy restricted subjects (body weight reduced by 5% during a 12 weeks intervention), there were clear signs of enhanced lipolysis as monitored by mRNA in adipose tissue as well as plasma concentration of free‐fatty acids. This increase was strongly related to increased expression of markers for M1‐like macrophages in adipose tissue. In the exercising subjects (glucose infusion rate increased by 29% during a 12‐week intervention), there was a marked reduction in the expression of markers of M2‐like macrophages and T cells, suggesting that physical exercise was especially important for reducing inflammation in adipose tissue with insignificant reduction in total body weight. Our data indicate that energy restriction and physical exercise affect energy‐related pathways as well as inflammatory processes in different ways, probably related to macrophages in adipose tissue.
Collapse
Affiliation(s)
- Sindre Lee
- Department of Nutrition, Institute of Basic Medical Sciences Faculty of Medicine University of Oslo, Oslo, Norway
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences Faculty of Medicine University of Oslo, Oslo, Norway.,Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Torgrim M Langleite
- Department of Nutrition, Institute of Basic Medical Sciences Faculty of Medicine University of Oslo, Oslo, Norway
| | - Hans J Noreng
- The Intervention Centre, Oslo University Hospital Oslo, Oslo, Norway
| | - Trygve H Storås
- The Intervention Centre, Oslo University Hospital Oslo, Oslo, Norway
| | - Lydia A Afman
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Gary Frost
- Division of Diabetes, Endocrinology and Metabolism, Dietetics, Imperial College Hammersmith Campus, London, UK
| | - Jimmy D Bell
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, UK
| | - E Louise Thomas
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, UK
| | - Kristoffer J Kolnes
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Daniel S Tangen
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Hans K Stadheim
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | | | - Hanne L Gulseth
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of medicine, University of Oslo, Oslo, Norway
| | - Kåre I Birkeland
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of medicine, University of Oslo, Oslo, Norway
| | - Jørgen Jensen
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences Faculty of Medicine University of Oslo, Oslo, Norway
| | - Torgeir Holen
- Department of Nutrition, Institute of Basic Medical Sciences Faculty of Medicine University of Oslo, Oslo, Norway
| | | |
Collapse
|
123
|
Alshahrani A, Bin Khunayfir A, Al Rayih M, Al Sayed H, Alsadoon A, Al Dubayee M, Zahra M, Alrumayyan Y, Al Zayer M, Nasr A, Aljada A. Phenotypic Characterization of Human Monocytes following Macronutrient Intake in Healthy Humans. Front Immunol 2017; 8:1293. [PMID: 29109719 PMCID: PMC5660602 DOI: 10.3389/fimmu.2017.01293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 09/26/2017] [Indexed: 01/04/2023] Open
Abstract
Background Three subsets of human monocytes in circulation have been identified and their characterization is still ill-defined. Although glucose and lipid intakes have been demonstrated to exert pro-inflammatory effects on mononuclear cells (MNCs) of healthy subjects, characterization of monocytes phenotypes following macronutrient (glucose, protein, and lipid) intake in humans remains to be determined. Methods Thirty-six healthy, normal weight volunteers were recruited in the study. Subjects were randomly assigned into three groups, each group consisting of 12 participants. Each group drank equal calories (300 kcal) of either glucose or lipids or whey proteins. Each subject served as his own control by drinking 300 mL of water 1 week before or after the caloric intake. Baseline blood samples were drawn at 0, 1, 2, and 3-h intervals post caloric or water intakes. MNCs were isolated, and the expression levels of different cluster of differentiation (CD) markers (CD86, CD11c, CD169, CD206, CD163, CD36, CD68, CD11b, CD16, and CD14) and IL-6 were measured by RT-qPCR. Results Equicaloric intake of either glucose or lipids or whey proteins resulted in different monocyte phenotypes as demonstrated by changes in the expression levels of CD and polarization markers. Whey proteins intake resulted in significant mRNA upregulation in MNCs of CD68 and CD11b at 1, 2, and 3 h post intake while mRNA of IL-6 was significantly inhibited at 1 h. Lipids intake, on the other hand, resulted in mRNA upregulation of CD11b at 2 and 3 h and CD206 at 1, 2, and 3 h. There were no significant changes in the other CD markers measured (CD86, CD163, CD169, CD36, CD16, and CD14) following either whey proteins or lipids intakes. Glucose intake did not alter mRNA expression of any marker tested except CD206 at 3 h. Conclusion Macronutrient intake alters the expression levels of polarization markers in MNCs of human subjects. A distinct population of different monocytes phenotypes may result in human circulation following the intake of different macronutrients. Further studies are required to characterize the immunomodulatory effects of macronutrients intake on monocytes phenotypes and their characteristics in humans.
Collapse
Affiliation(s)
- Awad Alshahrani
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abdalmalik Bin Khunayfir
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Mohammed Al Rayih
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Hasan Al Sayed
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abdullah Alsadoon
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Mohammed Al Dubayee
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Mahmoud Zahra
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Yousof Alrumayyan
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Maha Al Zayer
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Amre Nasr
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ahmad Aljada
- Department of Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| |
Collapse
|
124
|
Macrophage VLDLR mediates obesity-induced insulin resistance with adipose tissue inflammation. Nat Commun 2017; 8:1087. [PMID: 29057873 PMCID: PMC5651811 DOI: 10.1038/s41467-017-01232-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 08/25/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is closely associated with increased adipose tissue macrophages (ATMs), which contribute to systemic insulin resistance and altered lipid metabolism by creating a pro-inflammatory environment. Very low-density lipoprotein receptor (VLDLR) is involved in lipoprotein uptake and storage. However, whether lipid uptake via VLDLR in macrophages affects obesity-induced inflammatory responses and insulin resistance is not well understood. Here we show that elevated VLDLR expression in ATMs promotes adipose tissue inflammation and glucose intolerance in obese mice. In macrophages, VLDL treatment upregulates intracellular levels of C16:0 ceramides in a VLDLR-dependent manner, which potentiates pro-inflammatory responses and promotes M1-like macrophage polarization. Adoptive transfer of VLDLR knockout bone marrow to wild-type mice relieves adipose tissue inflammation and improves insulin resistance in diet-induced obese mice. These findings suggest that increased VLDL-VLDLR signaling in ATMs aggravates adipose tissue inflammation and insulin resistance in obesity. VLDLR regulates cellular lipoprotein uptake and storage. Here, the authors show that VLDLR, expressed on adipose tissue macrophages, is upregulated in obesity and promotes adipose tissue inflammation by upregulating ceramide production and facilitating M1-like macrophage polarization.
Collapse
|
125
|
ERV1 Overexpression in Myeloid Cells Protects against High Fat Diet Induced Obesity and Glucose Intolerance. Sci Rep 2017; 7:12848. [PMID: 28993702 PMCID: PMC5634420 DOI: 10.1038/s41598-017-13185-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/19/2017] [Indexed: 12/26/2022] Open
Abstract
Non-resolving inflammation is a central pathologic component of obesity, insulin resistance, type 2 diabetes and associated morbidities. The resultant hyperglycemia is deleterious to the normal function of many organs and its control significantly improves survival and quality of life for patients with diabetes. Macrophages play critical roles in both onset and progression of obesity-associated insulin resistance. Here we show that systemic activation of inflammation resolution prevents from morbid obesity and hyperglycemia under dietary overload conditions. In gain-of-function studies using mice overexpressing the human resolvin E1 receptor (ERV1) in myeloid cells, monocyte phenotypic shifts to increased patrolling-to-inflammatory ratio controlled inflammation, reduced body weight gain and protected from hyperglycemia on high-fat diet. Administration of a natural ERV1 agonist, resolvin E1, recapitulated the pro-resolving actions gained by ERV1 overexpression. This protective metabolic impact is in part explained by systemic activation of resolution programs leading to increased synthesis of specialized pro-resolving mediators.
Collapse
|
126
|
Murakami S. The physiological and pathophysiological roles of taurine in adipose tissue in relation to obesity. Life Sci 2017; 186:80-86. [DOI: 10.1016/j.lfs.2017.08.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/31/2017] [Accepted: 08/08/2017] [Indexed: 01/08/2023]
|
127
|
Adipose tissue conditioned media support macrophage lipid-droplet biogenesis by interfering with autophagic flux. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1001-1012. [DOI: 10.1016/j.bbalip.2017.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/04/2017] [Accepted: 06/19/2017] [Indexed: 01/20/2023]
|
128
|
Stanley EG, Jenkins BJ, Walker CG, Koulman A, Browning L, West AL, Calder PC, Jebb SA, Griffin JL. Lipidomics Profiling of Human Adipose Tissue Identifies a Pattern of Lipids Associated with Fish Oil Supplementation. J Proteome Res 2017; 16:3168-3179. [PMID: 28587463 DOI: 10.1021/acs.jproteome.7b00161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To understand the interaction between diet and health, biomarkers that accurately reflect consumption of foods of perceived health relevance are needed. The aim of this investigation was to use direct infusion-mass spectrometry (DI-MS) lipidomics to determine the effects of fish oil supplementation on lipid profiles of human adipose tissue. Adipose tissue samples from an n-3 polyunsaturated fatty acid (PUFA) supplementation study (n = 66) were analyzed to compare the pattern following supplementation equivalent to zero or four portions of oily fish per week. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) were incorporated into highly unsaturated (≥5 double bonds) triglycerides (TGs), phosphocholines, and phosphoethanolamines as well as being detected directly as the nonesterified fatty acid forms. Multivariate statistics demonstrated that phospholipids were the most accurate and sensitive lipids for the assessing EPA and DHA incorporation into adipose tissue. Potential confounding factors (adiposity, age, and sex of the subject) were also considered in the analysis, and adiposity was also associated with an increase in highly unsaturated TGs as a result of incorporation of the n-6 PUFA arachidonic acid. DI-MS provides a high-throughput analysis of fatty acid status that can monitor oily fish consumption, suitable for use in cohort studies.
Collapse
Affiliation(s)
- Elizabeth G Stanley
- MRC Human Nutrition Research, Elsie Widdowson Laboratory , Cambridge CB1 9NL, United Kingdom
| | - Benjamin J Jenkins
- MRC Human Nutrition Research, Elsie Widdowson Laboratory , Cambridge CB1 9NL, United Kingdom
| | - Celia G Walker
- MRC Human Nutrition Research, Elsie Widdowson Laboratory , Cambridge CB1 9NL, United Kingdom
| | - Albert Koulman
- MRC Human Nutrition Research, Elsie Widdowson Laboratory , Cambridge CB1 9NL, United Kingdom
| | - Lucy Browning
- MRC Human Nutrition Research, Elsie Widdowson Laboratory , Cambridge CB1 9NL, United Kingdom
| | - Annette L West
- Faculty of Medicine, University of Southampton , Southampton SO16 6YD, United Kingdom
| | - Philip C Calder
- Faculty of Medicine, University of Southampton , Southampton SO16 6YD, United Kingdom.,NIHR Southampton Biomedical Research Centre, University Hospital NHS Foundation Trust and University of Southampton , Southampton SO16 6YD, United Kingdom
| | - Susan A Jebb
- MRC Human Nutrition Research, Elsie Widdowson Laboratory , Cambridge CB1 9NL, United Kingdom.,Nuffield Department of Primary Care Health Sciences, University of Oxford , Radcliffe Observatory Quarter, Oxford OX2 6GG, United Kingdom
| | - Julian L Griffin
- MRC Human Nutrition Research, Elsie Widdowson Laboratory , Cambridge CB1 9NL, United Kingdom.,Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge , Sanger Building, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| |
Collapse
|
129
|
Gabriel TL, Mirzaian M, Hooibrink B, Ottenhoff R, van Roomen C, Aerts JMFG, van Eijk M. Induction of Sphk1 activity in obese adipose tissue macrophages promotes survival. PLoS One 2017; 12:e0182075. [PMID: 28753653 PMCID: PMC5533446 DOI: 10.1371/journal.pone.0182075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 07/12/2017] [Indexed: 02/07/2023] Open
Abstract
During obesity, adipose tissue macrophages (ATM) are increased in concert with local inflammation and insulin resistance. Since the levels of sphingolipid (SLs) in adipose tissue (AT) are altered during obesity we investigated the potential impact of SLs on ATMs. For this, we first analyzed expression of SL metabolizing genes in ATMs isolated from obese mice. A marked induction of sphingosine kinase 1 (Sphk1) expression was observed in obese ATM when compared to lean ATM. This induction was observed in both MGL-ve (M1) and MGL1+ve (M2) macrophages from obese WAT. Next, RAW264.7 cells were exposed to excessive palmitate, resulting in a similar induction of Sphk1. This Sphk1 induction was also observed when cells were treated with chloroquine, a lysosomotropic amine impacting lysosome function. Simultaneous incubation of RAW cells with palmitate and the Sphk1 inhibitor SK1-I promoted cell death, suggesting a protective role of Sphk1 during lipotoxic conditions. Interestingly, a reduction of endoplasmic reticulum (ER) stress related genes was detected in obese ATM and was found to be associated with elevated Sphk1 expression. Altogether, our data suggest that lipid overload in ATM induces Sphk1, which promotes cell viability.
Collapse
Affiliation(s)
- Tanit L. Gabriel
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Mina Mirzaian
- Department of Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Berend Hooibrink
- Department of Cell Biology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Roelof Ottenhoff
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Cindy van Roomen
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Johannes M. F. G. Aerts
- Department of Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Marco van Eijk
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
- Department of Biochemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
130
|
Sreedhar R, Arumugam S, Thandavarayan RA, Karuppagounder V, Koga Y, Nakamura T, Harima M, Watanabe K. Role of 14-3-3η protein on cardiac fatty acid metabolism and macrophage polarization after high fat diet induced type 2 diabetes mellitus. Int J Biochem Cell Biol 2017; 88:92-99. [DOI: 10.1016/j.biocel.2017.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/20/2017] [Accepted: 05/04/2017] [Indexed: 01/13/2023]
|
131
|
Rombaldova M, Janovska P, Kopecky J, Kuda O. Omega-3 fatty acids promote fatty acid utilization and production of pro-resolving lipid mediators in alternatively activated adipose tissue macrophages. Biochem Biophys Res Commun 2017; 490:1080-1085. [PMID: 28668396 DOI: 10.1016/j.bbrc.2017.06.170] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 12/31/2022]
Abstract
It is becoming increasingly apparent that mutual interactions between adipocytes and immune cells are key to the integrated control of adipose tissue inflammation and lipid metabolism in obesity, but little is known about the non-inflammatory functions of adipose tissue macrophages (ATMs) and how they might be impacted by neighboring adipocytes. In the current study we used metabolipidomic analysis to examine the adaptations to lipid overload of M1 or M2 polarized macrophages co-incubated with adipocytes and explored potential benefits of omega-3 polyunsaturated fatty acids (PUFA). Macrophages adjust their metabolism to process excess lipids and M2 macrophages in turn modulate lipolysis and fatty acids (FA) re-esterification of adipocytes. While M1 macrophages tend to store surplus FA as triacylglycerols and cholesteryl esters in lipid droplets, M2 macrophages channel FA toward re-esterification and β-oxidation. Dietary omega-3 PUFA enhance β-oxidation in both M1 and M2. Our data document that ATMs contribute to lipid trafficking in adipose tissue and that omega-3 PUFA could modulate FA metabolism of ATMs.
Collapse
Affiliation(s)
- Martina Rombaldova
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Praha 4, Czech Republic; Charles University in Prague, Faculty of Science, Department of Analytical Chemistry, Albertov 2030, 128 43 Prague, Czech Republic
| | - Petra Janovska
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Praha 4, Czech Republic
| | - Jan Kopecky
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Praha 4, Czech Republic
| | - Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Praha 4, Czech Republic.
| |
Collapse
|
132
|
Lin YW, Wei LN. Innate immunity orchestrates adipose tissue homeostasis. Horm Mol Biol Clin Investig 2017; 31:hmbci-2017-0013. [PMID: 28672736 DOI: 10.1515/hmbci-2017-0013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/01/2017] [Indexed: 02/07/2023]
Abstract
Obesity is strongly associated with multiple diseases including insulin resistance, type 2 diabetes, cardiovascular diseases, fatty liver disease, neurodegenerative diseases and cancers, etc. Adipose tissue (AT), mainly brown AT (BAT) and white AT (WAT), is an important metabolic and endocrine organ that maintains whole-body homeostasis. BAT contributes to non-shivering thermogenesis in a cold environment; WAT stores energy and produces adipokines that fine-tune metabolic and inflammatory responses. Obesity is often characterized by over-expansion and inflammation of WAT where inflammatory cells/mediators are abundant, especially pro-inflammatory (M1) macrophages, resulting in chronic low-grade inflammation and leading to insulin resistance and metabolic complications. Macrophages constitute the major component of innate immunity and can be activated as a M1 or M2 (anti-inflammatory) phenotype in response to environmental stimuli. Polarized M1 macrophage causes AT inflammation, whereas polarized M2 macrophage promotes WAT remodeling into the BAT phenotype, also known as WAT browning/beiging, which enhances insulin sensitivity and metabolic health. This review will discuss the regulation of AT homeostasis in relation to innate immunity.
Collapse
Affiliation(s)
- Yi-Wei Lin
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, 612 Jackson Hall, 321 Church St. SE., Minneapolis, MN 55455, USA, Phone: 612-625-9402, Fax: 612-625-8408
| |
Collapse
|
133
|
Chistiakov DA, Grechko AV, Myasoedova VA, Melnichenko AA, Orekhov AN. Impact of the cardiovascular system-associated adipose tissue on atherosclerotic pathology. Atherosclerosis 2017. [PMID: 28629772 DOI: 10.1016/j.atherosclerosis.2017.06.017] [Citation(s) in RCA: 37] [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] [Indexed: 01/07/2023]
Abstract
Cardiac obesity makes an important contribution to the pathogenesis of cardiovascular disease. One of the important pathways of this contribution is the inflammatory process that takes place in the adipose tissue. In this review, we consider the role of the cardiovascular system-associated fat in atherosclerotic cardiovascular pathology and a non-atherosclerotic cause of coronary artery disease, such as atrial fibrillation. Cardiovascular system-associated fat not only serves as the energy store, but also releases adipokines that control local and systemic metabolism, heart/vascular function and vessel tone, and a number of vasodilating and anti-inflammatory substances. Adipokine appears to play an important protective role in cardiovascular system. Under chronic inflammation conditions, the repertoire of signaling molecules secreted by cardiac fat can be altered, leading to a higher amount of pro-inflammatory messengers, vasoconstrictors, profibrotic modulators. This further aggravates cardiovascular inflammation and leads to hypertension, induction of the pathological tissue remodeling and cardiac fibrosis. Contemporary imaging techniques showed that epicardial fat thickness correlates with the visceral fat mass, which is an established risk factor and predictor of cardiovascular disease in obese subjects. However, this correlation is no longer present after adjustment for other covariates. Nevertheless, recent studies showed that pericardial fat volume and epicardial fat thickness can probably serve as a better indicator for atrial fibrillation.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Neurochemistry, Division of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center of Psychiatry and Narcology, 119991 Moscow, Russia
| | - Andrey V Grechko
- Federal Scientific Clinical Center for Resuscitation and Rehabilitation, 109240 Moscow, Russia
| | - Veronika A Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia
| | - Alexandra A Melnichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow 121609, Russia.
| |
Collapse
|
134
|
Adipocyte-Macrophage Cross-Talk in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:327-343. [DOI: 10.1007/978-3-319-48382-5_14] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
135
|
Pirola L, Ferraz JC. Role of pro- and anti-inflammatory phenomena in the physiopathology of type 2 diabetes and obesity. World J Biol Chem 2017; 8:120-128. [PMID: 28588755 PMCID: PMC5439163 DOI: 10.4331/wjbc.v8.i2.120] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/24/2017] [Accepted: 02/20/2017] [Indexed: 02/05/2023] Open
Abstract
In obesity, persistent low-grade inflammation is considered as a major contributor towards the progression to insulin resistance and type 2 diabetes while in lean subjects the immune environment is non-inflammatory. Massive adipose tissue (AT) infiltration by pro-inflammatory M1 macrophages and several T cell subsets as obesity develops leads to the accumulation - both in the AT and systemically - of numerous pro-inflammatory cytokines, including interleukin-1β (IL-1β), tumor necrosis factor α, IL-17 and IL-6 which are strongly associated with the progression of the obese phenotype towards the metabolic syndrome. At the same time, anti-inflammatory M2 macrophages and Th subsets producing the anti-inflammatory cytokines IL-10, IL-5 and interferon-γ, including Th2 and T-reg cells are correlated to the maintenance of AT homeostasis in lean individuals. Here, we discuss the basic principles in the control of the interaction between the AT and infiltrating immune cells both in the lean and the obese condition with a special emphasis on the contribution of pro- and anti-inflammatory cytokines to the establishment of the insulin-resistant state. In this context, we will discuss the current knowledge about alterations in the levels on pro- and anti-inflammatory cytokines in obesity, insulin resistance and type 2 diabetes mellitus, in humans and animal models. Finally, we also briefly survey the recent novel therapeutic strategies that attempt to alleviate or reverse insulin resistance and type 2 diabetes via the administration of recombinant inhibitory antibodies directed towards some pro-inflammatory cytokines.
Collapse
|
136
|
Role of immune cells in obesity induced low grade inflammation and insulin resistance. Cell Immunol 2017; 315:18-26. [DOI: 10.1016/j.cellimm.2017.03.001] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 02/09/2017] [Accepted: 03/05/2017] [Indexed: 12/11/2022]
|
137
|
Verdeguer F, Aouadi M. Macrophage heterogeneity and energy metabolism. Exp Cell Res 2017; 360:35-40. [PMID: 28341447 DOI: 10.1016/j.yexcr.2017.03.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 02/06/2023]
Abstract
Macrophages are versatile and multifunctional cell types present in most vertebrate tissues. They are the first line of defense against pathogens through phagocytosis of microbial infections, particles and dead cells. Macrophages harbor additional functions besides immune protection by participating in essential homeostatic and tissue development functions. The immune response requires a concomitant and coordinated regulation of the energetic metabolism. In this review, we will discuss how macrophages influence metabolic tissues and in turn how metabolic pathways, particularly glucose and lipid metabolism, affect macrophage phenotypes.
Collapse
Affiliation(s)
- Francisco Verdeguer
- Department of Medicine, KI/AZ Integrated Cardio Metabolic Center, Karolinska Institutet at Karolinska University Hospital Huddinge, C2-84, S-141 86 Stockholm, Sweden
| | - Myriam Aouadi
- Department of Medicine, KI/AZ Integrated Cardio Metabolic Center, Karolinska Institutet at Karolinska University Hospital Huddinge, C2-84, S-141 86 Stockholm, Sweden.
| |
Collapse
|
138
|
Microglia activation due to obesity programs metabolic failure leading to type two diabetes. Nutr Diabetes 2017; 7:e254. [PMID: 28319103 PMCID: PMC5380893 DOI: 10.1038/nutd.2017.10] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/04/2016] [Accepted: 01/22/2017] [Indexed: 02/06/2023] Open
Abstract
Obesity is an energy metabolism disorder that increases susceptibility to the development of metabolic diseases. Recently, it has been described that obese subjects have a phenotype of chronic inflammation in organs that are metabolically relevant for glucose homeostasis and energy. Altered expression of immune system molecules such as interleukins IL-1, IL-6, IL-18, tumor necrosis factor alpha (TNF-α), serum amyloid A (SAA), and plasminogen activator inhibitor-1 (PAI-1), among others, has been associated with the development of chronic inflammation in obesity. Chronic inflammation modulates the development of metabolic-related comorbidities like metabolic syndrome (insulin resistance, glucose tolerance, hypertension and hyperlipidemia). Recent evidence suggests that microglia activation in the central nervous system (CNS) is a priority in the deregulation of energy homeostasis and promotes increased glucose levels. This review will cover the most significant advances that explore the molecular signals during microglia activation and inflammatory stage in the brain in the context of obesity, and its influence on the development of metabolic syndrome and type two diabetes.
Collapse
|
139
|
Pereira JX, Cavalcante Y, Wanzeler de Oliveira R. The role of inflammation in adipocytolytic nonsurgical esthetic procedures for body contouring. Clin Cosmet Investig Dermatol 2017; 10:57-66. [PMID: 28260937 PMCID: PMC5327851 DOI: 10.2147/ccid.s125580] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background The adipocytolytic non-surgical esthetic procedures are indicated for the reduction of localized fat and are effective in reducing local adiposity through the ablation of adipocytes with fast and lasting results, besides causing local inflammation. Objective This study aimed to characterize the adipocytolytic procedures and correlate the phases of the inflammatory process with the results obtained from such procedures, in order to clarify the role of inflammation triggered by the adipocytolytic procedures and its relation with the lipolytic process, with a focus on body shaping. Methods This work is an integrative literature review that presents a total of 72 articles published between 1998 and 2015, derived from the PubMed database, in order to establish a relationship between clinical and basic science research, assuming an important role in medical practice based on evidence. Results The results show that the adipocytolytic procedures are characterized by triggering inflammation arising from the disruption of adipocytes by interfering with the lipolytic signaling pathways in both acute and chronic phases of inflammation through the direct action of proinflammatory cytokines or catecholamines. Therefore, inflammation plays an important role in modulating the lipolytic process, influencing body shaping. Conclusion The inflammatory process assists the adipolytic process in all stages of inflammation, contributing to the reduction of body contouring.
Collapse
|
140
|
Mechanistic interplay between ceramide and insulin resistance. Sci Rep 2017; 7:41231. [PMID: 28112248 PMCID: PMC5253739 DOI: 10.1038/srep41231] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
Recent research adds to a growing body of literature on the essential role of ceramides in glucose homeostasis and insulin signaling, while the mechanistic interplay between various components of ceramide metabolism remains to be quantified. We present an extended model of C16:0 ceramide production through both the de novo synthesis and the salvage pathways. We verify our model with a combination of published models and independent experimental data. In silico experiments of the behavior of ceramide and related bioactive lipids in accordance with the observed transcriptomic changes in obese/diabetic murine macrophages at 5 and 16 weeks support the observation of insulin resistance only at the later phase. Our analysis suggests the pivotal role of ceramide synthase, serine palmitoyltransferase and dihydroceramide desaturase involved in the de novo synthesis and the salvage pathways in influencing insulin resistance versus its regulation.
Collapse
|
141
|
McLaughlin T, Ackerman SE, Shen L, Engleman E. Role of innate and adaptive immunity in obesity-associated metabolic disease. J Clin Invest 2017; 127:5-13. [PMID: 28045397 DOI: 10.1172/jci88876] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation in adipose tissue, possibly related to adipose cell hypertrophy, hypoxia, and/or intestinal leakage of bacteria and their metabolic products, likely plays a critical role in the development of obesity-associated insulin resistance (IR). Cells of both the innate and adaptive immune system residing in adipose tissues, as well as in the intestine, participate in this process. Thus, M1 macrophages, IFN-γ-secreting Th1 cells, CD8+ T cells, and B cells promote IR, in part through secretion of proinflammatory cytokines. Conversely, eosinophils, Th2 T cells, type 2 innate lymphoid cells, and possibly Foxp3+ Tregs protect against IR through local control of inflammation.
Collapse
|
142
|
Abstract
Chronic inflammatory state in obesity causes dysregulation of the endocrine and paracrine actions of adipocyte-derived factors, which disrupt vascular homeostasis and contribute to endothelial vasodilator dysfunction and subsequent hypertension. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Adipose tissue inflammation, nitric oxide (NO)-bioavailability, insulin resistance and oxidized low-density lipoprotein (oxLDL) are main participating factors in endothelial dysfunction of obesity. In this chapter, disruption of inter-endothelial junctions between endothelial cells, significant increase in the production of reactive oxygen species (ROS), inflammation mediators, which are originated from inflamed endothelial cells, the balance between NO synthesis and ROS , insulin signaling and NO production, and decrease in L-arginine/endogenous asymmetric dimethyl-L-arginine (ADMA) ratio are discussed in connection with endothelial dysfunction in obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
143
|
Miron RJ, Zohdi H, Fujioka-Kobayashi M, Bosshardt DD. Giant cells around bone biomaterials: Osteoclasts or multi-nucleated giant cells? Acta Biomater 2016; 46:15-28. [PMID: 27667014 DOI: 10.1016/j.actbio.2016.09.029] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 09/14/2016] [Accepted: 09/22/2016] [Indexed: 12/31/2022]
Abstract
Recently accumulating evidence has put into question the role of large multinucleated giant cells (MNGCs) around bone biomaterials. While cells derived from the monocyte/macrophage lineage are one of the first cell types in contact with implanted biomaterials, it was originally thought that specifically in bone tissues, all giant cells were bone-resorbing osteoclasts whereas foreign body giant cells (FBGCs) were found associated with a connective tissue foreign body reaction resulting in fibrous encapsulation and/or material rejection. Despite the great majority of bone grafting materials routinely found with large osteoclasts, a special subclass of bone biomaterials has more recently been found surrounded by large giant cells virtually incapable of resorbing bone grafts even years after their implantation. While original hypotheses believed that a 'foreign body reaction' may be taking place, histological data retrieved from human samples years after their implantation have put these original hypotheses into question by demonstrating better and more stable long-term bone volume around certain bone grafts. Exactly how or why this 'special' subclass of giant cells is capable of maintaining long-term bone volume, or methods to scientifically distinguish them from osteoclasts remains extremely poorly studied. The aim of this review article was to gather the current available literature on giant cell markers and differences in expression patterns between osteoclasts and MNGCs utilizing 19 specific markers including an array of CD-cell surface markers. Furthermore, the concept of now distinguishing between pro-inflammatory M1-MNGCs (previously referred to as FBGCs) as well as wound-healing M2-MNGCs is introduced and discussed. STATEMENT OF SIGNIFICANCE This review article presents 19 specific cell-surface markers to distinguish between osteoclasts and MNGCs including an array of CD-cell surface markers. Furthermore, the concept of now distinguishing between pro-inflammatory M1-MNGCs (often previously referred to as FBGCs) as well as wound-healing M2-MNGCs is introduced and discussed. The proposed concepts and guidelines aims to guide the next wave of research facilitating the differentiation between osteoclast/MNGCs formation, as well as provides the basis for increasing our understanding of the exact function of MNGCs in bone tissue/biomaterial homeostasis.
Collapse
|
144
|
Contreras GA, Thelen K, Schmidt SE, Strieder-Barboza C, Preseault CL, Raphael W, Kiupel M, Caron J, Lock AL. Adipose tissue remodeling in late-lactation dairy cows during feed-restriction-induced negative energy balance. J Dairy Sci 2016; 99:10009-10021. [DOI: 10.3168/jds.2016-11552] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/19/2016] [Indexed: 12/12/2022]
|
145
|
Zhao Y, Gu X, Zhang N, Kolonin MG, An Z, Sun K. Divergent functions of endotrophin on different cell populations in adipose tissue. Am J Physiol Endocrinol Metab 2016; 311:E952-E963. [PMID: 27729337 PMCID: PMC6189636 DOI: 10.1152/ajpendo.00314.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/13/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022]
Abstract
Endotrophin is a cleavage product of collagen 6 (Col6) in adipose tissue (AT). Previously, we demonstrated that endotrophin serves as a costimulator to trigger fibrosis and inflammation within the unhealthy AT milieu. However, how endotrophin affects lipid storage and breakdown in AT and how different cell types in AT respond to endotrophin stimulation remain unknown. In the current study, by using a doxycycline-inducible mouse model, we observed significant upregulation of adipogenic genes in the white AT (WAT) of endotrophin transgenic mice. We further showed that the mice exhibited inhibited lipolysis and accelerated hypertrophy and hyperplasia in WAT. To investigate the effects of endotrophin in vitro, we incubated different cell types from AT with conditioned medium from endotrophin-overexpressing 293T cells. We found that endotrophin activated multiple pathological pathways in different cell types. Particularly in 3T3-L1 adipocytes, endotrophin triggered a fibrotic program by upregulating collagen genes and promoted abnormal lipid accumulation by downregulating hormone-sensitive lipolysis gene and decreasing HSL phosphorylation levels. In macrophages isolated from WAT, endotrophin stimulated higher expression of the collagen-linking enzyme lysyl oxidase and M1 proinflammatory marker genes. In the stromal vascular fraction isolated from WAT, endotrophin induced upregulation of both profibrotic and proinflammatory genes. In conclusion, our study provides a new perspective on the effect of endotrophin in abnormal lipid accumulation and a mechanistic insight into the roles played by adipocytes and a variety of other cell types in AT in shaping the unhealthy microenvironment upon endotrophin treatment.
Collapse
Affiliation(s)
- Yueshui Zhao
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Xue Gu
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Ningyan Zhang
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas
| | - Mikhail G Kolonin
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Zhiqiang An
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas; and
| |
Collapse
|
146
|
Paniagua JA. Nutrition, insulin resistance and dysfunctional adipose tissue determine the different components of metabolic syndrome. World J Diabetes 2016; 7:483-514. [PMID: 27895819 PMCID: PMC5107710 DOI: 10.4239/wjd.v7.i19.483] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 08/16/2016] [Accepted: 09/07/2016] [Indexed: 02/05/2023] Open
Abstract
Obesity is an excessive accumulation of body fat that may be harmful to health. Today, obesity is a major public health problem, affecting in greater or lesser proportion all demographic groups. Obesity is estimated by body mass index (BMI) in a clinical setting, but BMI reports neither body composition nor the location of excess body fat. Deaths from cardiovascular diseases, cancer and diabetes accounted for approximately 65% of all deaths, and adiposity and mainly abdominal adiposity are associated with all these disorders. Adipose tissue could expand to inflexibility levels. Then, adiposity is associated with a state of low-grade chronic inflammation, with increased tumor necrosis factor-α and interleukin-6 release, which interfere with adipose cell differentiation, and the action pattern of adiponectin and leptin until the adipose tissue begins to be dysfunctional. In this state the subject presents insulin resistance and hyperinsulinemia, probably the first step of a dysfunctional metabolic system. Subsequent to central obesity, insulin resistance, hyperglycemia, hypertriglyceridemia, hypoalphalipoproteinemia, hypertension and fatty liver are grouped in the so-called metabolic syndrome (MetS). In subjects with MetS an energy balance is critical to maintain a healthy body weight, mainly limiting the intake of high energy density foods (fat). However, high-carbohydrate rich (CHO) diets increase postprandial peaks of insulin and glucose. Triglyceride-rich lipoproteins are also increased, which interferes with reverse cholesterol transport lowering high-density lipoprotein cholesterol. In addition, CHO-rich diets could move fat from peripheral to central deposits and reduce adiponectin activity in peripheral adipose tissue. All these are improved with monounsaturated fatty acid-rich diets. Lastly, increased portions of ω-3 and ω-6 fatty acids also decrease triglyceride levels, and complement the healthy diet that is recommended in patients with MetS.
Collapse
|
147
|
Morgan A, Mooney K, Mc Auley M. Obesity and the dysregulation of fatty acid metabolism: implications for healthy aging. Expert Rev Endocrinol Metab 2016; 11:501-510. [PMID: 30058918 DOI: 10.1080/17446651.2016.1245141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The population of the world is aging. In 2010, an estimated 524 million people were aged 65 years or older representing eight percent of the global population. By 2050, this number is expected to nearly triple to approximately 1.5 billion, 16 percent of the world's population. Although people are living longer, the quality of their lives are often compromised due to ill-health. Areas covered: Of the conditions which compromise health as we age, obesity is at the forefront. Over half of the global older population were overweight or obese in 2010, significantly increasing the risk of a range of metabolic diseases. Although, it is well recognised excessive calorie intake is a fundamental driver of adipose tissue dysfunction, the relationship between obesity; intrinsic aging; and fat metabolism is less understood. In this review we discuss the intersection between obesity, aging and the factors which contribute to the dysregulation of whole-body fat metabolism. Expert commentary: Being obese disrupts an array of physiological systems and there is significant crosstalk among these. Moreover it is imperative to acknowledge the contribution intrinsic aging makes to the dysregulation of these systems and the onset of disease.
Collapse
Affiliation(s)
- Amy Morgan
- a Department of Chemical Engineering , University of Chester, Thornton Science Park , Chester , UK
| | - Kathleen Mooney
- b Faculty of Health and Social Care , Edge Hill University , Lancashire , UK
| | - Mark Mc Auley
- a Department of Chemical Engineering , University of Chester, Thornton Science Park , Chester , UK
| |
Collapse
|
148
|
Adipose tissue at the nexus of systemic and cellular immunometabolism. Semin Immunol 2016; 28:431-440. [DOI: 10.1016/j.smim.2016.09.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 09/25/2016] [Accepted: 09/30/2016] [Indexed: 12/13/2022]
|
149
|
Vieira WA, Sadie-Van Gijsen H, Ferris WF. Free fatty acid G-protein coupled receptor signaling in M1 skewed white adipose tissue macrophages. Cell Mol Life Sci 2016; 73:3665-76. [PMID: 27173059 PMCID: PMC11108433 DOI: 10.1007/s00018-016-2263-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 12/13/2022]
Abstract
Obesity is associated with the establishment and maintenance of a low grade, chronically inflamed state in the white adipose tissue (WAT) of the body. The WAT macrophage population is a major cellular participant in this inflammatory process that significantly contributes to the pathophysiology of the disease, with the adipose depots of obese individuals, relative to lean counterparts, having an elevated number of macrophages that are skewed towards a pro-inflammatory phenotype. Alterations in the WAT lipid micro-environment, and specifically the availability of free fatty acids, are believed to contribute towards the obesity-related quantitative and functional changes observed in these cells. This review specifically addresses the involvement of the five G-protein coupled free fatty acid receptors which bind exogenous FFAs and signal in macrophages. Particular focus is placed on the involvement of these receptors in macrophage migration and cytokine production, two important aspects that modulate inflammation.
Collapse
Affiliation(s)
- Warren Antonio Vieira
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
| | - Hanél Sadie-Van Gijsen
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
| | - William Frank Ferris
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa.
| |
Collapse
|
150
|
Namgaladze D, Brüne B. Macrophage fatty acid oxidation and its roles in macrophage polarization and fatty acid-induced inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1796-1807. [PMID: 27614008 DOI: 10.1016/j.bbalip.2016.09.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/26/2016] [Accepted: 09/02/2016] [Indexed: 12/14/2022]
Abstract
Recent research considerably changed our knowledge how cellular metabolism affects the immune system. We appreciate that metabolism not only provides energy to immune cells, but also actively influences diverse immune cell phenotypes. Fatty acid metabolism, particularly mitochondrial fatty acid oxidation (FAO) emerges as an important regulator of innate and adaptive immunity. Catabolism of fatty acids also modulates the progression of disease, such as the development of obesity-driven insulin resistance and type II diabetes. Here, we summarize (i) recent developments in research how FAO modulates inflammatory signatures in macrophages in response to saturated fatty acids, and (ii) the role of FAO in regulating anti-inflammatory macrophage polarization. In addition, we define the contribution of AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptors (PPARs), in controlling macrophage biology towards fatty acid metabolism and inflammation.
Collapse
Affiliation(s)
- Dmitry Namgaladze
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Bernhard Brüne
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|