Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Front Immunol ; 15: 1375138, 2024.
Article in English | MEDLINE | ID: mdl-38812501

ABSTRACT

Objectives: The effects of cold exposure on whole-body metabolism in humans have gained increasing attention. Brown or beige adipose tissues are crucial in cold-induced thermogenesis to dissipate energy and thus have the potential to combat metabolic disorders. Despite the immune regulation of thermogenic adipose tissues, the overall changes in vital immune cells during distinct cold periods remain elusive. This study aimed to discuss the overall changes in immune cells under different cold exposure periods and to screen several potential immune cell subpopulations on thermogenic regulation. Methods: Cibersort and mMCP-counter algorithms were employed to analyze immune infiltration in two (brown and beige) thermogenic adipose tissues under distinct cold periods. Changes in some crucial immune cell populations were validated by reanalyzing the single-cell sequencing dataset (GSE207706). Flow cytometry, immunofluorescence, and quantitative real-time PCR assays were performed to detect the proportion or expression changes in mouse immune cells of thermogenic adipose tissues under cold challenge. Results: The proportion of monocytes, naïve, and memory T cells increased, while the proportion of NK cells decreased under cold exposure in brown adipose tissues. Conclusion: Our study revealed dynamic changes in immune cell profiles in thermogenic adipose tissues and identified several novel immune cell subpopulations, which may contribute to thermogenic activation of adipose tissues under cold exposure.


Subject(s)
Adipose Tissue, Brown , Cold Temperature , Thermogenesis , Thermogenesis/immunology , Animals , Mice , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Mice, Inbred C57BL , Male , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/immunology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Monocytes/immunology , Monocytes/metabolism
2.
JCI Insight ; 6(16)2021 08 23.
Article in English | MEDLINE | ID: mdl-34423787

ABSTRACT

BACKGROUNDThe incidence of burn injuries in older patients is dramatically increasing as the population of older people grows. Despite the increased demand for elderly burn care, the mechanisms that mediate increased morbidity and mortality in older trauma patients are unknown. We recently showed that a burn injury invokes white adipose tissue browning that leads to a substantially increased hypermetabolic response associated with poor outcomes. Therefore, the aim of this study was to determine the effect of age on the metabolic adipose response of browning after a burn injury.METHODOne hundred and seventy patients with burn injury admitted to the Ross Tilley Burn Centre were prospectively enrolled and grouped by age as older (≥50 years) and young (≤35 years). Adipose tissue and sera were collected and analyzed for browning markers and metabolic state via histology, gene expression, and resting energy expenditure assays.RESULTSWe found that older patients with burn injury lacked the adipose browning response, as they showed significant reductions in uncoupling protein 1 (UCP1) expression. This failure of the browning response was associated with reduced whole-body metabolism and decreased survival in older patients with burn injury. Mechanistically, we found that the adipose of both aged patients after burn trauma and aged mice after a burn showed impairments in macrophage infiltration and IL-6, key immunological regulators of the browning process after a severe trauma.CONCLUSIONTargeting pathways that activate the browning response represents a potential therapeutic approach to improve outcomes after burn trauma for elderly patients.FUNDINGNIH (R01-GM087285-01), Canadian Institutes of Health Research (grant no. 123336), and Canada Foundation for Innovation Leaders Opportunity Fund (no. 25407).


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Aging/metabolism , Burns/pathology , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Adult , Age Factors , Aged , Animals , Basal Metabolism , Burns/diagnosis , Burns/metabolism , Burns/mortality , Canada , Disease Models, Animal , Female , Humans , Injury Severity Score , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Uncoupling Protein 1/analysis , Uncoupling Protein 1/metabolism
3.
Science ; 373(6550)2021 07 02.
Article in English | MEDLINE | ID: mdl-34210853

ABSTRACT

The mechanisms by which macrophages regulate energy storage remain poorly understood. We identify in a genetic screen a platelet-derived growth factor (PDGF)/vascular endothelial growth factor (VEGF)-family ortholog, Pvf3, that is produced by macrophages and is required for lipid storage in fat-body cells of Drosophila larvae. Genetic and pharmacological experiments indicate that the mouse Pvf3 ortholog PDGFcc, produced by adipose tissue-resident macrophages, controls lipid storage in adipocytes in a leptin receptor- and C-C chemokine receptor type 2-independent manner. PDGFcc production is regulated by diet and acts in a paracrine manner to control lipid storage in adipose tissues of newborn and adult mice. At the organismal level upon PDGFcc blockade, excess lipids are redirected toward thermogenesis in brown fat. These data identify a macrophage-dependent mechanism, conducive to the design of pharmacological interventions, that controls energy storage in metazoans.


Subject(s)
Adipocytes/immunology , Diet, High-Fat , Drosophila Proteins/metabolism , Energy Metabolism , Lymphokines/metabolism , Macrophages/immunology , Obesity/immunology , Platelet-Derived Growth Factor/metabolism , Thermogenesis , Adipose Tissue, Brown/immunology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Hemocytes/immunology , Liver/immunology , Lymphokines/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet-Derived Growth Factor/genetics , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
Sci Rep ; 11(1): 2752, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33531584

ABSTRACT

Neurogenic fever (NF) after subarachnoid hemorrhage (SAH) is a major cause of morbidity that is associated with poor outcomes and prolonged stay in the neurointensive care unit (NICU). Though SAH is a much more common cause of fever than sepsis in the NICU, it is often a diagnosis of exclusion, requiring significant effort to rule out an infectious source. NF does not respond to standard anti-pyretic medications such as COX inhibitors, and lack of good medical therapy has led to the introduction of external cooling systems that have their own associated problems. In a rodent model of SAH, we measured the effects of injecting whole blood, blood plasma, or erythrocytes on the sympathetic nerve activity to brown adipose tissue and on febrile thermogenesis. We demonstrate that following SAH the acute activation of brown adipose tissue leading to NF, is not dependent on PGE2, that subarachnoid space injection of whole blood or erythrocytes, but not plasma alone, is sufficient to trigger brown adipose tissue thermogenesis, and that activation of adenosine A1 receptors in the CNS can block the brown adipose tissue thermogenic component contributing to NF after SAH. These findings point to a distinct thermogenic mechanism for generating NF, compared to those due to infectious causes, and will hopefully lead to new therapies.


Subject(s)
Adipose Tissue, Brown/metabolism , Erythrocytes/immunology , Fever/immunology , Receptor, Adenosine A1/metabolism , Subarachnoid Hemorrhage/complications , Adipose Tissue, Brown/immunology , Animals , Disease Models, Animal , Fever/etiology , Fever/physiopathology , Humans , Male , Rats , Subarachnoid Hemorrhage/immunology , Subarachnoid Hemorrhage/physiopathology , Thermogenesis/immunology
5.
Front Immunol ; 11: 1800, 2020.
Article in English | MEDLINE | ID: mdl-32973755

ABSTRACT

White adipose tissue but recently also brown adipose tissue have emerged as endocrine organs. Age-associated obesity is accompanied by prolonged and elevated lipopolysaccharide (LPS)-induced sickness symptoms and increased cytokine and adipokine levels in the circulation partially originating from adipose tissue. In the present study, ex vivo fat explants were used to investigate how the exogenous pathogen-associated molecular pattern (PAMP) LPS or the endogenous danger-associated molecular patterns (DAMPs) high mobility group box-1 protein (HMGB1) and biglycan modulate the release of cytokines and adipokines/batokines and, thus, could influence systemic and/or local inflammation. The response of adipose tissue (epididymal, retroperitoneal, subcutaneous, and brown) was compared between young lean and old obese rats (2 vs. 24 months old). LPS induced a strong interleukin (IL)-6 and tumor necrosis factor (TNF) alpha release into the supernatant of all adipose tissue types investigated. HMGB1 (subcutaneous) and biglycan (retroperitoneal) led to an increased release of IL-6 and TNFalpha (HMGB1) and decreased visfatin and adiponectin (biglycan) secretion from epididymal adipose tissue (young rats). Visfatin was also decreased by HMGB1 in retroperitoneal adipose tissue of old rats. We found significantly higher leptin (all fat pads) and adiponectin (subcutaneous) levels in supernatants of adipose tissue from old compared to young rats, whereas visfatin secretion showed the opposite. The expression of the biglycan receptor Toll-like receptor (TLR) 2 as well as the LPS and HMGB1 receptors TLR4 and receptor for advanced glycation end products (RAGE) were reduced with age (TLR4/RAGE) and by stimulation with their ligands (subcutaneous). Overall, we revealed that adipokines/adipose-tissue released cytokines show some modulation of their release caused by mediators of septic (batokines) and sterile inflammation with potential implication for acute and chronic disease. Moreover, aging may increase or decrease the release of fat-derived mediators. These data show that DAMPS and LPS locally modulate cytokine secretion while only DAMPS but not LPS can locally alter adipokine secretion during inflammation.


Subject(s)
Adipokines/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Biglycan/pharmacology , Cytokines/metabolism , HMGB1 Protein/pharmacology , Lipopolysaccharides/pharmacology , Toll-Like Receptors/agonists , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Age Factors , Animals , Male , Rats, Wistar , Receptor for Advanced Glycation End Products/agonists , Receptor for Advanced Glycation End Products/metabolism , Secretory Pathway , Signal Transduction , Tissue Culture Techniques , Toll-Like Receptors/metabolism
6.
Sci Rep ; 10(1): 12374, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32704024

ABSTRACT

The functions of adipose tissue are associated with autoimmune diseases, such as rheumatoid arthritis (RA). Some studies have shown that the three compositions of adipose tissue (white, brown, and beige) have different functions. Brown adipose tissue (BAT) is known to secrete several factors that differ from those in white adipose tissue. This suggests that BAT might have potential positive advantages in the physiology of autoimmune diseases. We compared the functions of collagen-induced arthritis mice-derived BAT (CIA BAT) with normal mice-derived BAT. DBA/1J mice (6-7 weeks of age) were immunized by intradermal injection at the base of the tail with 100 µg of bovine type II collagen (CII) emulsified in complete Freund's adjuvant. Immunized mice then received booster immunizations by intraperitoneal injection with 100 µg of CII in incomplete Freund's adjuvant. We transplanted CIA BAT and normal BAT into CIA recipient mice. After transplantation, we measured the functions of CIA BAT and normal BAT in mice. Normal BAT-transplanted mice showed significantly lower scores of bone damage, inflammation, and cartilage damage. The proinflammatory cytokines in normal BAT-transplanted mice, such as IL-12, IL-17, IL-6, and tumor necrosis factor-α (TNF-α), tended to decrease. Microarray analysis showed that the PI3K-AKT signaling pathway and IL-17 levels of CIA BAT tissues were significantly higher than those of normal BAT tissues. These results suggest that the transplantation of normal brown fat may have a therapeutic effect in RA patients.


Subject(s)
Adipose Tissue, Brown/immunology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Cytokines/immunology , Signal Transduction/immunology , Th17 Cells/immunology , Adipose Tissue, Brown/pathology , Adipose Tissue, Brown/transplantation , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/therapy , Male , Mice , Th17 Cells/pathology
7.
FASEB J ; 34(7): 9755-9770, 2020 07.
Article in English | MEDLINE | ID: mdl-32510702

ABSTRACT

Regulatory T cells (Tregs) play essential roles in obesity and diabetes. Here, we report a role of Tregs in enhancing ß3-adrenergic receptor agonist CL316243 (CL)-stimulated thermogenic program in subcutaneous adipose tissue (SAT), but not in visceral fat. CL treatment for 7 days increased SAT adipocyte beiging and thermogenic gene expression in male or female mice. Adoptive transfer of Tregs enhanced this CL activity. Such Treg activity lost in male epididymal white adipose tissue (eWAT) and female gonadal gWAT. Adipocyte culture yielded the same conclusion. Tregs enhanced the expression of CL-induced thermogenic genes in SAT from male and female mice. This activity of Tregs reduced or disappeared in adipocytes from eWAT or gWAT. Both CL and Tregs induced much higher UCP-1 (uncoupling protein-1) expression in SAT from females than that from males. A mechanistic study demonstrated a role of Tregs in suppressing the expression of M1 macrophage markers (Tnfa, Il6, iNos, Ip10) and promoting the expression of M2 macrophage markers (Mrc1, Arg1, Il10) in bone-marrow-derived macrophages or in SAT from male or female mice. In female mice with pre-established obesity, Treg adoptive transfer reduced the gWAT weight in 2 weeks. Together with CL treatment, Treg adoptive transfer reduced the SAT weight and further improved CL-induced glucose metabolism and insulin sensitivity in female obese mice, but did not affect CL-induced body weight loss in male or female obese mice. This study revealed a predominant role of Tregs in female mice in promoting adipocyte beiging and thermogenesis in SAT, in part by slanting M2 macrophage polarization.


Subject(s)
Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Obesity/etiology , Subcutaneous Fat/pathology , T-Lymphocytes, Regulatory/immunology , Thermogenesis , Adipose Tissue, Brown/immunology , Adipose Tissue, White/immunology , Animals , Energy Metabolism , Female , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/pathology , Subcutaneous Fat/immunology , T-Lymphocytes, Regulatory/pathology
8.
Article in English | MEDLINE | ID: mdl-32265845

ABSTRACT

Obesity is characterized by a state of chronic inflammation in adipose tissue mediated by the secretion of a range of inflammatory cytokines. In comparison to WAT, relatively little is known about the inflammatory status of brown adipose tissue (BAT) in physiology and pathophysiology. Because BAT and brown/beige adipocytes are specialized in energy expenditure they have protective roles against obesity and associated metabolic diseases. BAT appears to be is less susceptible to developing inflammation than WAT. However, there is increasing evidence that inflammation directly alters the thermogenic activity of brown fat by impairing its capacity for energy expenditure and glucose uptake. The inflammatory microenvironment can be affected by cytokines secreted by immune cells as well as by the brown adipocytes themselves. Therefore, pro-inflammatory signals represent an important component of the thermogenic potential of brown and beige adipocytes and may contribute their dysfunction in obesity.


Subject(s)
Adipose Tissue, Brown/immunology , Inflammation/complications , Obesity/pathology , Thermogenesis , Adipogenesis , Adipose Tissue, Brown/metabolism , Animals , Humans , Inflammation/immunology , Obesity/etiology , Signal Transduction
9.
Obes Rev ; 21(2): e12963, 2020 02.
Article in English | MEDLINE | ID: mdl-31663251

ABSTRACT

Obesity epidemic responsible for increase in diabetes, heart diseases, infections and cancer shows no signs of abating. Obesity in children is also on rise, indicating the urgent need of strategies for prevention and intervention that must begin in early life. While originally posited that obesity results from the simple concept of consuming more calories, or genetics, emerging research suggests that the bacteria living in our gut (gut microbiome) and its interactions with immune cells and metabolic organs including adipose tissues (microbiome-immune-metabolic axis) play significant role in obesity development in childhood. Specifically, abnormal changes (dysbiosis) in the gut microbiome, stimulation of inflammatory cytokines, and shifts in the metabolic functions of brown adipose tissue and the browning of white adipose tissue are associated with increased obesity. Many factors from as early as gestation appear to contribute in obesity, such as maternal health, diet, antibiotic use by mother and/or child, and birth and feeding methods. Herein, using evidence from animal and human studies, we discuss how these factors impact microbiome-immune-metabolic axis and cause obesity epidemic in children, and describe the gaps in knowledge that are warranted for future research.


Subject(s)
Gastrointestinal Microbiome/immunology , Pediatric Obesity/immunology , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Child , Humans , Pediatric Obesity/metabolism , Pediatric Obesity/microbiology
10.
Mol Metab ; 28: 73-82, 2019 10.
Article in English | MEDLINE | ID: mdl-31427184

ABSTRACT

OBJECTIVE: Obesity and type-2 diabetes (T2D) are metabolic diseases that represent a critical health problem worldwide. Metabolic disease is differentially associated with fat distribution, while visceral white adipose tissue (VAT) is particularly prone to obesity-associated inflammation. Next to their canonical function of immune suppression, regulatory T cells (Tregs) are key in controlling adipose tissue homeostasis. Towards understanding the molecular underpinnings of metabolic disease, we focus on how environmental-metabolic stimuli impinge on the functional interplay between Tregs and adipose tissue. Here, cold exposure or beta3-adrenergic signaling are a promising tool to increase energy expenditure by activating brown adipose tissue, as well as by reducing local inflammation within fat depots by supporting immunosuppressive Tregs. However, in humans, the underlying mechanisms that enable the environmental-immune crosstalk in the periphery and in the respective tissue remain currently unknown. METHODS: We used combinatorial approaches of next generation humanized mouse models and in vitro and in vivo experiments together with beta3-adrenergic stimulation to dissect the underlying mechanisms of human Treg induction exposed to environmental stimuli such as cold. To test the translational relevance of our findings, we analyzed samples from the FREECE study in which human subjects were exposed to individualized cooling protocols. Samples were analyzed ex vivo and after in vitro Treg induction using qRT-PCR, immunofluorescence, as well as with multicolor flow cytometry and cell sorting. RESULTS: In vivo application of the beta3-adrenergic receptor agonist mirabegron in humanized mice induced thermogenesis and improved the Treg induction capacity of naïve T cells isolated from these animals. Using samples from the human FREECE study, we demonstrate that a short-term cold stimulus supports human Treg induction in vitro and in vivo. Mechanistically, we identify BORCS6 encoding the Ragulator-interacting protein C17orf59 to be significantly induced in human CD4+ T cells upon short-term cold exposure. Strong mTOR signaling is known to limit successful Treg induction and thus likely by interfering with mTOR activation at lysosomal surfaces, C17orf59 improves the Treg induction capacity of human naïve T cells upon cold exposure. CONCLUSIONS: These novel insights into the molecular underpinnings of human Treg induction suggest an important role of Tregs in linking environmental stimuli with adipose tissue function and metabolic diseases. Moreover, these discoveries shed new light on potential approaches towards tailored anti-inflammatory concepts that support human adipose tissue homeostasis by enabling Tregs.


Subject(s)
Cold Temperature , T-Lymphocytes, Regulatory/immunology , Acetanilides/pharmacology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/immunology , Adrenergic beta-Agonists/pharmacology , Adult , Animals , Female , Humans , Male , Mice , Middle Aged , Receptors, Adrenergic, beta/metabolism , T-Lymphocytes, Regulatory/drug effects , Thiazoles/pharmacology , Young Adult
11.
Cells ; 8(7)2019 06 30.
Article in English | MEDLINE | ID: mdl-31262098

ABSTRACT

Obesity is characterized by chronic and low-grade systemic inflammation, an increase of adipose tissue, hypertrophy, and hyperplasia of adipocytes. Adipose tissues can be classified into white, brown, beige and pink adipose tissues, which display different regulatory, morphological and functional characteristics of their adipocyte and immune cells. Brown and white adipocytes can play a key role not only in the control of energy homeostasis, or through the balance between energy storage and expenditure, but also by the modulation of immune and inflammatory responses. Therefore, brown and white adipocytes can orchestrate important immunological crosstalk that may deeply impact the tumor microenvironment and be crucial for cancer establishment and progression. Recent works have indicated that white adipose tissues can undergo a process called browning, in which an inducible brown adipocyte develops. In this review, we depict the mechanisms involved in the differential role of brown, white and pink adipocytes, highlighting their structural, morphological, regulatory and functional characteristics and correlation with cancer predisposition, establishment, and progression. We also discuss the impact of the increased adiposity in the inflammatory and immunological modulation. Moreover, we focused on the plasticity of adipocytes, describing the molecules produced and secreted by those cells, the modulation of the signaling pathways involved in the browning phenomena of white adipose tissue and its impact on inflammation and cancer.


Subject(s)
Adiposity/immunology , Carcinogenesis/immunology , Inflammation/immunology , Neoplasms/immunology , Obesity/immunology , Adipocytes, Brown/immunology , Adipocytes, Brown/metabolism , Adipocytes, White/immunology , Adipocytes, White/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , Carcinogenesis/pathology , Disease Models, Animal , Disease Progression , Energy Metabolism/immunology , Humans , Inflammation/metabolism , Inflammation/pathology , Neoplasms/metabolism , Neoplasms/pathology , Obesity/complications , Obesity/metabolism , Tumor Microenvironment/immunology
12.
Cell Metab ; 27(5): 954-961, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29719233

ABSTRACT

Immune cells were recently found to have an unexpected involvement in controlling the thermogenic activity of brown and beige adipose tissue. Here, we review how macrophages, eosinophils, type 2 innate lymphoid cells, and T lymphocytes are linked to this process. In particular, the recruitment of alternatively activated macrophages and eosinophils is associated with brown fat activation and white fat browning. Conversely, pro-inflammatory immune cell recruitment represses the thermogenic activity of brown and beige adipose tissues via cytokines that inhibit noradrenergic signaling. Macrophages also influence the noradrenergic tone by degrading norepinephrine locally and by inhibiting sympathetic innervation over time.


Subject(s)
Adipose Tissue, Beige/immunology , Adipose Tissue, Brown/immunology , Eosinophils/immunology , Macrophages/immunology , T-Lymphocytes/immunology , Thermogenesis/immunology , Adipocytes, Beige/cytology , Adipocytes, Beige/immunology , Adipocytes, Brown/cytology , Adipocytes, Brown/immunology , Adipose Tissue, Beige/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, White/immunology , Animals , Cytokines/immunology , Energy Metabolism , Humans , Mice , Norepinephrine/immunology
13.
J Physiol Biochem ; 74(3): 441-454, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29797227

ABSTRACT

Physical training (PT) has been considered as a treatment in metabolic syndrome (MS), since it induces thermogenic activity in brown (BAT) and white (WAT) adipose tissues. We evaluated the therapeutic effect of PT on activity of WAT and BAT in rats with MS induced by high-fat diet (30% lard) for 13 weeks and submitted, for the last 6 weeks, to swimming or kept sedentary (SED) rats. MS-SED rats compared to control diet (CT-SED) rats showed low physical fitness and high levels of glucose, insulin, homeostasis evaluation of insulin resistance (HOMA-IR), homeostasis evaluation of the functional capacity of ß-cells (HOMA-ß), and blood pressure. The gastrocnemius muscle decreased in peroxisome proliferator-activated receptor gamma coactivator 1-alpha and beta (PGC-1α, PGC-1ß), and uncoupled protein 2 and 3 (UCP2 and UCP3) expressions. Both WAT and BAT increased in the adipocyte area and decreased in blood vessels and fibroblast numbers. WAT increased in expression of pro-inflammatory adipokines and decreased in anti-inflammatory adipokine and adiponectin. WAT and gastrocnemius showed impairment in the insulin signaling pathway. In response to PT, MS rats showed increased physical fitness and restoration of certain biometric and biochemical parameters and blood pressure. PT also induced thermogenic modulations in skeletal muscle, WAT and BAT, and also improved the insulin signaling pathway. Collectively, PT was effective in treating MS by inducing improvement in physical fitness and interchangeable effects between skeletal muscle, WAT and BAT, suggesting a development of brown-like adipocyte cells.


Subject(s)
Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Adiposity , Insulin Resistance , Metabolic Syndrome/therapy , Physical Conditioning, Animal , Thermogenesis , Adipokines/genetics , Adipokines/metabolism , Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , Biomarkers/blood , Diet, High-Fat/adverse effects , Dietary Fats/adverse effects , Gene Expression Regulation , Hyperglycemia/etiology , Hyperglycemia/prevention & control , Hyperinsulinism/etiology , Hyperinsulinism/prevention & control , Hypertension/etiology , Hypertension/prevention & control , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Random Allocation , Rats, Inbred F344 , Weaning
14.
Parasite Immunol ; 40(2)2018 02.
Article in English | MEDLINE | ID: mdl-28423191

ABSTRACT

The identification, in the late 2000s, of innate lymphoid cells (ILCs) as a new class of non-B, non-T lymphocytes has led to global efforts to understand their functions, plasticity and evolutionary origins and to define their place within the leucocyte family. Although this work has uncovered striking similarities in the developmental cues, lineage-specific transcription factors and functional capacities of innate and adaptive lymphocytes, it has become clear that ILCs play a unique and defining role as stewards of barrier defence and that this sets them apart from their adaptive cousins. This review will explore how the dynamic environment of barrier surfaces has shaped ILC evolution and functionality. We highlight the critical importance of the microbiome and the unique role of ILCs as environmental sensors. We reflect on how these factors may have influenced the development of ILC2s and barrier immunity in the context of exposure to helminth parasites that have been driving forces of our evolution throughout human history. Finally, we argue that the plasticity of ILC function reflects their role as first responders to environmental change.


Subject(s)
Helminthiasis/immunology , Helminths/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Adipose Tissue, Brown/immunology , Animals , Cell Count , Cell Differentiation/immunology , Helminthiasis/parasitology , Humans , Lymphocytes/cytology , Microbiota/immunology , Transcription Factors
15.
Cell Mol Neurobiol ; 38(1): 349-361, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28801784

ABSTRACT

Continuous exposure to cold leads to activation of adaptive thermogenesis in brown adipose tissue but also to induction of brown/beige cell phenotype in white adipose tissue. The aim of this work was to investigate whether prior exposure to immobilization (IMO) stress may affect immune response associated with adipocyte "browning" in mesenteric adipose tissue (mWAT). In the first experiment, Sprague-Dawley rats were exposed to acute (3 h) or prolonged (7 days) cold exposure (4 ± 1 °C). 7-day cold stimulated gene expression of uncoupling protein 1 and other "browning"-associated factors. In the second experiment, rats were immobilized for 7 days (2 h daily) followed by exposure to continuous cold for 1 or 7 days. Prior IMO exaggerated cold-induced sympathetic response manifested by elevated tyrosine hydroxylase (TH) protein and norepinephrine in mWAT. Induction of non-sympathetic catecholamine production demonstrated by elevated TH and PNMT (phenylethanolamine N-methyltransferase) mRNAs was observed after 7-day cold; however, prior IMO attenuated this response. 7-day cold-induced gene expression of anti-inflammatory mediators (IL-4, IL-13, IL-10, adiponectin), markers of M2 macrophages (Arg1, Retnlα), and eosinophil-associated molecules (eotaxin, IL-5), while inhibited expression of pro-inflammatory cytokines (IFNγ, IL-1b, IL-6, IL-17) and monocytes (MCP-1, Ly6C). This immune response was accompanied by elevated expression of uncoupling protein-1 and other thermogenic factors. Rats exposed to prior IMO exhibited inhibition of cold-induced immune and "browning"-related expression pattern. Overall, we demonstrated that 7-day cold-induced browning"-associated changes in rat mWAT, while prior history of repeated stress prevented this response.


Subject(s)
Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Cold Temperature , Immunomodulation/physiology , Stress, Psychological/immunology , Stress, Psychological/metabolism , Animals , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Male , Rats , Rats, Sprague-Dawley
16.
Sci Rep ; 7(1): 16082, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29167565

ABSTRACT

Obesity is associated with severe metabolic diseases such as type 2 diabetes, insulin resistance, cardiovascular disease and some forms of cancer. The pathophysiology of obesity-induced metabolic diseases has been strongly related to white adipose tissue (WAT) dysfunction through several mechanisms such as fibrosis, apoptosis, inflammation, ER and oxidative stress. However, little is known of whether these processes are also present in brown adipose tissue (BAT) during obesity, and the potential consequences on mitochondrial activity. Here we characterized the BAT of obese and hyperglycemic mice treated with a high-fat diet (HFD) for 20 weeks. The hypertrophic BAT from obese mice showed no signs of fibrosis nor apoptosis, but higher levels of inflammation, ER stress, ROS generation and antioxidant enzyme activity than the lean counterparts. The response was attenuated compared with obesity-induced WAT derangements, which suggests that BAT is more resistant to the obesity-induced insult. In fact, mitochondrial respiration in BAT from obese mice was enhanced, with a 2-fold increase in basal oxygen consumption, through the upregulation of complex III of the electron transport chain and UCP1. Altogether, our results show that obesity is accompanied by an increase in BAT mitochondrial activity, inflammation and oxidative damage.


Subject(s)
Adipose Tissue, Brown/pathology , Inflammation/pathology , Mitochondria/metabolism , Oxidative Stress , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Animals , Antioxidants/metabolism , Cell Respiration , Diet, High-Fat , Endoplasmic Reticulum Stress , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/pathology , Phenotype , Reactive Oxygen Species/metabolism , Uncoupling Protein 1/metabolism
17.
EBioMedicine ; 24: 127-136, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29032027

ABSTRACT

Brown adipose tissues (BAT) burn lipids to generate heat through uncoupled respiration, thus representing a powerful target to counteract lipid accumulation and obesity. The tumor suppressor liver kinase b1 (Lkb1) is a key regulator of cellular energy metabolism; and adipocyte-specific knockout of Lkb1 (Ad-Lkb1 KO) leads to the expansion of BAT, improvements in systemic metabolism and resistance to obesity in young mice. Here we report the unexpected finding that the Ad-Lkb1 KO mice develop hindlimb paralysis at mid-age. Gene expression analyses indicate that Lkb1 KO upregulates the expression of inflammatory cytokines in interscapular BAT and epineurial brown adipocytes surrounding the sciatic nerve. This is followed by peripheral neuropathy characterized by infiltration of macrophages into the sciatic nerve, axon degeneration, reduced nerve conductance, and hindlimb paralysis. Mechanistically, Lkb1 KO reduces AMPK phosphorylation and amplifies mammalian target-of-rapamycin (mTOR)-dependent inflammatory signaling specifically in BAT but not WAT. Importantly, pharmacological or genetic inhibition of mTOR ameliorates inflammation and prevents paralysis. These results demonstrate that BAT inflammation is linked to peripheral neuropathy.


Subject(s)
Adipose Tissue, Brown/immunology , Paraplegia/pathology , Peripheral Nervous System Diseases/pathology , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Adenylate Kinase/metabolism , Adipose Tissue, Brown/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Humans , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Paraplegia/genetics , Paraplegia/immunology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/immunology , Phosphorylation , Sciatic Nerve/immunology , Up-Regulation
18.
J Nutr Biochem ; 49: 15-21, 2017 11.
Article in English | MEDLINE | ID: mdl-28863365

ABSTRACT

Fat browning has emerged as an attractive target for the treatment of obesity and related metabolic disorders. Its activation leads to increased energy expenditure and reduced adiposity, thus contributing to a better energy homeostasis. Green tea extracts (GTEs) were shown to attenuate obesity and low-grade inflammation and to induce the lipolytic pathway in the white adipose tissue (WAT) of mice fed a high-fat diet. The aim of the present study was to determine whether the antiobesity effect of an extract from green tea leaves was associated with the activation of browning in the WAT and/or the inhibition of whitening in the brown adipose tissue (BAT) in HF-diet induced obese mice. Mice were fed a control diet or an HF diet supplemented with or without 0.5% polyphenolic GTE for 8 weeks. GTE supplementation significantly reduced HF-induced adiposity (WAT and BAT) and HF-induced inflammation in WAT. Histological analysis revealed that GTE reduced the adipocyte size in the WAT and the lipid droplet size in the BAT. Markers of browning were induced in the WAT upon GTE treatment, whereas markers of HF-induced whitening were reduced in the BAT. These results suggest that browning activation in the WAT and whitening reduction in the BAT by the GTE could participate to the improvement of metabolic and inflammatory disorders mediated by GTE upon HF diet. Our study emphasizes the importance of using GTE as a nutritional tool to activate browning and to decrease fat storage in all adipose tissues, which attenuate obesity.


Subject(s)
Adipose Tissue, Brown/pathology , Anti-Obesity Agents/therapeutic use , Camellia sinensis/chemistry , Dietary Supplements , Obesity/prevention & control , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Adipogenesis , Adipose Tissue, Beige/immunology , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/pathology , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity , Animals , Biomarkers/metabolism , Cell Size , Diet, High-Fat/adverse effects , Food Handling , Lipid Droplets/immunology , Lipid Droplets/metabolism , Lipid Droplets/pathology , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Oxidation-Reduction , Polyphenols/therapeutic use , Random Allocation , Specific Pathogen-Free Organisms
19.
Vet Pathol ; 54(6): 885-891, 2017 11.
Article in English | MEDLINE | ID: mdl-28812533

ABSTRACT

Liposarcoma, rhabdomyosarcoma, and hibernoma share some overlapping histologic and immunohistochemical features. Although immunohistochemistry (IHC) is commonly used in the diagnosis of these neoplasms, expression of muscle markers has been reported in human liposarcoma and canine hibernoma in addition to rhabdomyosarcoma. Thus, these neoplasms are a diagnostic challenge but important to distinguish because of differences in prognosis and treatment. Rhabdomyosarcoma and liposarcoma are both malignant, but rhabdomyosarcoma has a higher potential for metastasis. In contrast, hibernomas are benign with low risk of recurrence. This study investigated expression of the muscle markers desmin, myogenin, and α-smooth muscle actin (α-SMA) and the brown fat marker uncoupling protein 1 (UCP1) in 25 cases of canine liposarcoma using IHC. Oil red O histochemistry was performed to confirm the presence of lipid and the diagnosis of liposarcoma in cases that were not well-differentiated. The 25 cases included 15 well-differentiated, 5 pleomorphic, 3 myxoid, and 2 dedifferentiated subtypes of liposarcoma. By IHC, 23 of 25 expressed UCP1, 7 of 25 expressed α-SMA, 7 of 25 expressed desmin, and 3 of 25 expressed myogenin with no clear relationship of antigen expression and tumor subtype. These findings clarify the immunohistochemical profile of canine liposarcoma and suggest overlap in the expression of several muscle antigens and UCP1 between liposarcoma, hibernoma, and rhabdomyosarcoma.


Subject(s)
Antigens/immunology , Biomarkers, Tumor/immunology , Dog Diseases/immunology , Liposarcoma/veterinary , Actins/immunology , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Animals , Desmin/immunology , Dog Diseases/pathology , Dogs , Female , Immunohistochemistry/veterinary , Liposarcoma/immunology , Liposarcoma/pathology , Male , Muscle, Smooth/metabolism , Myogenin/immunology , Uncoupling Protein 1/immunology
20.
J Mol Endocrinol ; 59(3): 245-255, 2017 10.
Article in English | MEDLINE | ID: mdl-28694301

ABSTRACT

Brown adipose tissue (BAT) activation and white adipose tissue (WAT) beiging can increase energy expenditure and have the potential to reduce obesity and associated diseases. The immune system is a potential target in mediating brown and beige adipocyte activation. Type 2 and anti-inflammatory immune cells contribute to metabolic homeostasis within lean WAT, with a prominent role for eosinophils and interleukin (IL)-4-induced anti-inflammatory macrophages. We determined eosinophil numbers in epididymal WAT (EpAT), subcutaneous WAT (ScAT) and BAT after 1 day, 3 days or 1 week of high-fat diet (HFD) feeding in C57Bl/6 mice. One day of HFD resulted in a rapid drop in eosinophil numbers in EpAT and BAT, and after 3 days, in ScAT. In an attempt to restore this HFD-induced drop in adipose tissue eosinophils, we treated 1-week HFD-fed mice with helminth antigens from Schistosoma mansoni or Trichuris suis and evaluated whether the well-known protective metabolic effects of helminth antigens involves BAT activation or beiging. Indeed, antigens of both helminth species induced high numbers of eosinophils in EpAT, but failed to induce beiging. In ScAT, Schistosoma mansoni antigens induced mild eosinophilia, which was accompanied by slightly more beiging. No effects were observed in BAT. To study type 2 responses on brown adipocytes directly, T37i cells were stimulated with IL-4. This increased Ucp1 expression and strongly induced the production of eosinophil chemoattractant CCL11 (+26-fold), revealing that brown adipocytes themselves can attract eosinophils. Our findings indicate that helminth antigen-induced eosinophilia fails to induce profound beiging of white adipocytes.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/metabolism , Antigens, Helminth/immunology , Diet, High-Fat , Eosinophils/immunology , Eosinophils/metabolism , Adipocytes/metabolism , Adipose Tissue/pathology , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Animals , Biomarkers , Chemokine CCL11/biosynthesis , Immunity , Interleukin-4/metabolism , Leukocyte Count , Male , Mice , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...