Volume 26, Issue 3 p. 547-558
Original Article
Free Access

Dietary Curcumin Intervention Targets Mouse White Adipose Tissue Inflammation and Brown Adipose Tissue UCP1 Expression

Zhuolun Song

Zhuolun Song

Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada

Department of Physiology, University of Toronto, Toronto, Ontario, Canada

Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada

Search for more papers by this author
Xavier Revelo

Xavier Revelo

Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada

Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada

Search for more papers by this author
Weijuan Shao

Weijuan Shao

Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada

Search for more papers by this author
Lili Tian

Lili Tian

Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada

Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada

Search for more papers by this author
Kejing Zeng

Kejing Zeng

Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada

Search for more papers by this author
Helena Lei

Helena Lei

Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada

Search for more papers by this author
Hong-Shuo Sun

Hong-Shuo Sun

Department of Physiology, University of Toronto, Toronto, Ontario, Canada

Department of Surgery, University of Toronto, Toronto, Ontario, Canada

Search for more papers by this author
Minna Woo

Minna Woo

Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada

Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada

Division of Endocrinology and Metabolism, Department of Medicine, University Health Network, Toronto, Ontario, Canada.

Search for more papers by this author
Daniel Winer

Daniel Winer

Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada

Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada

Search for more papers by this author
Tianru Jin

Corresponding Author

Tianru Jin

Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada

Department of Physiology, University of Toronto, Toronto, Ontario, Canada

Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada

Correspondence: Tianru Jin ([email protected])Search for more papers by this author
First published: 06 February 2018
Citations: 58

Funding agencies: This work was supported by an operating grant by the Canadian Institutes of Health Research (MOP 97790) to TJ. ZS is supported by a Banting and Best Diabetes Centre (BBDC) graduate studentship, and LT is supported by a BBDC postdoctoral fellowship. KZ is the recipient of the Scholarship for International Program for PhD candidates, Sun Yat-Sen University.

Disclosure: The authors declared no conflict of interest.

Author contributions: ZS, XR, WS, LT, KZ, HL, DW, and TJ: experimental data and study design. HS: assistance on rectal temperature recording. MW: assistance on mouse metabolic cage analyses. ZS and TJ: writing of the manuscript. XR, MW, and DW: editing of the manuscript. TJ is the guarantor of this work and, as such, had full access to all the data in this study and takes responsibility for the integrity of data and the accuracy of the data analyses.

Current address: Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.

Abstract

Objective

This study aimed to determine whether dietary curcumin intervention targets both white adipose tissue (WAT) inflammation and brown adipose tissue (BAT)-mediated energy expenditure.

Methods

C57BL/6J mice were fed with a low-fat diet, high-fat diet (HFD), or HFD plus curcumin. In addition to assessing the effect of curcumin intervention on metabolic profiles, this study assessed WAT macrophage infiltration and composition and inflammatory cytokine production. Metabolic cages were applied for determining energy expenditure. Raw264.7 (ATCC, Manassas, Virginia) and other cell models were utilized to test the in vitro effect of curcumin treatment.

Results

Curcumin intervention reduced WAT macrophage infiltration and altered macrophage functional polarity, as the ratio of M2-like versus M1-like macrophages increased after curcumin intervention. Curcumin treatment reduced M1-like macrophage markers or proinflammation cytokine expression in both macrophages and adipocytes. Curcumin intervention also increased energy expenditure and body temperature in response to a cold challenge. Finally, the in vivo and in vitro investigations suggested that curcumin increased expression of uncoupling protein 1 (UCP1), possibly involving PPAR-dependent and -independent mechanisms.

Conclusions

Curcumin intervention targets both WAT inflammation and BAT UCP1 expression. These observations advanced our knowledge on the metabolic beneficial effects of the curry compound curcumin, bringing us a novel perspective on dietary polyphenol research.

Introduction

Obesity is associated with type 2 diabetes (T2D), insulin resistance, and other metabolic disorders (1). Following the recognition that proinflammatory cytokines expressed by white adipose tissues (WAT) play direct roles in obesity-induced insulin resistance (2), great efforts have been made to explore mechanistic insights into how adipose tissue inflammation contributes to the development of obesity-induced metabolic disorders, including the clarification that WAT macrophage infiltration can be induced by high-calorie food intake (3).

Intensive investigations have revealed the existence of a full spectrum of macrophage activations in WAT and elsewhere (4). The differentiation of the M1-like macrophages, or classically activated macrophages, is mainly accelerated by proinflammatory factors, such as lipopolysaccharide (LPS) and interferon γ (IFNγ). The M1-like macrophages express proinflammatory cytokines, such as interleukin 1 (IL-1), IL-6, and monocyte chemoattractant protein 1 (MCP-1). On the other hand, the differentiation of M2-like macrophages or alternatively activated macrophages can be induced by IL-4, IL-10, or IL-13, and they express certain anti-inflammatory cytokines (5). Targeting inflammation in adipose tissues and elsewhere with IL-1β or tumor necrosis factor (TNF) antagonists or antibodies has been tested in both animal models and in clinical trials (6).

The development of obesity also reflects the existence of impaired energy homeostasis, while increased energy expenditure leads to reduced energy deposition and adipose tissue expansion. Recent studies have defined the role of the uncoupling protein 1 (UCP1) located in the mitochondria inner membrane, mostly found in brown adipose tissue (BAT), that mediates thermogenesis in response to cold exposure and other environmental changes (7). The upregulation of UCP1 production or function and WAT “browning” are potential novel therapeutic and preventive approaches for obesity and its related metabolic disorders (8).

Dietary interventions are alternative and promising tools for preventing or even treating obesity and its related metabolic disorders (9, 10). Previous rodent studies have attributed these effects of dietary polyphenols mainly to their anti-inflammation and antioxidation properties, secondary to their body weight lowering effect. We reported recently that one of these polyphenols, the curry compound curcumin, could improve insulin signaling in an anti-inflammation independent manner (11). Furthermore, curcumin and resveratrol were shown to stimulate the production and function of hepatic hormone fibroblast growth factor 21 (Fgf21) (12, 13).

The current study aimed to determine whether dietary polyphenols, such as curcumin, can exert the metabolic beneficial effects via targeting both WAT inflammation and BAT-mediated energy expenditure. We show here that dietary curcumin intervention in high-fat diet (HFD)-fed mice reduced not only WAT macrophage infiltration, but also the ratio of WAT M1-like macrophages versus M2-like macrophages. We did not observe the effect of dietary curcumin intervention on subcutaneous adipocyte browning, reported by another group in mice without HFD feeding (14), but we found that curcumin intervention increased BAT UCP1 production.

Methods

Materials

Escherichia coli LPS, peroxisome proliferator-activated receptor-gamma (PPARγ) antagonist GW9662 (M6191), and agonist rosiglitazone (R2408) were purchased from Sigma-Aldrich (L2880; Sigma-Aldrich Corp., St. Louis, Missouri). Murine IFNγ (AF-315-05) was purchased from Peprotech (Quebec, Canada). Human recombinant FGF21 was the product of Novoprotein Scientific Inc. (Summit, New Jersey). PPARα antagonist GW6471 (G5045) and agonist Wy14643 (C7081) were purchased from Santa Cruz Biotechnology (Santa Cruz, California). Curcumin was purchased from Sigma-Aldrich for cell culture experiments or from Organika Health Products (the 95% standardized curcumin extract; Richmond, British Columbia, Canada) for dietary intervention as we have documented previously (12).

Experimental animals

Male C57BL/6J mice (Charles River Laboratories, St. Laurent, Quebec, Canada) were housed under the conditions of a constant temperature (22°C, unless otherwise specified) and a 12-hour light/dark shift with free access to food and water.

For the experiments illustrated in Figure 1A, 45 mice were randomly divided into the following three groups: fed with the chow diet (low-fat diet [LFD], 3.6 kcal/g), HFD (Product# F3282 with 60% fat calories and 5.49 kcal/g; Bio-Serv, Flemington, New Jersey), or HFD plus 1% curcumin. Body weight and food intake were recorded weekly. Intraperitoneal glucose, insulin, and pyruvate tolerance tests were performed at indicated time points (Figure 1A) as we have previously described (15). Mice were sacrificed at the 18th week for blood and tissue collection.

Details are in the caption following the image

Long-term dietary curcumin intervention attenuates HFD-induced body weight gain and improves metabolic profiles. (A) Illustration of experimental procedures. (B) Body weight recording for the first 12 weeks (stopped when metabolic tolerance tests were started). Right panel shows areas under the curve (AUC) (n = 15 per each of the three groups). (C-E) Results of intraperitoneal glucose (i.p.GTT; n = 6), insulin (i.p.ITT; n = 3), and pyruvate tolerance tests (i.p.PTT; n = 4). (F) Pyruvate results presented as relative percentage declines, with the glucose level prior to insulin injection as 100%. (G) Fasting blood glucose levels (n = 5). (H) Random plasma insulin concentrations (n = 5). (I) Random plasma leptin concentrations (n = 5). (J) Epididymal adipose tissue Fgf21 mRNA expression levels (n = 6). *P < 0.05, **P < 0.01, ***P < 0.001. For panel B, *, HFD vs. Cur; #, LFD vs. HFD; P < 0.05. [Color figure can be viewed at wileyonlinelibrary.com]

For the experiments listed in Figure 5A, 24 mice were randomly divided into the following three groups: fed with the LFD, HFD, or HFD plus 1% curcumin. The cold challenge was performed after 2, 4, or 6 weeks after corresponding diet feeding. Mice were kept at room temperature prior to being put in a 4°C environmental chamber. Four hours later, rectal temperature recording was started by using an electronic thermometer equipped with a rectal probe (HH63K Stick Type Temperature Transducer and Thermometer; Omega, Quebec, Canada).

For metabolic cage analysis, mice fed HFD or HFD plus curcumin were housed individually in metabolic cages with sufficient food and water. After a 24-hour settlement period, data collection was started. Collected data for the 30-hour period were analyzed by using a lab animal monitoring system (Columbus Instruments Corp., Columbus, Ohio) as previously presented (16).

The animal experiments and protocols were approved by the University Health Network Animal Care Committee and performed in accordance with the guidelines of the Canadian Council of Animal Care.

Cell cultures and luciferase reporter analysis

The isolation of primary adipocytes from male Sprague Dawley rat (Charles River Laboratories) epididymal fat tissues has been previously described (17).

The mouse leukemic monocyte-macrophage Raw264.7 cell line was purchased from ATCC (Manassas, Virginia). M1-like macrophage differentiation was induced by LPS (100 ng/mL) and IFNγ (2.5 ng/mL).

The mouse brown adipocyte cell line (mBAC) was the gift of Shingo Kajimura (18). Cells were cultured with the growth medium (DMEM with 10% FBS). The differentiation medium contained the growth medium and the cocktail (5 µg/mL insulin, 1 nmol/L T3, 0.123 mmol/L indomethacin, 2 µg/mL dexamethasone, and 0.1 mmol/L IBMX).

For mouse peritoneal macrophage isolation, male C57BL/6J mice (8 weeks old) were injected intraperitoneally with 4% thioglycollate (1 mL per mouse). Mice were sacrificed 4 days later for peritoneal macrophage isolation.

The Ucp1-luciferase (LUC) reporter gene construct was described previously by the provider, in which the expression of the LUC reporter is driven by the 3.1-kilobase mouse Ucp1 gene 5' flanking region (19). The LUC assay was conducted as previously described (12).

Histology and immunohistochemistry

Mouse brown, epididymal (WAT), and inguinal (subcutaneous) adipose tissues were fixed in 4% paraformaldehyde followed by paraffin embedding. Tissue sections were stained with hematoxylin and eosin or immune-stained with the UCP1 antibody (15).

Processing of immune cells from epididymal adipose tissue and spleen

Epididymal adipose tissue immune cells were isolated as previously described (20). The cell culture medium was collected for the Luminex multiplex assay (Thermo Fisher Scientific, Waltham, Massachusetts), conducted by the Service Laboratories of University Health Network.

Flow cytometry

Immune cells were stained for 30 minutes with fluorophore-conjugated antibodies against CD206, CD11c, CD86, and CD80 by using recommended dilutions. Cells were identified by using an LSRFortessa cell analyzer (BD Biosciences, Mountain View, California). Data acquired on the flow cytometer were analyzed with FlowJo software (Tree Star; FlowJo LLC, Ashland, Oregon).

Western blotting and real-time reverse transcription polymerase chain reaction

Protein samples were subjected to Western blotting with antibodies against inducible nitric oxide synthase (iNOS) (13120S; Cell Signaling Technology Inc., Danvers, Massachusetts), UCP1 (Cell Signaling 14670S), or β-actin (Cell Signaling 3700S). Methods for RNA extraction, complementary DNA production, and real-time reverse transcription-polymerase chain reaction (RT-PCR) analyses have been described previously (15). RT-PCR primers are listed in Supporting Information Table S1.

Statistical analysis

All data were expressed as the mean ± SEM. Comparisons between groups were performed by using either a two-tailed t test or a one-way analysis of variance (ANOVA). P < 0.05 was considered significant.

Results

Curcumin intervention improves metabolic profiles

We first assessed the effect of curcumin intervention in HFD-fed mice during the 17-week period, as illustrated in Figure 1A. The body weight difference between HFD- and LFD-fed mice started at the third week, while the attenuation effect of curcumin intervention on HFD-induced body weight gain started at the fifth week (Figure 1B). During the first 12-week period, total calorie intake for mice on an HFD was higher than for mice on an LFD, while no difference was found for HFD-fed mice with or without curcumin intervention (Supporting Information Figure S1a-S1b). Supporting Information Figure S1c shows the weekly calorie intake normalized by body weight. Results of glucose, pyruvate, and insulin tolerance tests among the three groups of mice performed at indicated time points (Figure 1A) are shown in Figure 1C-1F. These results, along with the attenuation of hyperglycemia, hyperinsulinemia, and hyperleptinemia (Figure 1G-1I) with curcumin intervention, allowed us to conclude that curcumin administration improved insulin signaling and metabolic profiles, which were impaired by HFD consumption.

We observed very recently that HFD consumption increased plasma Fgf21 levels, while curcumin intervention blocked the increase and improved hepatocyte Fgf21 sensitivity (12). We show here that HFD consumption also increased epididymal fat tissue Fgf21 levels, and the increase was attenuated by curcumin intervention (Figure 1J).

Curcumin intervention alters macrophage functional polarity

We then assessed the effect of curcumin intervention on adipose tissues. HFD-induced adipose tissue and liver weight gains were attenuated by curcumin intervention (Figure 2A-2B). We then compared epididymal fat tissue macrophage volumes and the compositions in HFD-fed mice in the presence and absence of curcumin intervention. LFD-fed mice were not included as there were very few infiltrated macrophages in those mice for conducting our assays. Figure 2C-2G shows that curcumin intervention not only reduced WAT macrophage volume, but also reduced the percentages of M1-like macrophages and increased the percentages of M2-like macrophages. Such effects, however, were not observed in the spleen tissue (Figure 2H). Table 1 shows that curcumin intervention altered the levels of a panel of cytokines secreted by infiltrated immune cells in WAT. The stimulation of the anti-inflammatory cytokine IL-10, but not IL-4 and IL-13, was also observed. Evidently, dietary curcumin intervention reduced the mean fluorescence intensity (MFI) in WAT CD86 on macrophages but not CD80 cells (Figure 2I-2J). CD86 is expressed in antigen-presenting cells. Reduced CD86 MFI suggests decreased activation of WAT immune cells (3). We therefore suggest that curcumin intervention not only reduces WAT macrophage infiltration, but it also alters WAT macrophage polarity.

Details are in the caption following the image

Long-term curcumin intervention alters macrophage functional polarity. (A-B) Adipose tissue and liver weight (n = 8). (C) Epididymal adipose tissue total macrophage counts. (D-E) M1-like and M2-like macrophage percentages. (F-G) Flow cytometer data for M1 and M2 macrophages. Cells outside of the marked areas are considered CD11c and CD206 negative cells. (H) Lack of the effect of curcumin intevention on the control spleen tissue macrophage mass as well as M1-like and M2-like macrophage percentages. (I-J) CD86 or CD80 MFI. n = 4 for panels C-J. *, **, or ***, P < 0.05 between indicated groups. n.s., nonsignificant. [Color figure can be viewed at wileyonlinelibrary.com]

Table 1. Luminex multiplex assays for immune cell cytokines
HFD Significance Cur
IL-1α 28.33833 ± 3.081949 * 3.633333 ± 1.478967
IL-1β 358.23 ± 19.97456 * 83.24 ± 8.302201
IL-2 7.406667 ± 0.44464 N/A OOR<
IL-3 9.308333 ± 1.10071 ** 2.373333 ± 0.280139
IL-4 OOR< N/A OOR<
IL-5 1,598.705 ± 431.1624 * 45.53 ± 2.05052
IL-6 OOR> N/A 1,477.053 ± 130.1968
IL-9 OOR< N/A OOR<
IL-10 44.24667 ± 12.82369 *** 187.19333 ± 28.88765
IL-12(p40) 57.05333 ± 3.525397 * 22.48 ± 4.516868
IL-12(p70) 84.64167 ± 7.885371 * 27.99 ± 2.452393
IL-13 296.6633 ± 6.593959 ns 247.7433 ± 21.02634
IL-17 26.845 ± 9.642319 N/A OOR<
Eotaxin 452.095 ± 133.0914 N/A OOR<
G-CSF 35,136.61 ± 4,813.975 ** 1,102.503 ± 556.6802
GM-CSF 384.285 ± 121.4681 N/A OOR<
IFNγ 6.795 ± 1.041277 N/A OOR<
KC 22,473.01 ± 3,837.754 ns 19,687.42 ± 1,230.979
MCP-1 OOR> N/A 6,338.84 ± 1,519.185
MIP-1α 5,832.663 ± 980.4733 ** 178.5233 ± 47.066
MIP-1β 2,084.3 ± 380.2988 *** 51.14667 ± 9.034167
RANTES 5,716.025 ± 1,598.435 * 74.58333 ± 27.12339
TNFα 172.775 ± 9.916168 ** 119.4033 ± 5.524269
  • *, **, or ***, HFD vs. Cur, P < 0.05.
  • Data are mean ± SD. Unit picograms per milliliter.
  • OOR>, out of range above; OOR<, out of range below; Cur, curcumin; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; ns, nonsignificant.

In vitro curcumin treatment attenuates Raw264.7 macrophage M1-like cell differentiation

It has been reported previously that curcumin treatment affects the polarization of the murine Raw264.7 macrophages (21). We assessed the direct effect of curcumin treatment in vitro in this monocyte-macrophage cell model with the dosages of curcumin that generated no appreciable inhibition on cell viability (Supporting Information Figure S2). Indeed, LPS/IFNγ treatment in this cell model induced the dendritic morphological change, while curcumin treatment reversed the effect of LPS/IFNγ (Supporting Information Figure S3). Curcumin treatment also dose-dependently attenuated the stimulatory effect of LPS/IFNγ on iNOS protein levels (Figure 3A). Figure 3B-3D shows that LPS/IFNγ-induced expression of iNOS, MCP-1, and Il-6 messenger RNA (mRNA) (differentiation markers of the M1-like subtype) can be dose-dependently inhibited by curcumin treatment. The stimulation of the differentiation markers of the M2-like subtype (KFL4, PPARγ, and ARG1) as well as the anti-inflammation cytokine gene IL-4, however, was observed only when the dosage of curcumin reached 20 μM (Figure 3E-3H). Such a high dosage is unlikely reached in the in vivo settings. When mouse peritoneal macrophages were treated with 0.25 to 2 μM curcumin for 10 hours, we also did not observe the stimulation of M2-like subtype differentiation marker expression (Supporting Information Figure S4).

Details are in the caption following the image

Curcumin treatment attenuates Raw264.7 M1-like cell differentiation. (A) iNOS protein detection by Western blotting. Right panel shows the result of densitometric analyses. (B-D) M1-like macrophage marker detection by real-time RT-PCR. (E-H) M2-like macrophage marker detection. (I) M1-like marker detection in Raw264.7 cells incubated with rat adipocyte conditioned medium for 10 hours. (J) Detection of the expression of proinflammatory cytokine genes and (K) an anti-inflammatory cytokine gene (IL-4) in rat adipocytes that received indicated dose of curcumin treatment for 10 hours. L/I, LPS/IFNγ (M1-like subtype inducer). n = 3 for panels A-K. For panels A-H, *, **, or ***, Cur vs. L/I only; ## or ###, DMSO vehicle control vs. L/I; P < 0.05. For panels I-K, *, **, or ***, P < 0.05 between indicated groups. [Color figure can be viewed at wileyonlinelibrary.com]

Adipocytes can produce cytokines that may affect macrophage activity and differentiation (22, 23). We performed the in vitro assay, asking whether mature adipocyte-secreted cytokines affect the differentiation of Raw264.7 cells. Figure 3I shows that rat adipocyte conditioned cell culture medium also increased expression of the M1-like subtype markers in the Raw264.7 cell model. When rat adipocytes were treated with curcumin for 10 hours, the expression of a battery of proinflammatory cytokine genes was reduced, while the expression of the anti-inflammatory cytokine IL-4 was increased (Figure 3J-3K). We therefore suggest that in addition to the regulation of macrophage differentiation directly, curcumin can also inhibit the expression of proinflammatory cytokines produced by adipocytes, indirectly repressing M1 macrophage differentiation.

We reported very recently that in vitro curcumin treatment increased Fgf21 expression in mouse hepatocytes (12). Here, we show that curcumin also stimulated Fgf21 mRNA expression in rat primary adipocytes (Supporting Information Figure S5a). In vitro treatment with human recombinant FGF21, however, did not reduce the expression of the differentiation markers of the M1-like subtype in Raw264.7 cells (Supporting Information Figure S5b-S5e).

Dietary curcumin intervention increases energy expenditure and the thermogenic capacity

Mice fed with an HFD without or with curcumin intervention at the 16th week were subjected to metabolic cage analyses. Curcumin intervention was shown to increase CO2 production, O2 consumption, and energy expenditure (Figure 4A-4C), regardless of whether the data were expressed per animal or per body weight of the animals. During the metabolic cage analysis period, there was a trend of increased on respiratory exchange ratio (RER) in mice that received curcumin intervention (Figure 4D-4E). When we separated the data into the dark and the light cycles, a significant increase in RER was found in mice with curcumin intervention in the dark cycle but not in the light cycle (Figure 4F-4G). Increased RER usually suggests that the mice utilize relatively more carbohydrates as the source of energy (24, 25). How this can be found in curcumin intervention treated mice with improved insulin signaling, associated with increased energy expenditure, is an open question for further investigations.

Details are in the caption following the image

Long-term curcumin intervention increases energy expenditure. (A) Metabolic cage analyses of CO2 production, (B) O2 consumption, (C) and energy expenditure (EE). Data presented as absolute value, without the normalization against mouse body weight (left panels of A-C) or with the normalization against mouse body weight (right panels of A-C). (D) Diagram of RER generated by metabolic cage analyses. (E) Overall RER. (F-G) RER during the dark cycle and light cycles. (H) Food intake and (I) physical activity measured by metabolic cage analyses during the 30-hour period. n = 4 for panels A-H. *, **, or ***, P < 0.05 between indicated groups. n.s., nonsignificant. [Color figure can be viewed at wileyonlinelibrary.com]

To further verify the stimulatory effect of curcumin intervention on thermogenesis, we conducted another set of mouse experiments with a relatively short-term curcumin intervention, illustrated in Figure 5A. Mice were housed at room temperature (∼22oC) before being placed in a 4oC cold chamber. Two, four, and six weeks after the corresponding diet consumption, mice were subjected to cold exposure. Four hours after the mice were in the cold chamber, we started rectal temperature recording. As shown in Figure 5B-5D, curcumin intervention increased the thermogenic capacity during the cold exposure process, starting as early as 2 weeks after curcumin intervention. We noticed that the significant temperature differences appeared mostly during the dark periods. The differences, however, were not observed for these three groups of mice in the absence of the cold challenge (Supporting Information Figure S6).

Details are in the caption following the image

Curcumin intervention increases thermogenesis in response to cold exposure. (A) Illustration of experimental procedure for rectal temperature recording. (B-C) Results of rectal temperature monitoring during the 48-hour or (D) 72-hour cold challenge (4oC) period. n = 5 for panels B-D. In each experiment, mice were moved from the cage at room temperature (∼22oC) to the cold chamber (4oC) for 4 hours before starting rectal temperature recording. * or **, HFD vs. Cur, P < 0.05. AUC, area under curve. [Color figure can be viewed at wileyonlinelibrary.com]

Curcumin intervention increases BAT UCP1 expression

An improved ability of the mice to handle a cold challenge could be due to the increase of shivering thermogenesis, the increase of BAT-mediated nonshivering thermogenesis, or both (26-28). As curcumin intervention was shown to increase energy expenditure in our metabolic cage analysis, and curcumin gavage was shown to stimulate the inguinal adipocyte “browning” that was reported by another team (14), we asked whether long-term (17 weeks) curcumin intervention affects UCP1 expression and subcutaneous (inguinal) WAT browning. We did not see changes in UCP1 expression in subcutaneous adipose tissues after curcumin intervention and could not detect their UCP1 expression by Western blotting (Supporting Information Figure S7a-S7b). In addition, mice that received the 6-week curcumin intervention showed elevated BAT but not inguinal UCP1 expression (Supporting Information Figure S7c-S7d). We did find the “browning-like clusters” within the inguinal adipose tissue in mice that received curcumin intervention, but the numbers were few. Nevertheless, the 17-week dietary curcumin intervention reduced the sizes of adipocytes in both inguinal and epididymal adipose tissues (Supporting Information Figure S7e-S7h).

Curcumin intervention appears to reduce BAT adipocyte size, which is associated with increased UCP1 mRNA and protein expression (Figure 6A-6D). In BAT, increased UCP1 and Prdm16 (which encodes the protein PR domain containing 16) expression in response to long-term curcumin intervention was associated with increased PPARα but not PPARγ or PPARγ coactivator (PGC-1α) (Figure 6C), while in rat mature adipocytes, in vitro curcumin treatment increased UCP1 mRNA levels, which were associated with PPARα and PGC-1α elevation (Figure 6E). As PPARs are known transcriptional activators of UCP1 (29-31), the above observation indicates that curcumin may directly stimulate UCP1 transcription via PPAR activation. To examine this, we transfected the UCP-1/LUC fusion gene plasmid (19) into the 293T cells, followed by curcumin treatment in the absence and presence of PPARα or PPARγ antagonist. Figure 6F shows that curcumin or the PPARα agonist WY14643 treatment increased UCP1 promoter activity, while the PPARα antagonist GW6471 inhibited its activity. The stimulatory effect of curcumin, however, was not blocked by GW6471 pretreatment. Figure 6G shows that PPARγ agonist rosiglitazone also stimulated UCP1 promoter activity, while the stimulatory effect of curcumin was not blocked by the PPARγ antagonist GW9662. These observations suggest that curcumin can stimulate UCP1 expression via PPARα and PPARγ, while the existence of PPARα- and PPARγ-independent mechanisms cannot be excluded.

Details are in the caption following the image

Long-term dietary curcumin intervention increases BAT UCP1 expression. (A-B) BAT UCP1 immunostaining. (C) UCP1, Prdm16, PPARα, PPARγ, and PGC-1α mRNA expression in BAT (n = 5). (D) UCP1 protein detected in mouse BAT. Right panel shows the results of densitometric analyses (n = 4). (E) UCP1, PPARα, PPARγ, and PGC-1α mRNA expression in rat primary adipocytes followed by 10-hour 2 μM curcumin treatment (n = 3). (F-G) UCP1-LUC reporter gene analyses in 293T cells treated with curcumin and (F) PPARα or (G) PPARγ agonist or antagonist, shown as relative LUC activity. (n = 5). (H) UCP1, PPARα, PPARγ, and PGC-1α mRNA expression in differentiated brown adipocyte cell line after 2 μM curcumin treatment for 10 hours (n = 6). *, **, or ***, P < 0.05 between indicated groups. [Color figure can be viewed at wileyonlinelibrary.com]

Finally, we tested the effect of curcumin on UCP1 expression in a mBAC after its differentiation for 3 days (Supporting Information Figure S8a-S8b). As shown in Figure 6H, 10-hour curcumin treatment increased UCP1, PPARα, PPARγ, and PGC-1α mRNA levels in differentiated mBAC. Supporting Information Figure S8c shows that curcumin-stimulated UCP1 mRNA expression in mBAC cells could not be completely blocked by PPARα or PPARγ inhibition.

Discussion

Increased WAT inflammation and reduced energy expenditure are among key causative factors of obesity and its related metabolic disorders, tightly associated with urbanization and sedentary lifestyles (32). Therapeutic agents that target these two events simultaneously might provide better treatment or prevention of metabolic diseases, but they are currently unavailable. Our current study suggests that this can be achieved via dietary intervention, such as the use of the curry component curcumin.

We show here that curcumin intervention inhibits HFD-induced WAT inflammation and activates BAT UCP1 production. As illustrated in Figure 7, the inhibition of WAT inflammation by 17-week curcumin intervention is achieved not only by reducing macrophage infiltration, but also by regulating macrophage polarity. Furthermore, we demonstrated the direct effect of adipocyte-secreted proinflammatory cytokines on M1-like macrophage differentiation and the inhibitory effect of curcumin on proinflammatory cytokine production by both immune cells and adipocytes. The stimulation of anti-inflammatory cytokine production by curcumin, however, may occur mainly in adipocytes. Importantly, we demonstrated the stimulation of BAT UCP1 production by dietary curcumin intervention, suggesting that metabolic beneficial effects of dietary polyphenols should not be fully attributed to their anti-inflammation and antioxidation effects. Finally, our observations indicated that curcumin stimulates UCP1 transcription via PPARα and PPARγ, and we suggest the existence of PPAR-independent mechanisms, as the stimulation of UCP1 promoter activity cannot be completely blocked by a PPARα or PPARγ antagonist.

Details are in the caption following the image

Diagram summarizing the major findings of this study. Curcumin intervention reduces WAT inflammation, achieved by reducing proinflammatory cytokine expression in both macrophages and mature adipocytes and increasing the anti-inflammatory cytokine expression in mature adipocytes. These effects collectively lead to the attenuation of macrophage infiltration and the reduction in the ratio of M1-like macrophages vs. M2-like macrophages. Curcumin intervention may also increase energy expenditure via both PPARα/PPARγ-dependent and -independent and independent mechanisms in stimulating UCP1 expression in BAT. [Color figure can be viewed at wileyonlinelibrary.com]

Curcumin, anthocyanin, and resveratrol are the three most studied dietary polyphenols for their preventive feature and therapeutic potential in T2D and other metabolic disorders (33). A clinical trial showed that curcumin intervention reduced the occurrence of T2D in prediabetic subjects (34). These dietary polyphenols share the common feature of targeting multiple organs without a defined receptor. Another common feature for dietary polyphenols is their low bioavailabilities because of the low intestinal absorption rate. These characteristics have generated certain difficulties in dissecting the mechanistic insights of their functions and may have created challenges for conventional drug hunters, as debated recently (35, 36).

It should be pointed out that applications of dietary polyphenol research are far beyond conventional drug development. Indeed, scientists have been approaching the mysteries inside of curcumin and other plant polyphenols, including the recognition of the stimulatory effect of curcumin (12), resveratrol (13), and anthocyanin (unpublished data) on hepatic hormone Fgf21 production. We also demonstrated that curcumin intervention restored HFD feeding-induced Fgf21 resistance (12). More importantly, dietary polyphenol studies are among the driving forces for scientists to pay close attention to gut microbiota. We generally accepted that at least some of the biological functions of dietary polyphenols are exerted via their interactions with the gut, including the direct stimulation of certain duodenum signaling cascades (without entering the bloodstream) (37), the production of gut microbiota metabolites of polyphenols (38), or the generation of gut microbiota products that regulate metabolic homeostasis via various means (9, 37-41). Referring to the current study, further investigations are required to determine whether the observed dual-modulatory effects of curcumin intervention on WAT and BAT are mediated by the yet-to-be-identified gut microbiota metabolites of curcumin.

In exploring mechanisms underlying the stimulation of Fgf21 expression by curcumin, we also noticed the stimulation on the nuclear receptor (NR) PPARα (12). Curcumin, however, is unlikely to be a direct ligand of PPARα. We conducted a GAL4-NR luciferase (GAL4-NR-LUC) reporter assay in HEK293 cells. In this assay, curcumin did not activate exogenously introduced PPARα. Furthermore, a conditioned medium of primary hepatocytes treated with curcumin also cannot stimulate the GAL4-NR-LUC reporter, suggesting that curcumin may increase the intracellular PPARα ligand level or the ligand activity in hepatocytes (12). In the current study, PPARα and PPARγ expression was stimulated by curcumin in mBAC cells (Figure 6H). Unexpectedly, the direct stimulatory effect of curcumin on UCP-1 could not be completely blocked by PPARα or PPARγ antagonist, although they are known transactivators of UCP1 (29-31). These results, along with observations in intensive previous investigations on curcumin and other dietary polyphenols, prompted us to suggest that a revolution on our receptor theory is needed. The current theory cannot always explain physiological observations, even for certain peptide hormones such as glucagon-like peptide-1 with defined receptors (42).

A recent study showed that adipose tissue Fgf21 production was stimulated by invariant natural killer T cells (43). This signaling cascade appeared to activate thermogenesis and subcutaneous adipocyte browning (43). We found that FGF21 treatment did not repress LPS/IFNγ-induced Raw264.7 cells toward M1-like macrophage differentiation, suggesting that should Fgf21 possess the anti-inflammation property, it is not mediated directly via inhibiting the M1-like macrophage differentiation. Considering that curcumin treatment stimulates Fgf21 production in the liver and mature adipocytes, why did we not see the “browning” effect in our long-term curcumin intervention animals? One explanation is that although short-term curcumin gavage in mice on a chow diet increased Fgf21 production, in HFD-fed mice, concomitant curcumin intervention attenuated HFD-stimulated hepatic Fgf21 production (12).

We did not see the browning effect in mouse inguinal adipose tissue via curcumin intervention, which was reported by another group with 50-day daily curcumin gavage in mice on an LFD (14). This is likely because of various experiment differences between these two studies, including the animal models, the dosages and the sources of curcumin, the way to admit curcumin, and the length of the experiments. As we have discussed above, the browning effect might be attributed mainly to the increases of circulating and adipose tissue Fgf21 levels, which could be elevated in mice on a chow diet but not in HFD-fed mice with dietary curcumin intervention.

The Raw264.7 cells were utilized in this study as the in vitro cell model to assess the effect of curcumin on M1-like and M2-like macrophage differentiation. It appears that the repression of the M1-like macrophage marker expression, but not the stimulation of the M2-like macrophage marker expression, can be achieved with curcumin at acceptable physiologically relevant doses (2.0-5.0 μM). Thus, curcumin intervention may directly inhibit M1-like macrophage differentiation. Its stimulatory effect on M2-like macrophage differentiation could be secondary, as are the results of reduced M1-like macrophage differentiation in combining with the regulation of proinflammatory and anti-inflammatory cytokine expression in WAT. This notion is supported by our results utilizing the Luminex multiplex assay for WAT immune cells (Table 1). In epididymal adipose tissue immune cells of mice that received 17-week curcumin intervention, we observed the stimulation of IL-10 but not IL-4 or IL-13, two other well-known anti-inflammatory cytokines. Indeed, a study revealed that IL-13 is mainly produced in mouse and human adipocytes (44). Curiously, its adipocyte expression can be enhanced in humans and rodents with obesity, induced by IL-1β and TNFα, and repressed by inhibitor of nuclear factor kappa-B kinase subunit beta (IKKβ) deficiency (44).

Although we demonstrated the stimulation of BAT UCP1 expression and improvement in the capacity of the mice in handling the cold challenge by curcumin intervention, we cannot eliminate the involvement of an increase in shivering thermogenesis. Further investigations are needed to clarify whether curcumin or other multiple target dietary polyphenols can regulate both shivering and nonshivering thermogenesis.

Conclusion

This study advanced our knowledge on the metabolic beneficial effects of the curry compound curcumin. Dietary intervention with this plant polyphenol is capable of playing the following dual-modulatory effect in preventing obesity and its related metabolic disorders: attenuation of WAT inflammation and promotion of BAT UCP1 production. Observations made in this study, along with the discoveries that curcumin and other plant polyphenols (anthocyanin and resveratrol) regulate Fgf21 production and function (12, 13), bring a novel perspective to dietary polyphenol intervention studies.

Acknowledgments

We thank Dr. Shingo Kejimura (University of California, San Francisco) for providing the mBAC cell line and Dr. Qing Yang for providing the UCP1-LUC reporter construct.

      The full text of this article hosted at iucr.org is unavailable due to technical difficulties.