Compositions, methods and uses for controlling heat generation in organisms

文档序号:1880592 发布日期:2021-11-26 浏览:2次 中文

阅读说明:本技术 用于控制生物体内热量产生的组合物、方法及用途 (Compositions, methods and uses for controlling heat generation in organisms ) 是由 尹芝南 王倩 杨恒文 于 2021-08-26 设计创作,主要内容包括:本发明公开用于控制生物体内热量产生的组合物、方法及用途,本发明的组合物包含用于调节EBI-3和p28结合或两者的二聚体与其受体的结合、或者下游信号通路的分子。本发明揭示了IL27信号在调节热量产生中的作用。分子机制研究证实其直接作用于脂肪细胞,激活p38MAPK-PGC1信号并刺激UCP1的产生。IL-27的治疗性给药逆转EBI-3KO小鼠低温诱导的低体温症。因此,本发明揭示IL-27信号在能量代谢过程中的关键作用,为控制体温提供了一种可行方案。(The present invention discloses compositions, methods and uses for controlling heat generation in an organism, the compositions of the invention comprising molecules for modulating the binding of the dimers of EBI-3 and p28 or both to their receptors, or downstream signaling pathways. The present invention discloses the role of the IL27 signal in regulating heat generation. Molecular mechanism research proves that the protein directly acts on fat cells, activates p38MAPK-PGC1 signals and stimulates the generation of UCP 1. Therapeutic administration of IL-27 reversed hypothermia induced by EBI-3KO mice. Therefore, the invention discloses the key role of IL-27 signals in the energy metabolism process and provides a feasible scheme for controlling body temperature.)

1. A composition for controlling heat generation in an organism comprising a molecule for modulating EBI-3 and p28 binding to their receptors or the downstream signaling pathway.

2. The composition of claim 1, wherein the modulation is inhibition or reduction of binding of EBI-3 and p28, or inhibition or reduction of binding of dimers of both to their receptors, or down regulation of downstream signaling pathways, and the control is inhibition of heat generation, thereby reducing the body temperature of the organism.

3. The composition of claim 2, wherein the molecule comprises a biological macromolecule or small molecule compound comprising an anti-EBI-3 antibody, an anti-p 28 antibody, an anti-IL-27R antibody, an EBI-3 soluble fragment, a p28 soluble fragment, an IL-27R soluble fragment.

4. The composition of claim 1, wherein the modulation is promoting or enhancing binding of EBI-3 and p28, or promoting or enhancing binding of dimers of both to their receptors, or up-regulating downstream signaling pathways, and the control is promoting heat production and thereby increasing body temperature of the organism.

5. The composition of claim 4, wherein the molecule comprises a recombinant IL-27 protein, a modification thereof, a polypeptide or a small molecule compound that activates IL-27R activity.

6. A method for controlling heat generation in an organism, comprising administering to the organism a molecule for modulating EBI-3 and p28 binding or binding of the dimer of both to its receptor or downstream signaling pathway.

7. The method of claim 6, wherein the organism comprises a human subject or tissue or cell thereof, a mammal or tissue or cell thereof, or a microorganism.

8. The method of claim 7, wherein the tissue or cell is an in vitro tissue or cell.

9. A method for screening a candidate compound for the ability to control heat generation in an organism comprising the step of measuring EBI-3 and p28 binding, or the binding of the dimer of either to its receptor, or the downstream signaling pathway triggered by such binding, before and after administration of a test compound.

10. The method of claim 9, wherein the test compound is identified as a candidate compound for inhibiting heat generation in an organism when binding of EBI-3 and p28, or binding of the dimer of both to its receptor, is inhibited or reduced, or a downstream signal due to said binding is down-regulated, following administration of the test compound.

11. The method of claim 9, wherein the test compound is identified as a candidate compound for promoting heat generation in an organism when binding of EBI-3 and p28, or binding of the dimer of both to its receptor, is promoted or enhanced, or a downstream signal due to said binding is upregulated after administration of the test compound.

12. The method of claim 9, comprising the steps of:

(1) measuring a parameter of EBI-3 and p28 binding, or a parameter of dimer of EBI-3 and p28 binding to a receptor thereof, or a parameter of downstream signal pathway activation in the biological model to obtain a first measurement value;

(2) measuring a parameter of EBI-3 and p28 binding, or a parameter of dimer of the two binding to a receptor thereof, or a parameter of downstream signal pathway activation in the biological model after administering the test compound to the biological model, to obtain a second measurement value; and

(3) a step of comparing the first measurement value and the second measurement value.

13. The method of claim 9, comprising the steps of:

(1) measuring a parameter of EBI-3 and p28 binding, or a parameter of dimer of both binding to its receptor, or a parameter of downstream signaling pathway activation in the first biological model to obtain a first measurement;

(2) a step of measuring a parameter of EBI-3 and p28 binding, or a parameter of dimer of both binding to its receptor, or a parameter of downstream signaling pathway activation in a second organism model, wherein the first organism model and the second organism model belong to the same model and the first organism model is not administered with a test compound, and the second organism model is administered with a test compound, to obtain a second measurement value; and

(3) a step of comparing the first measurement value and the second measurement value.

14. The method of claim 9, comprising the steps of:

(1) measuring the parameters of EBI-3 and p28 binding, or the parameters of dimer of the two binding to the receptor thereof, or the parameters of downstream signal pathway activation in the biological model after the administration of the test compound to obtain measured values;

(2) a step of comparing said measurement value with a reference value, wherein said reference value is a parameter of EBI-3 and p28 binding, or a parameter of dimer of both binding to its receptor, or a parameter of downstream signaling pathway activation, measured from a biological model in the absence of administration of the test compound.

15. The method of any one of claims 12-14, wherein the biological model comprises an animal model or a cellular model.

Technical Field

The present invention relates to the field of biotechnology, in particular to compositions, methods and uses for controlling heat generation in an organism.

Background

Brown and beige lipogenesis play a key role in maintaining the development of body temperature. Although the targeting and activation of brown/beige fat is known, the diversity and abundance of immune factors in adipose tissue is not clear.

US20170128410a1 discloses a method and composition for achieving mammalian energy balance, in particular a method of achieving mammalian energy balance using forskolin during adipogenesis inhibition, wherein forskolin is added separately to pre-differentiated preadipocytes and also to mature adipocytes to comparatively assess adipogenesis inhibition potential, said process comprising the step of adding forskolin at a concentration of an amount in a pre-seeded microplate consisting of undifferentiated preadipocytes.

In the function and mechanism research of panda in Lkb1 regulation of brown adipose tissue, an Adipoq-Cre/Loxp system is adopted to establish an adipose tissue specific Lkb1KO mouse model (Ad-Lkb1), the influence of Lkb1 regulation of brown fat heat production and body energy metabolism is researched by combining histology, molecular level and in vitro and in vivo experiments, and a molecular mechanism of Lkb1 regulation of body energy metabolism and Ucp1 expression is disclosed through chromatin co-immunoprecipitation, protein co-immunoprecipitation and dual-luciferase report analysis.

The information in this background is only for the purpose of illustrating the general background of the invention and should not be taken as an acknowledgement or any form of suggestion that this information forms the prior art that is known to a person skilled in the art.

Disclosure of Invention

To solve at least some of the technical problems of the prior art, the present invention provides compositions, methods and uses for controlling heat generation in an organism. The research of the invention finds that (EBI-3/p28) signals have important effects on improving in vivo heat generation, and proves that IL-27 directly acts on adipocytes, activates p38MAPK-PGC1 signals and stimulates the generation of UCP 1. Specifically, the present invention includes the following.

In a first aspect of the invention, there is provided a composition for controlling heat generation in an organism comprising a molecule for modulating the binding of a dimer of EBI-3 and p28 or both to its receptor, or downstream signaling pathway.

Preferably, the modulation is inhibition or reduction of binding of EBI-3 and p28, or inhibition or reduction of binding of dimers of both to their receptors, or down regulation of downstream signaling pathways, and the control is inhibition of heat generation, thereby reducing the temperature of the organism.

Preferably, the molecules according to the composition of the present invention comprise biological macromolecules or small compounds, such as anti-EBI-3 antibodies, anti-p 28 antibodies, anti-IL-27R antibodies, EBI-3 soluble fragments, p28 soluble fragments, IL-27R soluble fragments, and the like.

Preferably, the composition according to the present invention, the modulation means promoting or enhancing the binding of EBI-3 and p28, or promoting or enhancing the binding of dimers of both to their receptors, or up-regulating downstream signaling pathways, and the control means promoting heat generation and thereby increasing the temperature of the organism.

Preferably, the molecule comprises recombinant IL-27 protein and its modified substance, polypeptide or small molecule compound with activity of activating IL-27R.

In a second aspect of the invention, there is provided a method for controlling heat generation in an organism by administering to the organism a molecule for modulating the binding of a dimer of EBI-3 and p28 or both to its receptor, or downstream signaling pathway.

According to the method for controlling heat generation in an organism of the present invention, preferably, the organism includes a human subject or a tissue or a cell thereof, a mammal or a tissue or a cell thereof, or a microorganism.

According to the method for controlling heat generation in an organism of the present invention, preferably, the tissue or cell is an in vitro tissue or cell.

In a third aspect of the present invention, there is provided a method for screening a candidate compound capable of controlling heat generation in an organism, comprising the step of measuring the binding of EBI-3 and p28, or the binding of the dimer of both to its receptor, or the downstream signaling pathway triggered by said binding, before and after administration of said test compound.

According to the method for screening a candidate compound capable of controlling the heat generation in an organism of the present invention, preferably, when the binding of EBI-3 and p28, or the binding of the dimer of both to its receptor, is inhibited or decreased or a downstream signal due to the binding is down-regulated after the administration of the test compound, the test compound is identified as a candidate compound inhibiting the heat generation in an organism.

According to the method for screening a candidate compound capable of controlling the heat generation in an organism of the present invention, preferably, when the binding of EBI-3 and p28, or the binding of the dimer of both to its receptor, is promoted or enhanced or a downstream signal due to the binding is up-regulated after the administration of the test compound, the test compound is identified as a candidate compound promoting the heat generation in an organism.

The method for screening a candidate compound capable of controlling heat generation in an organism according to the present invention preferably comprises the steps of:

(1) measuring a parameter of EBI-3 and p28 binding, or a parameter of dimer of EBI-3 and p28 binding to a receptor thereof, or a parameter of downstream signal pathway activation in the biological model to obtain a first measurement value;

(2) measuring a parameter of EBI-3 and p28 binding, or a parameter of dimer of the two binding to a receptor thereof, or a parameter of downstream signal pathway activation in the biological model after administering the test compound to the biological model, to obtain a second measurement value; and

(3) a step of comparing the first measurement value and the second measurement value.

The method for screening a candidate compound capable of controlling heat generation in an organism according to the present invention preferably comprises the steps of:

(1) measuring a parameter of EBI-3 and p28 binding, or a parameter of dimer of both binding to its receptor, or a parameter of downstream signaling pathway activation in the first biological model to obtain a first measurement;

(2) a step of measuring a parameter of EBI-3 and p28 binding, or a parameter of dimer of both binding to its receptor, or a parameter of downstream signaling pathway activation in a second organism model, wherein the first organism model and the second organism model belong to the same model and the first organism model is not administered with a test compound, and the second organism model is administered with a test compound, to obtain a second measurement value; and

(3) a step of comparing the first measurement value and the second measurement value.

The method for screening a candidate compound capable of controlling heat generation in an organism according to the present invention preferably comprises the steps of:

(1) measuring the parameters of EBI-3 and p28 binding, or the parameters of dimer of the two binding to the receptor thereof, or the parameters of downstream signal pathway activation in the biological model after the administration of the test compound to obtain measured values;

(2) a step of comparing said measurement value with a reference value, wherein said reference value is a parameter of EBI-3 and p28 binding, or a parameter of dimer of both binding to its receptor, or a parameter of downstream signaling pathway activation, measured from a biological model in the absence of administration of the test compound.

Preferably, the biological model comprises an animal model or a cell model, and the method for screening candidate compounds capable of controlling heat generation in an organism according to the present invention.

In a fourth aspect of the invention, there is provided the use of a composition of the first aspect in the manufacture of a formulation for metabolic disorders.

The present invention discloses the role of IL-27(EBI-3/p28) signaling in improving heat production in vivo. IL-27 was shown to act directly on adipocytes, activating p38MAPK-PGC1 signaling and stimulating UCP1 production. Therapeutic administration of IL-27 can reverse EBI-3KO mouse hypothermia induced by hypothermia.

Drawings

FIG. 1 is a graph of IL-27 signaling to promote heat generation and energy consumption.

FIG. 2 is a graph of IL-27 directly targeting adipocytes to promote heat generation.

FIG. 3 is a graph of IL-27 promoting heat generation.

FIG. 4 shows a reduction in energy expenditure and caloric production in IL-27 signaling deficient mice.

FIG. 5 shows that the thermogenic effect of IL-27R α signaling is not through direct effect on CD2+ lymphocytes or Lyz2+ myeloid cells.

FIG. 6 is a phenotype of IL-27R α KO and WT chimeras with adaptive heat production induced by HFD.

Figure 7 shows that IL-27 up-regulates UCP1 and improves browning of subcutaneous white adipose tissue.

FIG. 8 shows that IL-27 promotes caloric production.

Detailed Description

Reference will now be made in detail to various exemplary embodiments of the invention, the detailed description should not be construed as limiting the invention but as a more detailed description of certain aspects, features and embodiments of the invention.

It is to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. Further, for numerical ranges in this disclosure, it is understood that the upper and lower limits of the range, and each intervening value therebetween, is specifically disclosed. Every smaller range between any stated value or intervening value in a stated range and any other stated or intervening value in a stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included or excluded in the range.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although only preferred methods and materials are described herein, any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention. All documents mentioned in this specification are incorporated by reference herein for the purpose of disclosing and describing the methods and/or materials associated with the documents. In case of conflict with any incorporated document, the present specification will control. Unless otherwise indicated, "%" is percent by weight.

As used herein, the term "heat generation" may be used interchangeably with the terms "heat generation", "heat generation" and "heat generation". Thermogenesis in this context refers to the process by which mammals, including humans, regulate heat production.

In the present invention, EBI-3 and p28 are combined, which means that the EBI-3 subunit and the p28 subunit form a heterodimeric structure with a certain spatial arrangement through interaction, and the interaction can be connected in a disulfide bond mode.

The term "receptor" as used herein refers to a biological macromolecule capable of binding to the aforementioned dimers and of causing a change in cell function. It contains at least an active site that recognizes and binds to the dimer and a functional active site responsible for generating a response reaction, and thus initiates a series of biochemical reactions, ultimately leading to the generation of a biological effect by the target cell. The result of binding between the receptor and the dimer of the present invention is that the receptor is activated and the basic steps for the receptor to activate subsequent signaling occur. Under physiological conditions, the association between the receptor and the dimer is not mediated by covalent bonds, but rather by ionic bonds, hydrogen bonds, van der waals forces, and hydrophobic interactions.

The term "signaling pathway" as used herein refers to a phenomenon in which when a certain reaction occurs in a cell, a signal transmits a message from the outside of the cell to the inside of the cell, and the cell responds according to the message. The signal path of the invention particularly refers to a series of enzymatic reaction paths which can transmit extracellular molecular signals into cells through cell membranes to exert effects.

[ composition for controlling Heat Generation in organisms ]

The present invention provides a composition for controlling heat generation in an organism comprising a molecule for modulating the binding of a dimer of EBI-3 and p28 binding or both to its receptor, or downstream signaling pathway. Preferably, the compositions of the invention comprise molecules for modulating IL27 signaling or IL27 binding to its receptor or downstream signaling pathway. Downstream signaling pathways include, but are not limited to, JAK/STAT3, p38MAPK, and the like. In the present invention, thermogenesis and energy expenditure are mediated by adipocytes, including but not limited to brown/beige adipocytes and white adipocytes. In the present invention, the above-mentioned adipocytes act as IL27 target cells, not immune cells.

[ method for screening candidate Compound capable of controlling Heat production in an organism ]

In the present invention, the screening method comprises at least the step of measuring the binding between EBI-3 and p28, or the binding between the dimer of both and its receptor, or the downstream signaling pathway triggered by the binding, using an agent before and after the administration of the test compound. The above combination further comprises the step of measuring a parameter of interest by use of the reagent, said parameter comprising the amount of dimer formed by the reagent, or the metabolic phenotype, or the amount of the thermogenic protein. The thermogenic protein includes, but is not limited to UCP1, PPAR alpha, PGC-1 alpha. In the present invention, downstream signaling pathway activation (sometimes also referred to as activation) refers to further expression of a downstream target gene. Preferably, the reagent comprises the sequence shown in SEQ ID NO. 1-30.

Examples

1. Experimental Material

1.1 materials

1.1.1 mice

Interleukin-27R alpha KO (B6N.129P2-Il27ratm1Mak/J, numbering: 018078) and EBI-3KO (B6.129X1-Ebi 3)tm1Rsb/J, numbering: 008691) mice were purchased from Jackson Laboratory. Adipoq-cre mice were provided by professor Yong Liu. IL-27R alphaf/fMouse [ paper of the text ]]Prepared by the laboratory. Animal experiments were conducted in accordance with ethical norms and protocols approved by the institutional animal care and utilization committee of river-south university and the institutional animal care and utilization committee of yale university. Groups of gender and age matched mice were used for all experiments, with 4 to 6 animals per cage. IL-27 treatment experiments and bone marrow chimera production were performed in random groups. The mice were housed in a sterile temperature and humidity controlled facility for animals at 25 deg.C and under dark conditionsThe dark period is 12:12h, and food and water can be freely obtained.

1.1.2 reagents

Use of Bio-Plex-ProTMHuman Cytokine 17-Plex assay kit (Cat. No. M5000031YV) and Bio-Plex ProTMHuman serum samples were analyzed on a Bio-Plex system (BioRad) using Human infection Panel1(Cat. No.171-AL 001M). Human IL-27ELISA kit (434607) was purchased from Biolegend. Glucose (63005518) was purchased from chinese medicinal chemicals. Blood glucose test strips (one-touch) were purchased from Johnson and Jonhson. Anti-mouse pAKT mab (4060), anti-mouse AKT mab (4685), anti-mouse GAPDH mab (5174), anti-mouse HSP90 mab (4877), anti-mouse PPAR γ mab (2435), anti-mouse pSTAT3 mab (9145), anti-mouse STAT3 mab (9139), anti-mouse p-p38 MAPK (9211), anti-mouse p38MAPK (9212), anti-mouse pATF2 mab (9221), anti-mouse ATF2 mab (35031) were purchased from Cell Signaling Technology. Anti-mouse UCP1 monoclonal antibody (ab10983), anti-mouse PGC-1 alpha monoclonal antibody (ab54481), anti-mouse PPAR alpha monoclonal antibody (ab8934), anti-mouse PRDM16 monoclonal antibody (ab202344), anti-mouse IL-27R alpha monoclonal antibody (ab5997) purchased from Abcam. APC-CY 7-anti-mouse CD45 (clone 30-F11) was purchased from BD. APC anti-mouse CD11b (clone M1/70), PE anti-mouse F4/80 (clone BM8) were purchased from Sungene Biotech (Tianjin, China). Recombinant mouse IL-27(rmIL-27) (577408) was purchased from Biolegend. STAT3 inhibitor SH-4-54 and p38MAPK inhibitor SB203580 were purchased from Selleck Chemicals. Brown/beige fat differentiation agents dexamethasone (D4902), rosiglitazone (R2408), indomethacin (I7378), isoproterenol (I5627), forskolin (F6886), 3-isobutyl-1-methylxanthine (IBMX) (I5879), triglyceride detection kit (T2449), insulin (I3536) were purchased from Sigma. Leptin detection kit (MOB00), adiponectin detection kit (MRP300) were purchased from R and D. Insulin detection kit (90080) was purchased from Crystal Chem.

2. Experimental methods

2.1 enzyme-linked immunosorbent assay

Human serum was obtained as described above. Mouse sera were collected 10 weeks after HFD feeding. Human IL-27 and mouse insulin were detected using an ELISA kit according to the instructions.

2.2 Luminex immunoassay

Human serum was collected as described above. Use of Bio-Plex-ProTMHuman Cytokine 17-Plex assay kit (Cat. No. M5000031YV) and Bio-Plex ProTMHuman serum inflammatory factors were detected by Human Inflammation Panel1(Cat. No.171-AL001M) on a Bio-Plex200 flow liquid chip assay System (BioRad) according to the instructions of the user's manual.

2.3 Western blot

Whole cell or tissue lysates were extracted using RIPA lysis buffer (Beyotime, China) supplemented with the complete protease inhibitor (Roche) and phosphatase inhibitor Cocktail2(P5726, Sigma) and supernatants were used for subsequent analysis. The proteins were diluted in Loading dye (BL502A, Bio-sharp), heated at 95 ℃ for 10 min, and run on 4-12% polyacrylamide gels. Proteins were transferred to polyvinylidene fluoride membranes and western blot experiments were performed with commercial antibodies and methods-reagents mentioned in the figure.

2.4 histology

Adipose or liver tissue was fixed in 4% paraformaldehyde/1 × PBS overnight at 4 ℃ and embedded in paraffin prior to sectioning. Sections were stained with hematoxylin and eosin (H and E) or oil red o (lever) and photographed under a bright field microscope.

2.5 immunohistochemistry

UCP1 and immunohistochemistry were performed using rabbit specific HRP/dab (abc) detection IHC kit (ab64261, Abcam) according to the manufacturer's instructions.

2.6 core body temperature measurement and Cold stimulation experiments

Cold stimulation experiments were performed in a temperature controlled cold room. Mice were placed individually in individual cages (without bedding) at 4 ℃. The mice had free access to food and water. Rectal core body temperatures were recorded every 2 hours using a digital thermometer and rectal thermocouple probe (TH-212, HICHANCE, China). If the core body temperature falls below 20 ℃, individual mice are euthanized and scored as events for survival analysis.

2.7 metabolism cages

Integrated laboratory animal monitoring in rodent incubators using environmental control of yale universityA measurement system (CLAMS, Columbus Instruments, Columbus, OH) metabolic cage measures energy loss and energy consumption of mice. Mice were housed individually and acclimated in the metabolic chamber for 48 hours prior to data collection. The rats had free access to food and water. Each mouse was subjected to continuous monitoring of physical activity and food intake. During the experiment, each mouse collected CO 4 times per hour2/O2And (4) concentration.

2.8RNA extraction and Gene expression analysis

Total RNA was extracted from frozen tissue using TRNzol universal reagent (Tiangen, beijing) and quantified using a Nanodrop 2000 uv-vis spectrophotometer (Thermo). The cDNA was obtained using PrimeS scriptTMRT Reagent kit (TAKARA, large even) using 1 u g total RNA RT polymerase chain reaction detection preparation. Real-time quantitative PCR of cDNA using TB GreenTM Premix-EX-TaqTMII kit (TAKARA) in CFX96TMReal-time PCR detection was performed on a Bio-Rad detection system. Internal reference to mouse HPRT by Δ CtThe method uses immunohistochemical method to calculate fold change in expression. The primer pair sequences are as follows. Mouse HPRT: forward-TCATTATGCCGAGGATTTG (SEQ ID NO.1), reverse-GCCTCCCATCTCCTTCAT (SEQ ID NO. 2); TNF-alpha, ford ard-CTACTGAACTTCGGGGTGAT (SEQ ID NO.3), reverse-CAGGCTTGTCACTCGAATT (SEQ ID NO. 4); IL-6: forward-TCCAGTTGCCTTCTTGGGAC (SEQ ID NO.5), reverse-GTGTAATTAAGCGCCGACTTG (SEQ ID NO. 6); IL-12p40 forward-GGCTGGTGCAAAGAAACATGGACTTGA (SEQ ID NO.7), reverse-TGCAGACAGAGACGCCATTCCACAT (SEQ ID NO. 8); IL-1 beta is forward-CAACCAACAAGTGATATTCTCCATG (SEQ ID NO.9), reverse-GATCCACACTCTCCAGCTGCA (SEQ ID NO. 10); IL-4: forward-GAAAACTCCATGCTTGAAGAA (SEQ ID NO.11), reverse-TCTTTCAGTGATGTGGACTTG (SEQ ID NO.12), IL-5: forward-TCACCGAGCTCTGTTGACAA (SEQ ID NO.13), reverse-CCACACTTCTCTTTTTGGCG (SEQ ID NO. 14); IL-13 forward-TGAGCAACATCACACAAGACC (SEQ ID NO.15), reverse-GGCCTTGCGGTTACAGAGG (SEQ ID NO. 16); ucp1 forward-ACTGCCACACCTCCAGTCATT (SEQ ID NO.17), reverse-CTTTGCCTCACTCAGGATTGG (SEQ ID NO. 18); cox8b forward-GAACCATGAAGCCAACGACT (SEQ ID NO.19), reverse-GCGAAGTTCACAGTGGTTCC (SEQ ID NO. 20); cidea: forward-TGCTCTTCTGTATCGCCCAGT (SEQ ID NO.21), reverse-GCCGTGTTAAGGAATCTGCTG (SEQ ID NO.21)NO.22);Prdm16:forward-CAGCACGGTGAAGCCATTC(SEQIDNO.23),reverse-GCGTGCATCCGCTTGTG(SEQIDNO.24);Adiponectin:forward-GAATCATTATGACGGCAGCA(SEQIDNO.25),reverse-TCATGTACACCGTGATGTGGTA(SEQIDNO.26);Elovl3:forward-TCCGCGTTCTCATGTAGGTCT(SEQIDNO.27),reverse-GGACCTGATGCAACCCTATGA(SEQIDNO.28)。

2.9 bone marrow chimera

IL-27R α KO and WT mice at 10 weeks of age were used as bone marrow cell donors. Bone marrow was isolated from hind limb femurs, lysed by erythrocytes, filtered through a cell filter (40 μm), and the cell pellets were washed 3 times with sterile PBS, counted and stored on ice for injection. 10 weeks old IL-27R alpha KO and WT mice were lethally irradiated (900rads) and implanted 1X10 by ophthalmic intravenous injection7Bone marrow cells, mice were placed in a special pathogen-free facility, supplemented with sterile water and feed for 8 weeks to reconstitute the immune system, and then subjected to metabolic studies.

2.10 Primary beige adipocyte preparation

The inguinal subcutaneous adipose tissue was minced, digested in PBS containing collagenase II (1mg/ml) at 37 ℃ for 45 minutes, and the tissue suspension was filtered through a 100 μm cell filter, centrifuged at 600g for 5min, to granulate the Stromal Vascular Fraction (SVF). After granulation, further filtration through a 40 μm cell filter was performed and placed on collagen-coated plates. After overnight incubation, the supernatant containing non-adherent cells was removed. Precursor adipocytes were grown in fusion in DMEM containing 10% FBS + insulin (5. mu.g/ml). Treatment with dexamethasone (1 μ M), 3-isobutyl-1-methylxanthine (IBMX, 0.5mM), insulin (5 μ g/ml), indomethacin (125nM) and rosiglitazone (1 μ M) for 2 days induced differentiation of the fused cells, followed by separate treatment with insulin (5 μ g/ml) and triiodothyronine (T3, 1nM) for an additional 5 days. On day 7, cells were pretreated overnight with and without IL-27(100ng/ml) and then treated with isoproterenol (10 μ M) or forskolin (10 μ M) for 4-6 hours. And adding IL-27(100ng/ml) for treatment for 0-120min on day 7 for protein phosphorylation analysis, or for 12-24h for protein expression level detection. For signal inhibition experiments, STAT3 inhibitor (C188-9, 10 μ M) or p38MAPK inhibitor (SB203580, 10 μ M) were added to the medium 0.5h prior to and during IL-27 treatment.

2.11 flow cytometry analysis (FACS).

Epididymal adipose tissue was minced and digested as described above. SVF particles were used for surface staining of APC-CY7 anti-mouse CD45, APC anti-mouse CD11b, PE anti-mouse F4/80, incubated for 15min, and cells were washed with PBS and analyzed with BD-FACSVverse flow cytometer (BD).

2.12 production of IL-27R alpha conditional knockout mice. The development work of IL-27R alpha conditional knockout mice is carried out by utilizing a gene targeting technology. Briefly, a 997bps CKO region (GenBank NM-016671.3) containing exon 3 and exon 4 of the IL27ra gene, a 5.2kb 5 'homology arm and a 3kb 3' homology arm were amplified from BAC clones using high fidelity Taq technology and assembled into targeting vectors with recombination sites and selectable markers. The final targeting vector was determined by multiple restriction endonucleases and full sequence analysis and was transformed into 129s mouse embryonic stem cells by electroporation. The correct embryonic stem cell clone was identified and injected into C57BL/6J mouse blastocysts. After the chimeric mouse was unambiguously identified, it was backcrossed to C57BL/6J mice. Genotyping PCR demonstrated germline transmission. Mice with the IL-27R α f/w genotype were then bred with B6 adipoq-cre mice. Mice of the desired genotype were selected for the experiment. Genotyping primers for detection of the flox or wt band: forward CTGGTTCTGGTATGGTTTGGGGTT (SEQ ID NO.29) and reverse TGAAAGAACTCAACAGTGGGCCGG (SEQ ID NO. 30).

2.13 IL-27 treatment

WT mice at 8 weeks of age were HFD fed for 32 weeks and randomized into 2 groups. Mice were injected intraperitoneally with either rmI L-27(100 μ g/kg) or PBS for 15 consecutive days, and then subjected to metabolic analysis. 12-month-old EBI-3KO mice were randomly divided into 2 groups, and were injected intraperitoneally with rmIL-27 (100. mu.g/kg) or PBS for 7 consecutive days. The mice were then placed in a cold challenge experiment at 4 ℃.

2.14 statistical analysis

Mapping and statistical analysis were performed using Graphpad Prism software (version 7). All statistical tests are fully described in the chart legend and meet the criteria of a normal distribution with similar variances. The sample size was not predetermined using statistical methods. The two groups were compared using the t-test. For the evaluation data of the relevant samples, repeated measures analysis of variance (AVOVA) was performed. For more than two sets of evaluations, one-way anova and multiple comparisons were used. For the evaluation between two independent variables, a two-way analysis of variance with multiple comparisons was used. Survival analysis was performed using a log rank test. The correlation analysis was performed using linear regression analysis. Data are expressed as mean ± s.e.m., unless otherwise indicated. p <0.05 was considered to have statistical significance, denoted as p <0.05, p <0.01, p <0.001, NS, not significant.

3. Results of the experiment

FIG. 1 is a graph of IL-27 signaling to promote heat generation and energy consumption. il-27R α KO mice and WT control mice were fed HFD for 4 weeks and then housed in metabolic cages. Food intake (shown as a in the figure), oxygen consumption (shown as b in the figure) and energy consumption (shown as c in the figure) were monitored over 24 hours (n-7-8). d and e: IL-27R α KO mice and WT control mice were separately HFD fed for 6 weeks and then cold stimulated (4 ℃). The survival curves are shown in the figure (d, n-6-8) and the rectal temperatures are shown in the figure (e, n-11-15). F and g: IL-27R α KO and WT mice were fed with HFD for 6 weeks, followed by cold stimulation at 25 deg.C (shown as f in the figure) or 4 deg.C for 2 hours (shown as g in the figure). Subcutaneous fat (SCW) and Brown Adipose Tissue (BAT) were collected for UCP1 immunoblot analysis. h-j: IL-27R α KO and WT mice fed on a normal diet were placed in cold stimulation at 25 ℃ or 4 ℃ for 48 hours. SCW and BAT were collected for UCP1 immunoblot analysis (shown in panel H), i for UCP1 immunohistochemical staining (scale bar 100 μm) and j for H and E staining of BAT and SCW tissues (scale bar 50 μm). (f-h): the band density was quantified and normalized with ImageJ. Each lane represents a biologically independent sample, and the experiment was repeated at least twice with similar results. Data are mean ± s.e.m. of biologically independent samples, two-way analysis of variance (a-c and e); log rank test (d). P <0.05, p <0.01, p < 0.001.

FIG. 2 is a graph of IL-27 directly targeting adipocytes to promote heat generation. a. Four sets of bone marrow chimeras were generated by feeding HFD for 10 weeks with IL-27R α KO or WT mice as donors or recipients. Body weight was recorded weekly (n-10-13). b. Inlay for normal feed feeding by cold stimulationInfluence of rectal temperature in mice (4 ℃, n ═ 6-7). c. Chimeric mice fed normal chow were immunoblotted with SCW collected 12 hours after cold stimulation at 4 ℃. d.8 weeks old Adipoq-CreIl27raf/fMouse and Il27raf/fThe control group was fed HFD for 10 weeks. Body weight was recorded weekly (n-7-9). e. Effect of cold stimulation on rectal temperature in normal diet mice (4 ℃, n ═ 16-17). f. Adipoq-Cre Il27ra fed with normal dietf/fAnd Il27raf/fAfter cold stimulation of mice at 4 ℃ for 12 hours, SCW and BAT were collected for immunoblotting. g.8 Weekly-aged Ucp1-CreIl27raf/fMouse and Il27raf/fThe control group was fed HFD for 10 weeks. Body weight was recorded weekly (n-5-6). h. Effect of cold stimulation on rectal temperature in normal diet mice (4 ℃, n ═ 5-8). i. Ucp1-CreIl27ra for feeding normal foodf/fAnd Il27raf/fAfter cold stimulation of mice at 4 ℃ for 12 hours, SCW and BAT were collected for immunoblotting (c, f and i), and the band densities were quantified and normalized with ImageJ. Each lane represents a separate biological sample, and the experiment was repeated at least twice with similar results. Data are mean ± s.e.m. of biologically independent samples. Two-way analysis of variance (a, b, d, e, g and h). P<0.05,**p<0.01,**p<0.001。

FIG. 3 is a graph showing the effect of IL-27 in promoting caloric production. a. Primary cream-colored adipocytes were generated in vitro from SVF from WTSCW and then treated with rmIL-27(100ng/ml) or PBS for 24 hours. Cells were lysed and protein extracts were used for immunoblot analysis. b. Immunoblot analysis of protein phosphorylation in WT primary beige adipocyte extracts treated with rmIL-27(100ng/ml) for the indicated time. c. Primary beige adipocytes derived from WTSCW were generated in vitro. STAT3 inhibitor (C188-9, 10. mu.M) or p38MAPK inhibitor (SB203580, 10. mu.M) were added to the medium at 0.5 hours prior to culture and for the duration of rmIL-27 treatment (100ng/ml for 12 hours). Representative immunoblots of cell lysis and protein extracts are shown. d-j: WT mice were fed with HFD for 32 weeks and then injected intraperitoneally. Every other day, rmIL-27 (100. mu.g/kg) or PBS was injected for 15 days. d. Body weight was recorded at the indicated time points (n-8). 15 days after rmil-27 treatment, adipose tissues were collected and weighed (n-6-8) for glucose tolerance test (f, n-8) and insulin tolerance test (g, n-8). h. Mice were fasted overnight and then injected intraperitoneally with 0.75U/kg insulin (15 minutes). Immunoblot analysis of pAKT (pSer473) in epididymal fat was performed. i.rmIL-27 treatment was followed by oil red O liver tissue staining 15 days later. Scale bar 50 μm. j-m: UCP1-KO mice were fed HFD for 20 weeks, and then were injected intraperitoneally with rmIL-27 (100. mu.g/kg) or PBS every other day for 15 consecutive days. j. Body weight was recorded at the indicated time points (n-6). k. Adipose tissues and liver were collected and weighed 15 days after rmIL-27 treatment (n-6), and GTT (l, n-6) and ITT (m, n-4) tests were performed 15 days after rmIL-27 treatment. The band densities were quantified with ImageJ and normalized (a and c), and the experiment was repeated at least twice with similar results. Data are mean ± s.e.m. of biologically independent samples. Unpaired student t-test (e and k); two-way analysis of variance (d, f, g, j, l and m). P < 0.05.

FIG. 4 shows the reduction in energy expenditure and caloric production in IL-27 signaling deficient mice. a-c: IL-27R α KO and WT mice, 8 weeks old, were placed in metabolic cages. Wherein, the food intake (a), the oxygen consumption (b) and the energy consumption (c) (n is 7-8) are shown in the figure. d. BAT and SCW tissues were isolated from 6-8 week old WT-ND and IL-27R α KO, cut into small pieces (about 0.003g BAT and about 0.004g SCW), and the basal oxygen consumption rate was measured using a Seahorse XF analyzer (n: 5/group, 4-5 pieces/group/mouse). Il-27 ralpha KO and WT mice were fed a ND response survival curve to cold challenge (4 ℃, n ═ 9-11). f. Effect of cold stimulation on rectal temperature in ND-fed mice (4 ℃, n ═ 9-11). g. Expression of IL-27R α KO and WT mouse genes from normal diet in SCW was detected using real-time PCR (n-9-11). EBI-3KO mice and WT control group fed with normal diet were recorded at 4 ℃ cold stimulation and their survival curves (h) and rectal temperatures (i) (n-5-6) are shown. BAT and SCW were collected 12 hours after cold stimulation, lysed and used for immunoblot analysis of UCP1 (j). The band density was quantified by ImageJ and the UCP1/HSP90 ratio was normalized. Experiments were performed twice showing similar results. Data are mean ± s.e.m. of biologically independent samples. Unpaired t-tests (d and g); two-way analysis of variance (a-c, f and i); log rank test (e and h). P <0.05, p <0.01, p < 0.001.

FIG. 5 shows IL-2The thermogenesis of the 7ra signal is not through direct effects on CD2+ lymphocytes or Lyz2+ bone marrow cells. Wherein, a.Il27raflox/floxSchematic model generated by mice. The Il27ra site (top) was targeted by a targeting vector (second) containing the homologous sequences of Il27ra, including two LoxP sites flanking exons 3 and 4 and one Neo selection cassette. The linearized vector was then delivered to embryonic stem cells by electroporation (C57BL/6), followed by drug selection, PCR screening, and Southern blot confirmation. Homologous recombination generates a floxed allele (Third). After correct targeting of ES clones as determined by Southern blotting, selected clones were used for blastocyst microinjection to generate F0 passages. It was identified that F0 was crossed with Flp-deleter, confirming that F1 was the germline passage to delete the Neo cassette (Forth). After Cre recombination, a mismatch of the floxedIl27ra allele will lead to the deletion of exons 3 and 4 (bottom). Il27raf/fGenotyping of mice. c. Detection of Il27ra by real-time fluorescent quantitative PCR methodf/fAnd expression of Il27ra gene in WT mouse spleen (n ═ 3). d. Detection of Il27ra by real-time fluorescent quantitative PCR (n-3, left)f/fAnd Cd2-Cre Il27raf/fExpression of Il27ra gene in mouse spleen CD 3T cells. Intraperitoneal injection of ethanethiolate into Il27raf/fAnd Lyz2-Cre Il27raf/fAbdominal macrophages were collected after 4 days in mice and tested for Il27ra gene expression by real-time PCR (n-5-6, right panel). e-h 8 week old Il27raf/f、Cd2-Cre-Il27raf/fAnd Lyz2-Cre-Il27raf/fMice were fed HFD for 10 weeks. Wherein, body weight was recorded weekly (n-11-21). Gtt (f) and itt (g) trials (n-11-19) were performed 10 weeks after HFD treatment. h. Adipose tissue was collected 10 weeks after HFD treatment and weighed (n-11-21). Data are mean ± s.e.m. of biologically independent samples. Unpaired t-test (c, d); two-way analysis of variance (e-g); one-way analysis of variance (h) p<0.05,***p<0.001。

FIG. 6 is a phenotype of IL-27R α KO and WT chimeras with adaptive heat production induced by HFD. Bone marrow cells from CD45.1 WT strain were transferred to either CD45.2 IL-27R α KO or CD45.2 WT host (1X 10)7Cells/mouse) to generate chimeras. Mice were housed for 8 weeks to reconstitute the immune system. As shown in the figureAs shown, immune cells were isolated from different tissues and analyzed by flow cytometry for the percentage (n-3) of donor (CD45.1+) and host (CD45.2+) cells, CD19+ B (middle) or F4/80+ macrophages (bottom) in CD4+ T (top). b-d: bone marrow chimeras were obtained and fed with HFD as shown in panel a. After 10 weeks of HFD treatment GTT (b, n-10-13) and ITT (c, n-5-7) tests were performed. d. Adipose tissue and liver were collected 10 weeks after HFD treatment and weighed (n-4-6). E-g. chimeric mice fed normal diet were cold stimulated at 4 ℃ for 12 hours, and SCW and BAT were collected for histological analysis (E, H and E, scale bar 100 μm) or UCP1 immunohistochemical staining (f, scale bar 100 μm). Real-time PCR analysis of gene expression of SCW was performed (g, n ═ 12). Data are mean ± s.e.m. of biologically independent samples. Unpaired t-tests (a, f and g); one-way anova (d); two-way analysis of variance (b and c). P<0.05,**p<0.01,**p<0.001。

Figure 7 shows that IL-27 up-regulates UCP1 and improves browning of subcutaneous white adipose tissue. a-c: 8 week old Il27ra fed with HFDf/fAnd Ucp1-CreIl27raf/fMice were used for 10 weeks. Gtt (a) and itt (b) (n-5-6) trials were performed 10 weeks after HFD treatment, adipose tissue and liver were collected and weighed (c, n-5-6). d. Il27ra fed with normal foodf/fAnd Ucp1-Cre Il27raf/fAfter cold stimulation of mice at 4 ℃ for 12 hours, SCW and BAT were collected for histological analysis (H and E). Scale bar 100 μm. e. Primary cream-colored adipocytes were generated in vitro from SVF from WTSCW and then treated with rmIL-27(100ng/ml) or PBS for 24 hours. The cells were lysed and the protein extracts were used for immunoblot analysis. BAT tissue from WT mice served as positive control. f. Immunoblot analysis of STAT1 phosphorylation from WT primary beige adipocyte extracts treated with rmIL-27(100ng/ml) for the indicated time. g. WT mice were cultured in vitro. Two different p38MAPK inhibitors (SB203580, 10. mu.M and SB202190, 5. mu.M) or three STAT3 inhibitors (C188-9, 10. mu.M; Static, 10. mu.M or HO3867, 20. mu.M) were added to the medium 0.5h before and during the treatment of rmIL-27(100ng/ml, 12 h). The expression of UCP1 was detected by immunoblotting. h-i: primary beige adipocytes from SCW of WT (h) or IL-27R α KO (i) mice were obtained by in vitro culture. p38MAPK inhibitionAgents (SB203580, 10. mu.M) or STAT3 inhibitors (C188-9, 10. mu.M) were added to the medium 0.5h prior to and during treatment (100ng/ml, 12h) with rmIL-27. Expression of UCP1 or PPAR α was analyzed by immunoblot analysis. j-m: EBI-3KO mice were injected intraperitoneally with either rmIL-27 (100. mu.g/kg) or PBS for 7 consecutive days, followed by cold stimulation at 4 ℃. j. Rectal temperature of mice to cold stimulation (n ═ 6). SCW and BAT protein extracts were used for immunoblot analysis 24 hours after cold stimulation. UCP1 staining (l) and histological analysis (m) (scale bar 100 μm) are shown. Representative portions are shown. The band density was quantified and normalized with ImageJ. Two repetitions of the experiment showed similar results. Data are mean ± s.e.m. of biologically independent samples. Unpaired t-test (c); two-factor differential analysis (a, b and j) × p<0.05,**p<0.01,**p<0.001。

FIG. 8 shows the effect of IL-27 in promoting caloric production. a-i: il27raf/fAnd Adipoq-CreIl27raf/fMice were fed HFD for 32 weeks and were injected intraperitoneally with either rmIL-27 (100. mu.g/kg) or PBS every other day for 15 days. Detecting inflammatory factors in serum by adopting a Bio-RadBio-Plex200 multifunctional analyzer. b. Representative tissue sections (H and E) of the indicated tissue. c. The percentage of infiltrated CD4 or CD8T cells and the cytokine production by hepatic CD 4T cells were analyzed by flow cytometry. Detecting and recording Il27raf/fBody weight of mice (d), gtt (e), and itt (f) (n-5-8). Detecting and recording Adipoq-CreIl27raf/fBody weight (g), gtt (h), and itt (i) (n-4-5) of mice. j-m: feeding Ucp1-CreIl27ra with HFDf/fMice were injected intraperitoneally with rmIL-27 (100. mu.g/kg) or PBS every other day for 16 weeks for 15 days. Body weight (j), tissue weight (k), glucose tolerance test (l), and insulin tolerance test (m) (n-5-6) were measured and recorded. Data are mean ± s.e.m. of biologically independent samples. One-way anova (a and c), two-way anova (d-j, l and m); unpaired t-test (k). P<0.05,**p<0.01,**p<0.001。

4. Conclusion

Browning of white adipose tissue predisposes mice to increased energy expenditure. To examine the systemic metabolic status, the present invention examined the energy and nutritional consumption of IL-27 ra KO mice and WT control groups under normal diet or HFD treatment. Energy expenditure in normally-fed IL-27 ra KO mice was significantly reduced without changing food intake (shown as a-c in figure 7). The phenotype was more pronounced when fed HFD (shown as a-c in figure 1). These data reveal that IL-27 signaling has the effect of maintaining systemic metabolic homeostasis. To determine whether the effect of IL-27 Ra deficiency on energy expenditure may be attributed to reduced thermogenesis, the present invention next examined the effect of IL-27 signaling deficiency on adaptive heat generation.

IL-27R α KO mice were sensitive to cold-induced hypothermia under normal dietary conditions (shown in FIG. 4 as f and g), and this intolerance was more severe after HFD treatment (shown in FIG. 1 as d and e). The expression of the key protein uncoupling protein 1(UCP1) that regulates thermogenesis in IL-27 ralpha KO mice was also significantly reduced (shown as f-h in figure 1). Tissue sections also showed less browning in the IL-27R α KO mouse Brown Adipose Tissue (BAT) and subcutaneous white adipose tissue (SCW) (shown in i in fig. 1) and fewer cells containing multilumen lipid droplets (shown in j in fig. 1). These data clearly indicate that thermogenic activity of IL-27R α KO mice is severely impaired. Notably, UCP1 was already reduced in IL-27R α KO mice prior to cold stimulation (shown in fig. 1 as f and h), identical to the other thermogenic genes (shown in fig. 4 as f), which is consistent with a reduction in systemic energy expenditure (shown in fig. 1 as b and c, and in fig. 4 as b and c). In addition, cold stimulation of EBI-3KO mice recapitulates findings in IL-27R α KO mice (shown in FIG. 4 as g-i), further confirming the pyrogenic effect of IL-27 signaling.

Next, the cell types responsible for IL-27 mediated signaling were dissected. First, 4 groups of chimeric mice (WT) were generated using the bone marrow of IL-27R α KO or WT mice>WT、WT>KO、KO>WT and KO>KO) and HFD treatment was performed on these mice. Surprisingly, only those chimeras made with IL-27R α KO, whatever the donor cells, were highly susceptible to HFD-induced fat accumulation and cold stimulation (shown in FIG. 2 as a-c). The IL-27R α KO chimera was consistently inhibited from heat generation compared to the wild-type chimera, as evidenced by reduced body temperature, reduced multilumen lipid droplets and reduced UCP1 production (shown as a-c in FIG. 2). These studies indicate that the dominant target of IL-27 is adipocytes and not immune cells. In fact, IL-27R alpha is expressed on mature adipocytes and primary beige adipocytes. Furthermore, IL-27R α△adipoThe mouse's caloric production was also inhibited by intolerance to cold stimuli, reduced browning, and reduced UCP1 production (shown in fig. 2 as c, e, f, h). Taken together, the above data indicate that IL-27 promotes caloric production by directly targeting adipocytes.

To investigate the molecular mechanism of IL-27 action on adipocytes, we treated primary beige adipocytes directly with IL-27 in vitro. The expression of UCP1 was significantly increased after IL-27 treatment, while up-regulating the expression of peroxisome proliferator-activated receptor alpha (PPAR α), peroxisome proliferator-activated receptor gamma co-activator 1 α (PGC-1 α) and key transcriptional co-activators mediating energy metabolism (shown as a in fig. 3). Typical downstream signaling pathways for IL-27R α in immune cells are JAK/STAT3 and p38 MAPK. Next we examined whether IL-27 promotes adipocyte heat production via these pathways. phosphorylation of p38MAPK was significantly increased under IL-27 stimulation, whereas pSTAT3 was only transiently induced and rapidly resolved (shown in fig. 3 b). P38MAPK is known to activate ATF 2-mediated transcription of PGC-1 α, and PGC-1 α is a major regulator of UCP1 and thermogenesis. As expected, IL-27 treatment also enhanced the activation of ATF2 in primary beige adipocytes, and therewith upregulated PGC-1 α, which could be eliminated by pharmacological inhibition of the activity of p38MAPK (shown as a-c in fig. 3). In addition, cells with p38MAPK inhibition promoted tolerance to IL-27 induced UCP1 (shown as c in FIG. 3). However, IL-27 up-regulated UCP1 response to STAT3 inhibition was essentially unaffected, although it also decreased PGC-1 α expression (shown as c in FIG. 3). These results indicate that IL-27 enhances caloric production primarily through the p38 MAPK-PGC-1. alpha. signaling pathway. Interestingly, inhibition of both p38 and STAT3 failed to abrogate PPAR α upregulation, suggesting that IL-27 action may involve additional pathways.

Furthermore, supplementation of EBI-3KO mice with IL-27 improved UCP 1-mediated caloric production and avoided low temperatures in cold stimulation (fig. 4).

In summary, our results indicate that the IL-27 signal is required for full adaptive heat generation. Direct targeting of IL-27 to adipocytes induced activation of p38MAPK, driving activation of ATF2 and subsequent expression of PGC-1 α and UCP 1. Taken together, these findings provide new insights that extend our understanding of the mechanisms of heat generation in the body.

While the present invention has been described with reference to exemplary embodiments, it is to be understood that the invention is not limited to the disclosed exemplary embodiments. Many modifications and variations may be made to the exemplary embodiments of the present description without departing from the scope or spirit of the present invention. The scope of the claims is to be accorded the broadest interpretation so as to encompass all modifications and equivalent structures and functions.

22页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:ASS1或BCKDK抑制剂在制备治疗溃疡性结肠炎的药物中的应用

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!