Sodium butyrate alleviates adipocyte inflammation by inhibiting NLRP3 pathway

Xukai Wang, Gang He, Yan Peng, Weitian Zhong, Yan Wang, Bo Zhang, Xukai Wang, Gang He, Yan Peng, Weitian Zhong, Yan Wang, Bo Zhang

Abstract

Insulin resistance (IR) is a common feature of Type II diabetes, metabolic disorders, hypertension and other vascular diseases. Recent studies showed that obesity-induced inflammation may be critical for IR. To investigate the anti-inflammatory effect of sodium butyrate (NaB) on obesity-induced inflammation, the db/db mice were intraperitoneally injected with NaB for 6 weeks. Glucose control was evaluated by glucose tolerance test (GTT) and insulin tolerance test (ITT). Adipose tissue was harvested for gene expression analysis. 3T3-L1 adipocytes were treated with Tnf-α to mimic the inflammatory state and gene expression was detected by realtime PCR and Western blotting. Our results showed that NaB treatment improved glucose control in db/db mice as determined by GTT and ITT tests. Gene expression analysis showed that NaB inhibited cytokines and immunological markers including CD68, Interferon-γ and Mcp in adipose tissues in db/db mice. Moreover, NaB inhibited cytokine releasing in 3T3-L1 adipocytes treated with TNF-α. Further analysis of inflammation pathway showed that NLRP3 was activated in db/db mice, which was efficiently inhibited by NaB treatment. Our data suggest that inhibition of obesity-induced inflammation alleviates IR, and NaB might be a potential anti-inflammatory agent for obesity.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Figure 1. NaB improved glucose control in…
Figure 1. NaB improved glucose control in db/db mice.
(A) NaB treatment significantly decreased the body weight of db/db mice. Intraperitoneal glucose tolerance test (B), insulin tolerance test (C) and fasting glucose (D) were detected at the end of treatment as described in Materials and Methods. Data are the means ± SE. *P < 0.05, by Student’s t test (N vs NaB). N: PBS-treated controls; NaB: NaB-treated group.
Figure 2. NaB inhibited lymphocyte infiltration into…
Figure 2. NaB inhibited lymphocyte infiltration into adipose tissues.
(AB) Epididymal adipose tissues (EAT) and subcutaneous adipose tissue (SAT) were harvested from db/db mice treated with or without NaB for six weeks. Total RNA was isolated for realtime RT-PCR. Data are expressed in ΔCt values normalized against the mean Ct of GAPDH. Fold changes were calculated as 2^(ΔΔCtN-NaB). *P < 0.05, #P < 0.01 by Student’s t test. (C) Immunohistochemical staining of macrophage marker F4/80 in mouse EAT. N: Sham-treated group; NaB: NaB-treated group.
Figure 3. NaB inhibited cytokine expression in…
Figure 3. NaB inhibited cytokine expression in db/db mice adipose tissues.
Epididymal adipose tissues (A) and subcutaneous adipose tissue (B) were harvested from db/db mice treated with or without NaB for six weeks. Total RNA was isolated for realtime RT-PCR. Data are expressed in ΔCt values normalized against the mean Ct of GAPDH. Fold changes were calculated as 2^(ΔΔCtN-NaB). *P < 0.05, #P < 0.01 by Student’s t-test. N: PBS-treated group; NaB: NaB-treated group.
Figure 4. HDACi inhibited the cytokine expression…
Figure 4. HDACi inhibited the cytokine expression in 3T3-L1 adipocytes.
3T3-L1 cells were treated with TNF-α (50 ng/mL) combined with or without NaB(1 mM) or TSA(0.5 μM) for 8 hr. Total RNA was isolated for realtime RT-PCR to detect the gene expression of IL-1β and IL-6 (A). Supernatants of cell culture were harvested for ELISA assay to detect IL-1 and IL-6 in the culture medium (B). N: Normal control. *P < 0.05 by t-test (TNF vs NaB or TSA).
Figure 5. HDACi inhibited NLRP3 inflammasome pathway…
Figure 5. HDACi inhibited NLRP3 inflammasome pathway in 3T3-L1 cells.
3T3-L1 cells were treated with TNF-α combined without or with NaB (1 mM) and TSA (0.5 μM) for 8 hr. (A) Total RNA was isolated for realtime RT-PCR. mRNA expression was normalized to that of GAPDH. (B) Total proteins were isolated from treated 3T3-L1 cells for Western blots to detect the protein level of NLRP3 and IL-1. GAPDH was used as controls for equal loading. N: Normal control. (C) Densitometry measurement of Western blots. The experiments were repeated at least three times. *P < 0.05, #P < 0.01 by t-test (TNF vs NaB or TSA).
Figure 6. NaB inhibited LRP3 inflammasome pathway…
Figure 6. NaB inhibited LRP3 inflammasome pathway in db/db mice.
Epididymal adipose tissues (EAT) and subcutaneous adipose tissue (SAT) were harvested from db/db mice treated with or without NaB for six weeks. Total RNA was isolated for realtime RT-PCR (A). Data are expressed in ΔCt values normalized against the mean Ct of GAPDH. Fold changes were calculated as 2^(ΔΔCtN-NaB). Total proteins were isolated for Western-blot (B). The bar graph of the densitometry measurement showed the decreased expression of NLRP3 and active IL-1 in db/db adipose tissues treated without or with NaB (C). *P < 0.05, #P < 0.01 by Student’s t test.

References

    1. Lee B. C. & Lee J. Cellular and molecular players in adipose tissue inflammation in the development of obesity-induced insulin resistance. Biochim. Biophys. Acta 1842, 446–462 (2014).
    1. Winer D. A., Winer S., Chng M. H., Shen L. & Engleman E. G. B Lymphocytes in obesity-related adipose tissue inflammation and insulin resistance. Cell Mol. Life Sci. 71, 1033–1043 (2014).
    1. Watanabe Y., Nagai Y. & Takatsu K. Activation and regulation of the pattern recognition receptors in obesity-induced adipose tissue inflammation and insulin resistance. Nutrients 5, 3757–3778 (2013).
    1. Galic S., Sachithanandan N., Kay T. W. & Steinberg G. R. Suppressor of cytokine signalling (SOCS) proteins as guardians of inflammatory responses critical for regulating insulin sensitivity. Biochem J. 461, 177–188 (2014).
    1. Ramkhelawon B. et al. Netrin-1 promotes adipose tissue macrophage retention and insulin resistance in obesity. Nat Med. 20, 377–384 (2014).
    1. Gauger K. J. et al. Mice deficient in sfrp1 exhibit increased adiposity, dysregulated glucose metabolism, and enhanced macrophage infiltration. PLoS One 8, e78320 (2014).
    1. Nteeba J., Ortinau L. C., Perfield J. W. 2nd & Keating A. F. Diet-induced obesity alters immune cell infiltration and expression of inflammatory cytokine genes in mouse ovarian and peri-ovarian adipose depot tissues. Mol. Reprod. Dev. 80, 948–958 (2013).
    1. Matsubara T. et al. PGRN is a key adipokine mediating high fat diet-induced insulin resistance and obesity through IL-6 in adipose tissue. Cell Metab. 15, 38–50 (2012).
    1. Matsunaga T. et al. γ-Tocotrienol attenuates TNF-α-induced changes in secretion and gene expression of MCP-1, IL-6 and adiponectin in 3T3-L1 adipocytes. Mol. Med. Rep. 5, 905–909 (2012).
    1. Takimoto T. et al. 4β-Hydroxywithanolide E isolated from Physalis pruinosa calyx decreases inflammatory responses by inhibiting the NF-κB signaling in diabetic mouse adipose tissue. Int. J. Obes. (Lond) 38, 1432–1439 (2014).
    1. Gu D. et al. Inhibition of ERK1/2 pathway suppresses adiponectin secretion via accelerating protein degradation by Ubiquitin- proteasome system: relevance to obesity-related adiponectin decline. Metabolism 62, 1137–1148 (2013).
    1. Stanojcic M. et al. Leukocyte Infiltration and Activation of the NLRP3 Inflammasome in White Adipose Tissue Following Thermal Injury. Crit Care Med 42, 1357–1364 (2014).
    1. Dalmas E. et al. T cell-derived IL-22 amplifies IL-1β-driven inflammation in human adipose tissue: relevance to obesity and type 2 diabetes. Diabetes 63, 1966–1977 (2014).
    1. Wang X., Chrysovergis K., Kosak J. & Eling T. E. Lower NLRP3 inflammasome activity in NAG-1 transgenic mice is linked to a resistance to obesity and increased insulin sensitivity. Obesity (Silver Spring) 22, 1256–1263 (2014).
    1. Vandanmagsar B. et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat. Med. 17, 179–188 (2011).
    1. Stienstra R. et al. Inflammasome is a central player in the induction of obesity and insulin resistance. Proc. Natl. Acad. Sci. USA 108, 15324–15329 (2011).
    1. Goossens G. H. et al. Expression of NLRP3 inflammasome and T cell population markers in adipose tissue are associated with insulin resistance and impaired glucose metabolism in humans. Mol. Immunol. 50, 142–149 (2012).
    1. Frew A. J., Johnstone R. W. & Bolden J. E. Enhancing the apoptotic and therapeutic effects of HDAC inhibitors. Cancer Lett. 280, 125–133 (2009).
    1. Vieira E. L. et al. Oral administration of sodium butyrate attenuates inflammation and mucosal lesion in experimental acute ulcerative colitis. J. Nutr. Biochem. 23, 430–436 (2012).
    1. Huuskonen J., Suuronen T., Nuutinen T., Kyrylenko S. & Salminen A. Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids. Br. J. Pharmacol. 141, 874–880 (2004).
    1. Halili M. A. et al. Differential effects of selective HDAC inhibitors on macrophage inflammatory responses to the Toll-like receptor 4 agonist LPS. J. Leukoc. Biol. 87, 1103–1114 (2010).
    1. Revelo X. S., Luck H., Winer S. & Winer D. A. Morphological and inflammatory changes in visceral adipose tissue during obesity. Endocr. Pathol. 25, 93–101 (2014).
    1. Harte A. L. et al. NFκB as a potent regulator of inflammation in human adipose tissue, influenced by depot, adiposity, T2DM status, and TNFα. Obesity (Silver Spring) 21, 2322–2330 (2013).
    1. Kim J. K. et al. Prevention of fat-induced insulin resistance by salicylate. J. Clin. Invest. 108, 437–446 (2001).
    1. Robbins G. R., Wen H. & Ting J. P. Inflammasomes and Metabolic Disorders: Old Genes in Modern Diseases. Mol. Cell 54, 297–308 (2014).
    1. Esser N. et al. Obesity phenotype is related to NLRP3 inflammasome activity and immunological profile of visceral adipose tissue. Diabetologia 56, 2487–2497 (2013).
    1. Miao H. et al. Macrophage CGI-58 deficiency activates ROS-inflammasome pathway to promote insulin resistance in mice. Cell Rep. 7, 223–235 (2011).
    1. Falkenberg K. J. & Johnstone R. W. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat Rev Drug Discov. 13, 673–691 (2014).
    1. Zhou Y., Peng J. & Jiang S. Role of histone acetyltransferases and histone deacetylases in adipocyte differentiation and adipogenesis. Eur J Cell Biol. 93, 170–177 (2014).
    1. Chatterjee T. K. et al. Histone deacetylase 9 is a negative regulator of adipogenic differentiation. J Biol. Chem. 286, 27836–27847 (2011).
    1. Pryde S. E., Duncan S. H., Hold G. L., Stewart C. S. & Flint H. J. The microbiology of butyrate formation in the human colon. FEMS Microbiol. Lett. 217, 133–139 (2002).
    1. Guilloteau P. et al. From the gut to the peripheral tissues: the multiple effects of butyrate. Nutr. Res. Rev. 23, 366–384 (2010).
    1. Guilloteau P. et al. Dietary sodium butyrate supplementation increases digestibility and pancreatic secretion in young milk-fed calves. J. Dairy Sci. 93, 5842–5850 (2010).
    1. Moyer B. D., Loffing-Cueni D., Loffing J., Reynolds D. & Stanton B. A. Butyrate increases apical membrane CFTR but reduces chloride secretion in MDCK cells. Am. J. Physiol. 277, F271–F276 (1999).
    1. Tian J. et al. Oral treatment with γ-aminobutyric acid improves glucose tolerance and insulin sensitivity by inhibiting inflammation in high fat diet-fed mice. PLoS One 6, e25338 (2011).
    1. Vinolo M. A. et al. Tributyrin attenuates obesity-associated inflammation and insulin resistance in high-fat-fed mice. Am. J. Physiol. Endocrinol. Metab. 303, E272–E282 (2012).
    1. Gao Z. et al. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58, 1509–1517 (2009).
    1. Galisteo M., Duarte J. & Zarzuelo A. Effects of dietary fibers on disturbances clustered in the metabolic syndrome. J Nutr Biochem. 19, 71–84 (2009).
    1. Tang T. et al. Uncoupling of inflammation and insulin resistance by NF-kappaB in transgenic mice through elevated energy expenditure. J Biol Chem. 285, 4637–4644 (2010).
    1. Cantley M. D. & Haynes D. R. Epigenetic regulation of inflammation: progressing from broad acting histone deacetylase (HDAC) inhibitors to targeting specific HDACs. Inflammopharmacology 21, 301–307 (2013).
    1. Sweet M. J., Shakespear M. R., Kamal N. A. & Fairlie D. P. HDAC inhibitors: modulating leukocyte differentiation, survival, proliferation and inflammation. Immunol. Cell Biol. 90, 14–22 (2012).
    1. Miletta M. C. et al. Butyrate increases intracellular calcium levels and enhances growth hormone release from rat anterior pituitary cells via the G-protein-coupled receptors GPR41 and 43. PLoS One. 9, e107388 (2014).
    1. Masui R. et al. G protein-coupled receptor 43 moderates gut inflammation through cytokine regulation from mononuclear cells. Inflamm Bowel Dis. 19, 2848–2856 (2013).
    1. Li H. et al. Sodium butyrate stimulates expression of fibroblast growth factor 21 in liver by inhibition of histone deacetylase 3. Diabetes. 61, 797–806 (2012).

Source: PubMed

3
Se inscrever