Review
Toll-like receptors: linking inflammation to metabolism

https://doi.org/10.1016/j.tem.2010.08.007Get rights and content

Obesity has been characterized as a state of chronic inflammation. Inflammatory signaling not only causes peripheral insulin resistance, but also promotes neuronal insulin and leptin resistance, which further propagates a positive energy balance. Upon development of obesity, numerous conditions, including increased circulating cytokine concentrations and cell autonomous dysregulation of homeostatic signaling pathways, such as the endoplasmic reticulum stress response, promote activation of stress kinases, to cause peripheral insulin as well as central insulin and leptin resistance. Recently, activation of toll-like receptor (TLR) signaling has been recognized as an alternative activator of obesity-induced inflammation. In this paper, we review recent progress in defining the molecular basis of obesity-associated TLR activation and its role in the development of metabolic syndrome.

Section snippets

Obesity causes chronic low-grade inflammation

The worldwide prevalence of obesity has progressively increased over the past 30 years. The World Health Organization estimates that by 2015, approximately 700 million adults will be obese [1]. Cardiovascular disease, type 2 diabetes mellitus (T2DM), fatty liver disease and obesity related cancers are the main health problems accounting for the morbidity associated with the increasing prevalence of obesity [2].

The link between obesity, insulin resistance and T2DM has been recognized for

Etiology of obesity

Obesity is characterized by increased storage of FAs in an expanded adipose tissue mass, and by chronic activation of inflammatory pathways 7, 8. Survival of multicellular organisms ultimately depends on the ability to fight infection, and the capability of storing energy for times of low nutrient availability. Thus, nutrient and pathogen-sensing systems are highly conserved across species, from organisms such as Caenorhabditis elegans and Drosophila to mammals 4, 8.

Adipose tissue, apart from

Central regulation of energy homeostasis

Both energy intake and energy expenditure are, at least partly, under the tight control of defined neuronal circuits in the CNS that are targeted by leptin. These neural circuits also receive multiple inputs from peripheral organs to control food intake, energy expenditure and locomotor activity, and they also integrate a coordinated response that adapts metabolic processes such as regulation of hepatic gluconeogenesis, glucose disposal, insulin secretion and lipid partitioning, depending on

Obesity causes peripheral insulin resistance

The past decade has revealed important new insights into the role of innate immune response pathways in the development of obesity-associated peripheral insulin resistance. Obesity is characterized by a broad inflammatory response, and transcriptional profiling studies have revealed that inflammatory and stress response genes are among the most tightly regulated genes in the adipose tissue of obese animals [8].

The first indication for increased cytokine release in obesity was provided by the

Molecular mechanisms of insulin resistance

Inflammatory signaling cascades activated in obesity have been shown to be causally linked to the development of insulin resistance in peripheral tissues such as skeletal muscle and liver [8]. However, the interaction of inflammatory signaling and systemic insulin resistance is complex, as lack of IL-6 and IL-18, either in the whole body or specific tissues, also results in obesity and/or insulin resistance 41, 42, 43.

The molecular basis for the observed impairment in insulin action as a result

Role of FAs in insulin resistance

The consumption of fat-rich diets also contributes to the development of insulin resistance, T2DM and dyslipidemia [59]. As inflammatory signaling pathways contribute to these pathologies, intensive efforts have been made to identify potential mechanisms that might link the intake of dietary fat with inflammatory signaling. One proposed link has emerged from the finding that key molecular components of the innate immune system, TLRs, play a pivotal role in the development of insulin resistance

TLRs and innate immunity

TLRs are pattern-recognition receptors (PRRs) and play a crucial role in the innate immune system, which detects the presence and the nature of pathogenic microbial infection, and thus provides the first line of host defense (Box 1). TLRs, of which 12 members have been identified to date in mammals, comprise a family of type I transmembrane receptors, which are characterized by an extracellular leucine rich repeat (LRR) domain and an intracellular Toll/IL1 receptor (TIR) domain, which shows

TLR signaling induces peripheral insulin resistance

TLR4 is expressed in a variety of cell types, predominantly in the cells of the immune system, including macrophages and dendritic cells. TLR4 binds to the lipopolysaccharides (LPS) of Gram-negative bacterial cell walls [69]. Interestingly, the medium-chain FA component of LPS, lauric acid, is sufficient to trigger TLR4 signaling in a macrophage cell line 74, 75, 76. SFAs, circulating levels of which are often increased in obesity, thus represent important candidates for causing diet-induced

TLR signaling mediates central insulin and leptin resistance

In peripheral tissues, sustained exposure to a supply of nutrients exceeding energy requirements induces insulin resistance via multiple, convergent mechanisms [92]. Diverse tissues are affected by nutrient excess, so it is not surprising that the CNS, especially the hypothalamus, is also susceptible [93]. However, this carries the potential to cause obesity, rather than simply being its consequence, because insulin and leptin are crucial hormonal signals that inform the brain of the body's

Concluding remarks

The recognition that obesity is associated with chronic low-grade inflammation and that activation of inflammatory signaling contributes to the development of obesity-associated insulin resistance and T2DM has revolutionized our understanding of the pathophysiology of this common and socioeconomically important endocrine disorder. Although the identification of intracellular stress kinases such as JNK and IKK, which are activated upon obesity development, both in peripheral tissues and in the

Acknowledgements

We apologize to all colleagues whose important contributions could not be cited because of space limitations. We thank G. Schmall and T. Rayle for excellent secretarial assistance. This work was supported by the Center for Molecular Medicine (CMMC), University of Cologne (TVA1 to J.C.B.), the European Community's Seventh Framework Program (FP7/2007–2013) under grant agreement number 201608 (TOBI), the European Union (FP7-HEALTH-2009–241592, EurOCHIP, to J.C.B.), the DFG (BR 1492/7–1 to J.C.B.),

References (103)

  • S. Seino et al.

    Physiological and pathophysiological roles of ATP-sensitive K+ channels

    Prog. Biophys. Mol. Biol.

    (2003)
  • F.T. Wunderlich

    Interleukin-6 signaling in liver-parenchymal cells suppresses hepatic inflammation and improves systemic insulin action

    Cell Metab.

    (2010)
  • V. Aguirre

    The c-Jun NH(2)-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser(307)

    J. Biol. Chem.

    (2000)
  • Z. Gao

    Serine phosphorylation of insulin receptor substrate 1 by inhibitor kappa B kinase complex

    J. Biol. Chem.

    (2002)
  • A.V. Chibalin

    Downregulation of diacylglycerol kinase delta contributes to hyperglycemia-induced insulin resistance

    Cell

    (2008)
  • Y. Lin

    The hyperglycemia-induced inflammatory response in adipocytes: the role of reactive oxygen species

    J. Biol. Chem.

    (2005)
  • J.J. Senn

    Toll-like receptor-2 is essential for the development of palmitate-induced insulin resistance in myotubes

    J. Biol. Chem.

    (2006)
  • M.T. Nguyen

    A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways

    J. Biol. Chem.

    (2007)
  • H. Kumar

    Toll-like receptors and innate immunity

    Biochem. Biophys. Res. Commun.

    (2009)
  • T. Kawai

    Unresponsiveness of MyD88-deficient mice to endotoxin

    Immunity

    (1999)
  • C. Pasare et al.

    Toll-like receptors: linking innate and adaptive immunity

    Microbes Infect.

    (2004)
  • J.Y. Lee

    Saturated fatty acids, but not unsaturated fatty acids, induce the expression of cyclooxygenase-2 mediated through Toll-like receptor 4

    J. Biol. Chem.

    (2001)
  • J.Y. Lee

    Reciprocal modulation of Toll-like receptor-4 signaling pathways involving MyD88 and phosphatidylinositol 3-kinase/AKT by saturated and polyunsaturated fatty acids

    J. Biol. Chem.

    (2003)
  • J.B. Lockridge

    Bioinformatic profiling of the transcriptional response of adult rat cardiomyocytes to distinct fatty acids

    J. Lipid. Res.

    (2008)
  • O. Takeuchi et al.

    Pattern recognition receptors and inflammation

    Cell

    (2010)
  • M. Saberi

    Hematopoietic cell-specific deletion of toll-like receptor 4 ameliorates hepatic and adipose tissue insulin resistance in high-fat-fed mice

    Cell Metab.

    (2009)
  • W.L. Holland

    Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance

    Cell Metab.

    (2007)
  • P.D. Cani et al.

    Interplay between obesity and associated metabolic disorders: new insights into the gut microbiota

    Curr. Opin. Pharmacol.

    (2009)
  • S.C. Woods

    Consumption of a high-fat diet alters the homeostatic regulation of energy balance

    Physiol. Behav.

    (2004)
  • P.J. Enriori

    Diet-induced obesity causes severe but reversible leptin resistance in arcuate melanocortin neurons

    Cell Metab.

    (2007)
  • A. Kleinridders

    MyD88 signaling in the CNS is required for development of fatty acid-induced leptin resistance and diet-induced obesity

    Cell Metab.

    (2009)
  • X. Zhang

    Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity

    Cell

    (2008)
  • WHO. (2006) Obesity and overweight. Fact sheet number 311....
  • A.H. Mokdad

    Prevalence of obesity, diabetes, and obesity-related health risk factors, 2001

    JAMA

    (2003)
  • A.V. Ferreira

    High-carbohydrate diet selectively induces tumor necrosis factor-alpha production in mice liver

    Inflammation

    (2010)
  • G.S. Hotamisligil

    Inflammation and metabolic disorders

    Nature

    (2006)
  • L. Plum

    Central insulin action in energy and glucose homeostasis

    J. Clin. Invest.

    (2006)
  • K.E. Wellen et al.

    Inflammation, stress, and diabetes

    J. Clin. Invest.

    (2005)
  • Y. Zhang

    Positional cloning of the mouse obese gene and its human homologue

    Nature

    (1994)
  • J.M. Friedman

    Obesity: Causes and control of excess body fat

    Nature

    (2009)
  • Q. Gao

    Disruption of neural signal transducer and activator of transcription 3 causes obesity, diabetes, infertility, and thermal dysregulation

    Proc. Natl. Acad. Sci. U. S. A.

    (2004)
  • J.W. Hill

    Acute effects of leptin require PI3K signaling in hypothalamic proopiomelanocortin neurons in mice

    J. Clin. Invest.

    (2008)
  • S.H. Bates

    STAT3 signalling is required for leptin regulation of energy balance but not reproduction

    Nature

    (2003)
  • K.D. Niswender

    Intracellular signalling. Key enzyme in leptin-induced anorexia

    Nature

    (2001)
  • K.K. Bence

    Neuronal PTP1B regulates body weight, adiposity and leptin action

    Nat. Med.

    (2006)
  • M.B. Ernst

    Enhanced Stat3 activation in POMC neurons provokes negative feedback inhibition of leptin and insulin signaling in obesity

    J. Neurosci.

    (2009)
  • E. Rother

    Neurocircuits integrating hormone and nutrient signaling in control of glucose metabolism

    Am. J. Physiol. Endocrinol. Metab.

    (2008)
  • E. Gropp

    Agouti-related peptide-expressing neurons are mandatory for feeding

    Nat. Neurosci.

    (2005)
  • S. Luquet

    NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates

    Science

    (2005)
  • Q. Gao et al.

    Neurobiology of feeding and energy expenditure

    Annu. Rev. Neurosci.

    (2007)
  • Cited by (278)

    • Editorial: Obesity, metabolic phenotypes and COVID-19

      2022, Metabolism: Clinical and Experimental
    View all citing articles on Scopus
    View full text