Skip to main content

MINI REVIEW article

Front. Endocrinol., 06 December 2016
Sec. Cellular Endocrinology
This article is part of the Research Topic Obesity, smoking and fatty liver disease View all 11 articles

Distinct Hepatic Macrophage Populations in Lean and Obese Mice

\r\nRafael Mayoral Monibas,*Rafael Mayoral Monibas1,2* \r\nAndrew M. F. JohnsonAndrew M. F. Johnson3 \r\nOlivia OsbornOlivia Osborn3 \r\nPaqui G. TravesPaqui G. Traves4Sushil K. Mahata,*\r\nSushil K. Mahata5,6*
  • 1Merck Research Laboratories, Kenilworth, NJ, USA
  • 2CIBERehd – Networked Biomedical Research Center, Hepatic and Digestive Diseases, Madrid, Spain
  • 3Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
  • 4Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA, USA
  • 5Metabolic Physiology & Ultrastructural Biology Laboratory, Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
  • 6Metabolic Physiology & Ultrastructural Biology Laboratory, Department of Medicine, University of California San Diego, La Jolla, CA, USA

Obesity is a complex metabolic disorder associated with the development of non-communicable diseases such as cirrhosis, non-alcoholic fatty liver disease, and type 2 diabetes. In humans and rodents, obesity promotes hepatic steatosis and inflammation, which leads to increased production of pro-inflammatory cytokines and acute-phase proteins. Liver macrophages (resident as well as recruited) play a significant role in hepatic inflammation and insulin resistance (IR). Interestingly, depletion of hepatic macrophages protects against the development of high-fat-induced steatosis, inflammation, and IR. Kupffer cells (KCs), liver-resident macrophages, are the first-line defense against invading pathogens, clear toxic or immunogenic molecules, and help to maintain the liver in a tolerogenic immune environment. During high fat diet feeding and steatosis, there is an increased number of recruited hepatic macrophages (RHMs) in the liver and activation of KCs to a more inflammatory or M1 state. In this review, we will focus on the role of liver macrophages (KCs and RHMs) during obesity.

Introduction

The rising prevalence of obesity represents a major global health challenge, not least because it is considered a significant risk factor for a wide array of non-communicable diseases. Prominent among these are diseases of the liver, ranging from steatosis through to cirrhosis, collectively termed non-alcoholic fatty liver disease (NAFLD) (1). However, the etiology linking obesity with liver pathology is incompletely understood, hindering attempts to treat these conditions.

A landmark discovery offering therapeutic potential for the metabolic syndrome was the finding that the adipose tissue of obese mice and humans displays hallmarks of an inflammatory state, including increased concentrations of tumor necrosis factor alpha (TNF-α) and increased monocyte/macrophage infiltration (24). Indeed, TNF-α is sufficient to induce features of the metabolic syndrome, such as insulin resistance (IR), and many chemical and genetic depletion studies have demonstrated the importance of inflammation and inflammatory macrophages in this process [recently reviewed in Ref. (5)]. Macrophage accumulation also occurs in other key metabolic tissues including muscle (69), liver (1012), and pancreas (13, 14), which contribute to the dysregulation of glucose homeostasis. In this review, we focus on the composition and behavior of hepatic macrophage populations in obese mice and highlight recent advances that could aid in the targeting of this axis to treat aspects of the metabolic syndrome.

The Liver at the Interface between Metabolism and Immunity

The liver is a key metabolic organ, which regulates a variety of processes vital for maintaining metabolic homeostasis. These include control of glucose production and lipid metabolism, dysregulation of which are symptomatic of the metabolic syndrome. The liver also plays key roles as part of the immune system secreting acute-phase proteins, complement components, cytokines, chemokines, and being positioned, along with the gastrointestinal tract, at the major interface between ourselves and our external, even microbial environment (15, 16). This unique position where metabolism and immunity are intertwined is reflected in the liver architecture, whereby immune cells are intimately connected to hepatocytes and liver sinusoidal endothelial cells (LSECs) (17, 18), as well as the cross-regulation whereby metabolic stress can result in hepatic immune activation leading to metabolic dysregulation (19, 20).

The liver maximizes nutrient absorption as blood flows through a system of sinusoidal vessels and fenestrations through beds of hepatocytes (17). The majority of blood within the sinusoid derives from the intestines via the hepatic portal vein and is rich in both nutrients, and also potentially immunogenic microbial molecules, or in cases of opportunistic infection microbes themselves (17). Therefore, in addition to facilitating nutrient absorption, sinusoids must also enable the removal of immunogenic material and allow the immune system to combat of infection. Kupffer cells (KCs) are located in the hepatic sinusoids and play a key role in this process (18). They bind a range of microbes or microbial ligands via microbe-associated molecular patterns (MAMPs), and by phagocytosis prevent them penetrating into the general circulation (18). Lipopolysaccharide (LPS), for example, is readily detectable in portal blood, but only rarely detectable in systemic circulation (21). Compared with macrophages from other locations, KCs are predisposed to respond to activation signals in a less inflammatory fashion and are especially characterized by producing high concentrations of the anti-inflammatory cytokine, interleukin 10 (IL-10) (22). Furthermore, KCs, along with other antigen-presenting cells in the liver, express low levels of co-stimulatory molecules required to initiate an adaptive immune response and high levels of molecules that suppress T cell activation, such as programed death-ligand 1 (PDL-1) (17). Thus, during homeostasis KCs in collaboration with other hepatic immune cell populations clear microbial material while maintaining the inflammatory tone of the liver at a level sufficient for essential functions such as pathogen killing, tissue remodeling, and sinusoidal permeability, but below that which would result in overt inflammation and tissue damage (5, 18, 23). The factors maintaining KCs in this tolerogenic state are not completely clear but are critically important when we consider how these cells and the hepatic macrophage pool in general are altered during obesity.

The phenotype of tissue macrophages is thought to be dependent on their respective ontogeny, as well as their respective polarization state in the tissue environment (24). Polarization was most clearly described by in vitro studies, which used cytokines to induce different extremes of macrophage phenotype classified as M1 or classically activated macrophages, considered more pro-inflammatory, and M2 or alternatively activated macrophages that have an anti-inflammatory tone (25). M1 macrophage differentiation can be induced by interferon gamma (IFN-γ), alone or with microbial products such as LPS or inflammatory cytokine TNF-α. In contrast, interleukin 4 (IL-4), interleukin 10 (IL-10), interleukin 13 (IL-13), interleukin 33 (IL-33), transforming growth factor beta (TGF-β), and granulocyte colony-stimulating factor (G-CSF) activate macrophages to differentiate to M2. However, given the range of factors now known to influence macrophage polarization, including cellular metabolic state (26), it is likely that a spectrum of macrophage phenotypes occur in vivo even within the same tissue macrophage pool (25). In lean mice, KCs have an M2-like phenotype maintained by the type 2 cytokine, IL-4, and the nuclear hormone receptor peroxisome proliferator activator receptor delta (PPAR-δ) (27, 28). Thus, KCs are specialized by virtue of their derivation from the yolk sac early in development (24, 29), and by factors in the liver environment, which maintain them in a less inflammatory, M2-like state (27, 28).

Parenchymal and Non-Parenchymal Cells in Liver

Hepatocytes are the major parenchymal cells, while the non-parenchymal cells integrate five cell populations including resident macrophages or KCs (30), recruited hepatic macrophages (RHMs), resident innate lymphocytes or natural killer cells (NKs) (31, 32), fat storing cells termed Ito or stellate cells (HSCs) (33), and LSECs (34) (Figure 1).

FIGURE 1
www.frontiersin.org

Figure 1. Schematic diagram showing parenchymal and non-parenchymal cells in liver. (A) Lean liver showing parenchymal hepatocytes (HC) and non-parenchymal anti-inflammatory Kupffer cells (M2-KC), natural killer cells (NK), hepatic stellate cells (HSC), and liver sinusoidal endothelial cells (LSEC). (B) High fat diet-induced obese liver showing activated pro-inflammatory Kupffer cells (M1-KC), recruited hepatic macrophages (RHM), and lipid droplets (L). CV, central vein; E, erythrocyte; GA, Golgi apparatus; Mt, mitochondria; N, nucleus; S, sinusoid.

These non-parenchymal cell populations can be identified by a variety of cell surface markers. In general, KCs and RHMs both express epidermal growth factor-like module-containing mucin-like hormone receptor-like 1 (F4/80) (35), NKs form two pools distinguished by mutually exclusive expression of CD49a or DX5 (36), HSCs express glial fibrillary acidic protein (GFAP) (37, 38), and LSECs express CD34 (39). In addition, these liver cell populations can also be distinguished by their physical location within the liver and specific ultrastructural characteristics. For example, hepatocytes contain many microvilli, which project into space of Disse (perisinusoidal space) between the endothelial cells and hepatocytes. KCs (~15% of all liver cells) represent the largest population of tissue macrophages (80–90% of resident macrophages in the whole body) (40). KCs are found attached to the luminal surface or inserted in the endothelial lining of hepatic sinusoids (41, 42), which make them the first macrophages to come into contact with gut-derived foreign and potentially noxious material. The size and function of KCs also depend on their specific location in the liver (43) with KCs in periportal regions being larger and more phagocytic with higher lysosomal enzyme activity than KCs in midzonal and perivenous locations (44). Unlike hepatocytes, KCs are amoeboid in shape. Fenestrae form open connections between the lumen of the sinusoid and the space of Disse (45). The transport and exchange of fluid, solutes, and particles between the sinusoidal lumen and the space of Disse containing the parenchymal cell surface are believed to occur through these open fenestrae (46). While KCs utilize phagocytosis to incorporate large particles such as erythrocytes and bacteria, they take up small particles and molecules via pinocytic vesicles (4750). NKs reside in sinusoids and eliminate virus-infected or transformed cells and regulate adaptive immune responses via contact-dependent signals and the secretion of cytokines (36, 5153). HSCs are perisinusoidal cells, which contain characteristic lipid droplets. HSCs maintain vitamin A homeostasis as they store 80% of total vitamin A in the body. Inflammatory signals transform HSCs into myofibroblasts, resulting in collagen production and development of liver fibrosis (54, 55). LSECs possess a high-rate, high-capacity system to remove colloids and water-soluble waster macromolecules from the circulation (34, 56). At the ultrastructural level, LSECs constitute the only mammalian endothelial cells that combine non-diaphragmed fenestrae with a discontinuous basement membrane, which allows blood plasma to enter the space of Disse.

Liver Macrophage Populations During Obesity

During the course of obesity, the adipose tissue’s ability to store excess energy is compromised, leading to ectopic lipid accumulation in non-adipose tissues such as muscle and liver (57). Intracellular lipid accumulation in ectopic tissues is associated with a phenomenon known as lipotoxicity, which induces cell death, cytokine secretion, and activation of inflammatory processes, especially in the liver (58, 59). Furthermore, dietary stress and obesity can lead to excessive activation of the hepatic immune system due to increased penetration of microbial material (6062). The response of the liver to damage and inflammation is a complex process involving parenchymal (hepatocytes) and non-parenchymal cells (KCs, NKs, HSCs, and LSECs), as well as monocyte-derived hepatic macrophages, RHMs (12, 63). The failure to regulate this inflammation during the progression of the obesity causes pathological chronic hepatic inflammation characterized by the advance of fatty liver to steatohepatitis, fibrosis, cirrhosis, and eventually liver failure (18, 64). Depletion of phagocytic cells in the liver (including both KCs and RHMs) through the administration of either liposome-encapsulated clodronate or gadolinium chloride protects against high-fat- or high-sucrose-induced steatosis, inflammation, and IR, demonstrating critical role of hepatic macrophages in the development of metabolic dysfunction (65).

Macrophage Regulation During NAFLD/NASH

Hepatic lipid accumulation and peroxidation leads to chronic hepatocyte endoplasmic reticulum (ER) stress, the production of reactive oxygen species (ROS), and toll-like receptor (TLR) activation, which converts KCs into an M1 phenotype defined by production of pro-inflammatory cytokines, oncostatin, and prostaglandins (Figure 2) (20, 66, 67). Circulating cytokines, adipokines, and free fatty acids (FFAs) released from inflamed adipose tissue in the obese state or immunogenic material derived from an altered intestinal microbiota can also contribute to KC polarization. M1-KCs secrete chemokine (C-C motif) ligand 2 (CCL2), pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6), macrophage inflammatory protein (MIP)-1a, MIP1b, RANTES, oncostatin, and prostaglandins (PGE2), which contribute to the alteration of the liver homeostasis and worsen the hepatic inflammatory response (29). PGE2 regulates cytokine production (IL-1, IL-6, TNF-α, and TGF-β) (68, 69), acts synergistically with IL-6 to induce IR (70), and induces production of oncostatin M (OSM) in KCs (71). Increased OSM contributes to hepatic IR and the development of non-alcoholic steato hepatitis (NASH) (71). High levels of TNF-α released by M1-KCs stimulate hepatic expression of CCL2 (also known as MCP1), a powerful monocyte chemoattractant, which recruits CCR2+Ly6Chigh monocytes from the vasculature into the liver (72), where they differentiate into Ly6Chigh macrophages. The Ly6Chigh macrophages amplify the severity of obesity-induced inflammation and hepatic IR through the secretion of TNF-α and interleukin 6 (IL-6) (12). C-C chemokine receptor type 2 (CCR2)-deficient mice are protected against weight gain and display reduced development of obesity, illustrating the importance of this chemokine system (73). Once established, this vicious circle of immune cell attraction, infiltration and activation, hepatocyte injury, and further inflammation promotes and defines the pathophysiology of NASH (74).

FIGURE 2
www.frontiersin.org

Figure 2. Schematic diagram showing the effects of resident (KC) and recruited hepatic macrophages (Ly6Chigh) in regulation of non-alcoholic fatty liver disease (NAFLD) and fibrosis. (A) Healthy liver showing parenchymal hepatocytes (HC) and non-parenchymal Kupffer cells (M2-KC), natural killer cells (NK), hepatic stellate cells (HSC), and liver sinusoidal endothelial cells (LSEC). (B) Healthy adipose tissue showing adipocytes (AC), adipocyte macrophage 1 (ATM1), and ATM2 macrophages. (C) Obese liver showing accumulation of lipid droplets in hepatocytes (HC), activated Kupffer cells (M1-KC), and activated hepatic stellate cells (HSC). Note increased production of TNF-α, IL-1β, IFNγ, ROS, and CCL2. (D) Obese adipose tissue showing larger adipocytes (AC), infiltrated ATM1 macrophages, and increased production of TNF-α and IL-1β. (E) Obese liver showing NAFLD and NASH. (F) Obese liver showing fibrosis. Increased production of CCL2 recruits Ly6Chigh monocytes, which convert to Ly6Chigh macrophages inside the liver. Ly6Chigh macrophages produce TGFβ, connective tissue growth factor (CTGF), and PDGF, which act on HSC and transform HSC to activated myofibroblast. Activated myofibroblast in turn results in fibrosis. Ly6Chigh macrophage is transformed into Ly6Clow macrophage upon eating dead hepatocytes and erythrocytes. Ly6Clow macrophage deactivates activated myofibroblasts and decrease fibrosis.

Macrophage Regulation of Hepatic Fibrosis

Fibrosis is increasingly appreciated as a major contributor to metabolic dysregulation in obese humans and type 2 diabetic patients (75). Both KCs and recruited Ly6Chigh macrophages contribute to the development of hepatic fibrosis. KCs activate HSCs through increased production of pro-fibrotic cytokine TGF-β and platelet-derived growth factor (PDGF) (76) leading to fibrosis. Ly6Chigh macrophages also interact with HSCs to promote fibrosis through increased production of TGF-β, connective tissue growth factor (CTGF), and PDGF (77). Therefore, inhibition of monocyte recruitment through depletion of the pro-inflammatory signal CCL2 results in attenuation of liver fibrosis (7779). In addition, pharmacological inhibition of CCL2 by the RNA-aptamer mNOX-E36 attenuates liver fibrosis, thereby strengthening a pro-fibrotic function of Ly6Chigh macrophages (80, 81).

Macrophage Surface Markers

Due to the distinct functions of RHMs and KCs in suppressing or perpetuating the immune activation (29, 82), it is important to be able to clearly isolate pure populations of each cell type. However, distinguishing RHM from KC has proven difficult mainly due to technical difficulties in isolating and identifying macrophages from the obese liver. KCs (CXCR1) appear histologically as larger cells with multiple phagocytic granules and have been defined by surface marker expression as CD45+/CD11c/F4/80high/CD11blow (12, 83). RHMs (CXCR1+) are smaller than KC, contain fine granules in the cytoplasm, and have been defined by surface marker expression as F4/80dim/CD45+/CD11b+/CCR2+ (10), CD11b+/Ly6Chigh/Ly6G (83), or CD45+/CD11c/F4/80low/CD11bhigh markers (83) depending on the publication. However, these factors alone do not sufficiently identify pure KC or RHM populations as there is significant size and surface marker overlap with other cell populations, including dendritic cells (DCs), eosinophils, and undifferentiated monocytes (84). KCs, unlike RHMs, have the unique ability to survive to lethal irradiation (85), which has enabled studies into these distinct cell types. The result of these investigations suggests that the number of KCs remains unchanged during the course of obesity, whereas accumulation of RHMs increases several-fold (12). Transcriptome analysis of these RHM and KC populations isolated from lean and diet-induced obese (DIO) mice revealed statistically marked differences between the two cell types on both diets. Furthermore, the Gene Ontology analysis of these transcriptomes showed a restricted list of 16 KC marker genes and 11 RHM markers genes differentially expressed from lean to DIO mice that could provide the opportunity for direct isolation strategies using specific surface markers (12). Interestingly, factors secreted in the culture media from isolated high fat diet (HFD)-RHMs, but not from isolated HFD-KCs, can promote hepatic glucose output and attenuate insulin’s normal inhibitory effects on this aspect of hepatic metabolism suggesting that RHMs are the dominant immune cell type inducing hepatic IR (12, 82).

Hepatic Gene Expression Changes During Obesity

To identify potential mechanisms underlying the development of obesity and diabetes, many studies have been conducted to characterize changes in hepatic gene expression (8691). Complex phenotypes such as obesity and IR involve many different interacting biological pathways, but recent technological advances in high throughput sequencing have greatly improved our ability to quantitatively detect gene expression changes in an unbiased way. Investigation of the hepatic gene expression profiles in obese db/db (leptin receptor deficient) mice compared with control mice revealed significant changes in lipid metabolism, gluconeogenesis, mitochondrial dysfunction, and oxidative stress (88, 89). Similar studies using HFD feeding to generate obesity resulted in increased hepatic expression of genes involved in fatty acid catabolism and ketone body synthesis, such as acyl-CoA oxidase1 (Acox1) and HMG-CoA lyase (Hmgcl), while genes involved in lipogenesis and cholesterol synthesis, such as fatty acid synthase (Fasn) and acetyl-CoA synthetase 2 (Acsl6), were drastically decreased in the HFD group (86). Further studies also identified upregulation of hepatic gluconeogenic genes and downregulation of expression of lipogenic genes in diabetic Zucker rats (92), with activation of distinct transcriptional regulatory networks during diabetic progression (93).

Due to the practical limitations in obtaining human liver tissue, the most detailed hepatic expression studies have, so far, been conducted in rodent models (8689, 92, 93). However, with the increasing use of gastric bypass surgery in obese patients, obtaining liver biopsies has become more feasible (91). Comparison of hepatic gene expression before and after weight loss in morbidly obese women identified differentially expressed genes involved in lipid and energy homeostasis, pro-inflammatory tissue repair, and bile acid transport (91). Liver samples from morbidly obese patients with all stages of NAFLD and controls were analyzed by array, and NAFLD specific expression differences were seen for nine genes involved in intermediate metabolism including pyruvate carboxylase (Pc), ATP citrate lyase (Acly), and phospholipase C-gamma-1 (Plcg1) as well as insulin/insulin-like signaling including insulin-like growth factor-1 (Igf1), insulin-like growth factor binding protein 2 (Igfbp2), and protein kinase C epsilon (Prkce) (94). In additional studies, comparison of transcriptional profiles from NASH patients versus non-obese controls also revealed significant changes in genes involved in metabolism, insulin signaling, and inflammation (90). For example, high levels of the central enzyme controlling unesterified arachidonic acid levels of Acyl-CoA synthetase long chain family member 4 (Acsl4) and lower levels of insulin signaling genes including Igfbp2 were observed in NASH versus non-obese controls (95).

Therefore, many hepatic gene expression studies in rodents and humans have been conducted at the level of the whole liver, but whether these changes occur within the hepatocyte or non-parenchymal cells is yet to be fully investigated. Increased understanding of the changes induced in the obese state in the hepatocytes, liver-resident macrophages, and each immune cell population may allow us to specifically target potentially harmful populations while promoting anti-inflammatory populations (96). These studies will also help clarify the molecular mechanisms behind the development of IR and identify potential targets for therapeutic intervention. Furthermore, future integration of transcriptomics data with metabolomics and proteomics data will further our understanding of the mechanisms behind obesity-associated liver disease and help identify biomarkers for the development of disease progression (89).

Conclusion and Future Perspectives

Although KCs are reemerging in obesity and metabolic syndrome as a critical player in the onset of hepatic IR, as well as NAFLD, their role in metabolism is still largely unknown. We are yet to define the direct role of KCs in metabolic diseases as well as their interactions with neighboring cells and distant organs that modulate liver function and whole body metabolism. After a hepatic insult, KCs secrete important factors involved in the recruitment and transformation of blood monocytes, which are involved in the subsequent development of the hepatic IR. During obesity, the inflammatory state in the liver is associated with a large increase in RHMs with a M1 phenotype, targeting specifically these immune cells or manipulating the activation of KC may be an effective therapeutic strategy in obesity-related chronic liver and NASH. The use of new technologies such as next-generation or single-cell sequencing at different stages of obesity and IR and approaches to isolate and identify the diverse macrophage population and profile their transcriptomes in the liver could provide the opportunity for a direct targeting strategy using specific surface markers. Further research in the field of immunometabolism, including a better understanding of how changes in the microbiota affect the development of inflammation and more knowledge about the factors that direct the polarization state of macrophages toward either the pro- or anti-inflammatory state, is necessary to design new therapeutic strategies for treating T2D and NAFLD.

Author Contributions

SM and RM conceived the idea. RM, AJ, OO, PT, and SM contributed equally to researching the data and writing of the manuscript.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Funding

The research was supported by Mahata’s personal funding.

References

1. Fotbolcu H, Zorlu E. Nonalcoholic fatty liver disease as a multi-systemic disease. World J Gastroenterol (2016) 22(16):4079–90. doi:10.3748/wjg.v22.i16.4079

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science (1993) 259(5091):87–91. doi:10.1126/science.7678183

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest (2003) 112(12):1796–808. doi:10.1172/JCI200319246

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest (2003) 112(12):1821–30. doi:10.1172/JCI200319451

PubMed Abstract | CrossRef Full Text | Google Scholar

5. McNelis JC, Olefsky JM. Macrophages, immunity, and metabolic disease. Immunity (2014) 41(1):36–48. doi:10.1016/j.immuni.2014.05.010

CrossRef Full Text | Google Scholar

6. Varma V, Yao-Borengasser A, Rasouli N, Nolen GT, Phanavanh B, Starks T, et al. Muscle inflammatory response and insulin resistance: synergistic interaction between macrophages and fatty acids leads to impaired insulin action. Am J Physiol Endocrinol Metab (2009) 296(6):E1300–10. doi:10.1152/ajpendo.90885.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Fink LN, Costford SR, Lee YS, Jensen TE, Bilan PJ, Oberbach A, et al. Pro-inflammatory macrophages increase in skeletal muscle of high fat-fed mice and correlate with metabolic risk markers in humans. Obesity (Silver Spring) (2014) 22(3):747–57. doi:10.1002/oby.20615

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Patsouris D, Cao JJ, Vial G, Bravard A, Lefai E, Durand A, et al. Insulin resistance is associated with MCP1-mediated macrophage accumulation in skeletal muscle in mice and humans. PLoS One (2014) 9(10):e110653. doi:10.1371/journal.pone.0110653

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Boon MR, Bakker LE, Haks MC, Quinten E, Schaart G, Van Beek L, et al. Short-term high-fat diet increases macrophage markers in skeletal muscle accompanied by impaired insulin signalling in healthy male subjects. Clin Sci (Lond) (2015) 128(2):143–51. doi:10.1042/CS20140179

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Obstfeld AE, Sugaru E, Thearle M, Francisco AM, Gayet C, Ginsberg HN, et al. C-C chemokine receptor 2 (CCR2) regulates the hepatic recruitment of myeloid cells that promote obesity-induced hepatic steatosis. Diabetes (2010) 59(4):916–25. doi:10.2337/db09-1403

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Bian Z, Peng Y, You Z, Wang Q, Miao Q, Liu Y, et al. CCN1 expression in hepatocytes contributes to macrophage infiltration in nonalcoholic fatty liver disease in mice. J Lipid Res (2013) 54(1):44–54. doi:10.1194/jlr.M026013

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Morinaga H, Mayoral R, Heinrichsdorff J, Osborn O, Franck N, Hah N, et al. Characterization of distinct subpopulations of hepatic macrophages in HFD/obese mice. Diabetes (2015) 64(4):1120–30. doi:10.2337/db14-1238

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Ehses JA, Perren A, Eppler E, Ribaux P, Pospisilik JA, Maor-Cahn R, et al. Increased number of islet-associated macrophages in type 2 diabetes. Diabetes (2007) 56(9):2356–70. doi:10.2337/db06-1650

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Richardson SJ, Willcox A, Bone AJ, Foulis AK, Morgan NG. Islet-associated macrophages in type 2 diabetes. Diabetologia (2009) 52(8):1686–8. doi:10.1007/s00125-009-1410-z

CrossRef Full Text | Google Scholar

15. Racanelli V, Rehermann B. The liver as an immunological organ. Hepatology (2006) 43(2 Suppl 1):S54–62. doi:10.1002/hep.21060

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Nemeth E, Baird AW, O’Farrelly C. Microanatomy of the liver immune system. Semin Immunopathol (2009) 31(3):333–43. doi:10.1007/s00281-009-0173-4

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Crispe IN. The liver as a lymphoid organ. Annu Rev Immunol (2009) 27:147–63. doi:10.1146/annurev.immunol.021908.132629

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Robinson MW, Harmon C, O’Farrelly C. Liver immunology and its role in inflammation and homeostasis. Cell Mol Immunol (2016) 13(3):267–76. doi:10.1038/cmi.2016.3

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Cai D, Yuan M, Frantz DF, Melendez PA, Hansen L, Lee J, et al. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat Med (2005) 11(2):183–90. doi:10.1038/nm1166

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Jager J, Aparicio-Vergara M, Aouadi M. Liver innate immune cells and insulin resistance: the multiple facets of Kupffer cells. J Intern Med (2016) 280(2):209–20. doi:10.1111/joim.12483

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Jacob AI, Goldberg PK, Bloom N, Degenshein GA, Kozinn PJ. Endotoxin and bacteria in portal blood. Gastroenterology (1977) 72(6):1268–70.

PubMed Abstract | Google Scholar

22. Knolle P, Schlaak J, Uhrig A, Kempf P, Meyer zum Buschenfelde KH, Gerken G. Human Kupffer cells secrete IL-10 in response to lipopolysaccharide (LPS) challenge. J Hepatol (1995) 22(2):226–9. doi:10.1016/0168-8278(95)80433-1

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Devisscher L, Verhelst X, Colle I, Van Vlierberghe H, Geerts A. The role of macrophages in obesity-driven chronic liver disease. J Leukoc Biol (2016) 99(5):693–8. doi:10.1189/jlb.5RU0116-016R

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Varol C, Mildner A, Jung S. Macrophages: development and tissue specialization. Annu Rev Immunol (2015) 33:643–75. doi:10.1146/annurev-immunol-032414-112220

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep (2014) 6:13. doi:10.12703/P6-13

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Galvan-Pena S, O’Neill LA. Metabolic reprograming in macrophage polarization. Front Immunol (2014) 5:420. doi:10.3389/fimmu.2014.00420

CrossRef Full Text | Google Scholar

27. Kang K, Reilly SM, Karabacak V, Gangl MR, Fitzgerald K, Hatano B, et al. Adipocyte-derived Th2 cytokines and myeloid PPARdelta regulate macrophage polarization and insulin sensitivity. Cell Metab (2008) 7(6):485–95. doi:10.1016/j.cmet.2008.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Odegaard JI, Ricardo-Gonzalez RR, Red Eagle A, Vats D, Morel CR, Goforth MH, et al. Alternative M2 activation of Kupffer cells by PPARdelta ameliorates obesity-induced insulin resistance. Cell Metab (2008) 7(6):496–507. doi:10.1016/j.cmet.2008.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Dey A, Allen J, Hankey-Giblin PA. Ontogeny and polarization of macrophages in inflammation: blood monocytes versus tissue macrophages. Front Immunol (2015) 5:683. doi:10.3389/fimmu.2014.00683

CrossRef Full Text | Google Scholar

30. Widmann JJ, Cotran RS, Fahimi HD. Mononuclear phagocytes (Kupffer cells) and endothelial cells. Identification of two functional cell types in rat liver sinusoids by endogenous peroxidase activity. J Cell Biol (1972) 52(1):159–70. doi:10.1083/jcb.52.1.159

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, et al. Innate or adaptive immunity? The example of natural killer cells. Science (2011) 331(6013):44–9. doi:10.1126/science.1198687

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Yokoyama WM, Sojka DK, Peng H, Tian Z. Tissue-resident natural killer cells. Cold Spring Harb Symp Quant Biol (2013) 78:149–56. doi:10.1101/sqb.2013.78.020354

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Hautekeete ML, Geerts A. The hepatic stellate (Ito) cell: its role in human liver disease. Virchows Arch (1997) 430(3):195–207. doi:10.1007/BF01324802

PubMed Abstract | CrossRef Full Text | Google Scholar

34. DeLeve LD. Liver sinusoidal endothelial cells and liver regeneration. J Clin Invest (2013) 123(5):1861–6. doi:10.1172/JCI66025

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Austyn JM, Gordon S. F4/80, a monoclonal antibody directed specifically against the mouse macrophage. Eur J Immunol (1981) 11(10):805–15. doi:10.1002/eji.1830111013

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Peng H, Jiang X, Chen Y, Sojka DK, Wei H, Gao X, et al. Liver-resident NK cells confer adaptive immunity in skin-contact inflammation. J Clin Invest (2013) 123(4):1444–56. doi:10.1172/JCI66381

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Neubauer K, Knittel T, Aurisch S, Fellmer P, Ramadori G. Glial fibrillary acidic protein – a cell type specific marker for Ito cells in vivo and in vitro. J Hepatol (1996) 24(6):719–30. doi:10.1016/S0168-8278(96)80269-8

CrossRef Full Text | Google Scholar

38. Gard AL, White FP, Dutton GR. Extra-neural glial fibrillary acidic protein (GFAP) immunoreactivity in perisinusoidal stellate cells of rat liver. J Neuroimmunol (1985) 8(4–6):359–75. doi:10.1016/S0165-5728(85)80073-4

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Baratta JL, Ngo A, Lopez B, Kasabwalla N, Longmuir KJ, Robertson RT. Cellular organization of normal mouse liver: a histological, quantitative immunocytochemical, and fine structural analysis. Histochem Cell Biol (2009) 131(6):713–26. doi:10.1007/s00418-009-0577-1

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Bouwens L, Baekeland M, De Zanger R, Wisse E. Quantitation, tissue distribution and proliferation kinetics of Kupffer cells in normal rat liver. Hepatology (1986) 6(4):718–22. doi:10.1002/hep.1840060430

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Wisse E. Observations on the fine structure and peroxidase cytochemistry of normal rat liver Kupffer cells. J Ultrastruct Res (1974) 46(3):393–426. doi:10.1016/S0022-5320(74)90064-1

CrossRef Full Text | Google Scholar

42. Wisse E. Kupffer cell reactions in rat liver under various conditions as observed in the electron microscope. J Ultrastruct Res (1974) 46(3):499–520. doi:10.1016/S0022-5320(74)90070-7

CrossRef Full Text | Google Scholar

43. Laskin DL, Weinberger B, Laskin JD. Functional heterogeneity in liver and lung macrophages. J Leukoc Biol (2001) 70(2):163–70.

PubMed Abstract | Google Scholar

44. Sleyster EC, Knook DL. Relation between localization and function of rat liver Kupffer cells. Lab Invest (1982) 47(5):484–90.

PubMed Abstract | Google Scholar

45. Hickey MJ, Kubes P. Intravascular immunity: the host-pathogen encounter in blood vessels. Nat Rev Immunol (2009) 9(5):364–75. doi:10.1038/nri2532

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Wisse E, De Zanger RB, Charels K, Van Der Smissen P, McCuskey RS. The liver sieve: considerations concerning the structure and function of endothelial fenestrae, the sinusoidal wall and the space of Disse. Hepatology (1985) 5(4):683–92. doi:10.1002/hep.1840050427

CrossRef Full Text | Google Scholar

47. Bouwens L, Geerts A, Van Bossuyt H, Wisse E. Recent insights into the function of hepatic sinusoidal cells. Neth J Med (1987) 31(3–4):129–48.

Google Scholar

48. Wake K, Decker K, Kirn A, Knook DL, McCuskey RS, Bouwens L, et al. Cell biology and kinetics of Kupffer cells in the liver. Int Rev Cytol (1989) 118:173–229. doi:10.1016/S0074-7696(08)60875-X

CrossRef Full Text | Google Scholar

49. Toth CA, Thomas P. Liver endocytosis and Kupffer cells. Hepatology (1992) 16(1):255–66. doi:10.1002/hep.1840160137

CrossRef Full Text | Google Scholar

50. Smedsrod B, De Bleser PJ, Braet F, Lovisetti P, Vanderkerken K, Wisse E, et al. Cell biology of liver endothelial and Kupffer cells. Gut (1994) 35(11):1509–16. doi:10.1136/gut.35.11.1509

CrossRef Full Text | Google Scholar

51. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol (2008) 9(5):503–10. doi:10.1038/ni1582

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Fasbender F, Widera A, Hengstler JG, Watzl C. Natural killer cells and liver fibrosis. Front Immunol (2016) 7:19. doi:10.3389/fimmu.2016.00019

CrossRef Full Text | Google Scholar

53. Peng H, Wisse E, Tian Z. Liver natural killer cells: subsets and roles in liver immunity. Cell Mol Immunol (2016) 13(3):328–36. doi:10.1038/cmi.2015.96

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Sato M, Suzuki S, Senoo H. Hepatic stellate cells: unique characteristics in cell biology and phenotype. Cell Struct Funct (2003) 28(2):105–12. doi:10.1247/csf.28.105

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Li JT, Liao ZX, Ping J, Xu D, Wang H. Molecular mechanism of hepatic stellate cell activation and antifibrotic therapeutic strategies. J Gastroenterol (2008) 43(6):419–28. doi:10.1007/s00535-008-2180-y

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Elvevold K, Smedsrod B, Martinez I. The liver sinusoidal endothelial cell: a cell type of controversial and confusing identity. Am J Physiol Gastrointest Liver Physiol (2008) 294(2):G391–400. doi:10.1152/ajpgi.00167.2007

PubMed Abstract | CrossRef Full Text | Google Scholar

57. van Herpen NA, Schrauwen-Hinderling VB. Lipid accumulation in non-adipose tissue and lipotoxicity. Physiol Behav (2008) 94(2):231–41. doi:10.1016/j.physbeh.2007.11.049

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Wree A, Kahraman A, Gerken G, Canbay A. Obesity affects the liver – the link between adipocytes and hepatocytes. Digestion (2011) 83(1–2):124–33. doi:10.1159/000318741

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Neuschwander-Tetri BA. Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology (2010) 52(2):774–88. doi:10.1002/hep.23719

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature (2012) 482(7384):179–85. doi:10.1038/nature10809

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Johnson AM, Costanzo A, Gareau MG, Armando AM, Quehenberger O, Jameson JM, et al. High fat diet causes depletion of intestinal eosinophils associated with intestinal permeability. PLoS One (2015) 10(4):e0122195. doi:10.1371/journal.pone.0122195

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Llorente C, Schnabl B. The gut microbiota and liver disease. Cell Mol Gastroenterol Hepatol (2015) 1(3):275–84. doi:10.1016/j.jcmgh.2015.04.003

CrossRef Full Text | Google Scholar

63. Gressner AM, Bachem MG. Molecular mechanisms of liver fibrogenesis – a homage to the role of activated fat-storing cells. Digestion (1995) 56(5):335–46. doi:10.1159/000201257

CrossRef Full Text | Google Scholar

64. Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism (2016) 65(8):1038–48. doi:10.1016/j.metabol.2015.12.012

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Gruben N, Shiri-Sverdlov R, Koonen DP, Hofker MH. Nonalcoholic fatty liver disease: a main driver of insulin resistance or a dangerous liaison? Biochim Biophys Acta (2014) 1842(11):2329–43. doi:10.1016/j.bbadis.2014.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Dixon LJ, Barnes M, Tang H, Pritchard MT, Nagy LE. Kupffer cells in the liver. Compr Physiol (2013) 3(2):785–97. doi:10.1002/cphy.c120026

CrossRef Full Text | Google Scholar

67. Kolios G, Valatas V, Kouroumalis E. Role of Kupffer cells in the pathogenesis of liver disease. World J Gastroenterol (2006) 12(46):7413–20. doi:10.3748/wjg.v12.i46.7413

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Goss JA, Mangino MJ, Callery MP, Flye MW. Prostaglandin E2 downregulates Kupffer cell production of IL-1 and IL-6 during hepatic regeneration. Am J Physiol (1993) 264(4 Pt 1):G601–8.

PubMed Abstract | Google Scholar

69. Roland CR, Goss JA, Mangino MJ, Hafenrichter D, Flye MW. Autoregulation by eicosanoids of human Kupffer cell secretory products. A study of interleukin-1, interleukin-6, tumor necrosis factor-alpha, transforming growth factor-beta, and nitric oxide. Ann Surg (1994) 219(4):389–99. doi:10.1097/00000658-199404000-00010

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Henkel J, Neuschafer-Rube F, Pathe-Neuschafer-Rube A, Puschel GP. Aggravation by prostaglandin E2 of interleukin-6-dependent insulin resistance in hepatocytes. Hepatology (2009) 50(3):781–90. doi:10.1002/hep.23064

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Henkel J, Gartner D, Dorn C, Hellerbrand C, Schanze N, Elz SR, et al. Oncostatin M produced in Kupffer cells in response to PGE2: possible contributor to hepatic insulin resistance and steatosis. Lab Invest (2011) 91(7):1107–17. doi:10.1038/labinvest.2011.47

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Klueh U, Czajkowski C, Ludzinska I, Qiao Y, Frailey J, Kreutzer DL. Impact of CCL2 and CCR2 chemokine/receptor deficiencies on macrophage recruitment and continuous glucose monitoring in vivo. Biosens Bioelectron (2016) 86:262–9. doi:10.1016/j.bios.2016.06.026

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest (2006) 116(1):115–24. doi:10.1172/JCI24335C1

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Tosello-Trampont AC, Landes SG, Nguyen V, Novobrantseva TI, Hahn YS. Kuppfer cells trigger nonalcoholic steatohepatitis development in diet-induced mouse model through tumor necrosis factor-alpha production. J Biol Chem (2012) 287(48):40161–72. doi:10.1074/jbc.M112.417014

CrossRef Full Text | Google Scholar

75. Sun K, Tordjman J, Clement K, Scherer PE. Fibrosis and adipose tissue dysfunction. Cell Metab (2013) 18(4):470–7. doi:10.1016/j.cmet.2013.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Pradere JP, Kluwe J, De Minicis S, Jiao JJ, Gwak GY, Dapito DH, et al. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology (2013) 58(4):1461–73. doi:10.1002/hep.26429

CrossRef Full Text | Google Scholar

77. Karlmark KR, Weiskirchen R, Zimmermann HW, Gassler N, Ginhoux F, Weber C, et al. Hepatic recruitment of the inflammatory Gr1+ monocyte subset upon liver injury promotes hepatic fibrosis. Hepatology (2009) 50(1):261–74. doi:10.1002/hep.22950

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Baeck C, Wehr A, Karlmark KR, Heymann F, Vucur M, Gassler N, et al. Pharmacological inhibition of the chemokine CCL2 (MCP-1) diminishes liver macrophage infiltration and steatohepatitis in chronic hepatic injury. Gut (2012) 61(3):416–26. doi:10.1136/gutjnl-2011-300304

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Ehling J, Bartneck M, Wei X, Gremse F, Fech V, Mockel D, et al. CCL2-dependent infiltrating macrophages promote angiogenesis in progressive liver fibrosis. Gut (2014) 63(12):1960–71. doi:10.1136/gutjnl-2013-306294

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Baeck C, Wei X, Bartneck M, Fech V, Heymann F, Gassler N, et al. Pharmacological inhibition of the chemokine C-C motif chemokine ligand 2 (monocyte chemoattractant protein 1) accelerates liver fibrosis regression by suppressing Ly-6C(+) macrophage infiltration in mice. Hepatology (2014) 59(3):1060–72. doi:10.1002/hep.26783

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Zimmermann HW, Tacke F. In search of the magic bullet: can liver inflammation and fibrosis be reversed with medications? Expert Rev Gastroenterol Hepatol (2015) 9(9):1139–41. doi:10.1586/17474124.2015.1063417

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Lackey DE, Olefsky JM. Regulation of metabolism by the innate immune system. Nat Rev Endocrinol (2016) 12(1):15–28. doi:10.1038/nrendo.2015.189

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Movita D, Kreefft K, Biesta P, van Oudenaren A, Leenen PJ, Janssen HL, et al. Kupffer cells express a unique combination of phenotypic and functional characteristics compared with splenic and peritoneal macrophages. J Leukoc Biol (2012) 92(4):723–33. doi:10.1189/jlb.1111566

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Lloyd CM, Phillips AR, Cooper GJ, Dunbar PR. Three-colour fluorescence immunohistochemistry reveals the diversity of cells staining for macrophage markers in murine spleen and liver. J Immunol Methods (2008) 334(1–2):70–81. doi:10.1016/j.jim.2008.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Klein I, Cornejo JC, Polakos NK, John B, Wuensch SA, Topham DJ, et al. Kupffer cell heterogeneity: functional properties of bone marrow derived and sessile hepatic macrophages. Blood (2007) 110(12):4077–85. doi:10.1182/blood-2007-02-073841

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Kim S, Sohn I, Ahn JI, Lee KH, Lee YS, Lee YS. Hepatic gene expression profiles in a long-term high-fat diet-induced obesity mouse model. Gene (2004) 340(1):99–109. doi:10.1016/j.gene.2004.06.015

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Kelder T, Eijssen L, Kleemann R, van Erk M, Kooistra T, Evelo C. Exploring pathway interactions in insulin resistant mouse liver. BMC Syst Biol (2011) 5:127. doi:10.1186/1752-0509-5-127

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Zhang F, Xu X, Zhang Y, Zhou B, He Z, Zhai Q. Gene expression profile analysis of type 2 diabetic mouse liver. PLoS One (2013) 8(3):e57766. doi:10.1371/journal.pone.0057766

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Connor SC, Hansen MK, Corner A, Smith RF, Ryan TE. Integration of metabolomics and transcriptomics data to aid biomarker discovery in type 2 diabetes. Mol Biosyst (2010) 6(5):909–21. doi:10.1039/b914182k

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Wruck W, Kashofer K, Rehman S, Daskalaki A, Berg D, Gralka E, et al. Multi-omic profiles of human non-alcoholic fatty liver disease tissue highlight heterogenic phenotypes. Sci Data (2015) 2:150068. doi:10.1038/sdata.2015.68

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Elam MB, Cowan GS Jr, Rooney RJ, Hiler ML, Yellaturu CR, Deng X, et al. Hepatic gene expression in morbidly obese women: implications for disease susceptibility. Obesity (Silver Spring) (2009) 17(8):1563–73. doi:10.1038/oby.2009.49

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Suh YH, Kim Y, Bang JH, Choi KS, Lee JW, Kim WH, et al. Analysis of gene expression profiles in insulin-sensitive tissues from pre-diabetic and diabetic Zucker diabetic fatty rats. J Mol Endocrinol (2005) 34(2):299–315. doi:10.1677/jme.1.01679

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Zhou H, Saito S, Piao G, Liu ZP, Wang J, Horimoto K, et al. Network screening of Goto-Kakizaki rat liver microarray data during diabetic progression. BMC Syst Biol (2011) 5(Suppl 1):S16. doi:10.1186/1752-0509-5-S1-S16

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Ahrens M, Ammerpohl O, von Schonfels W, Kolarova J, Bens S, Itzel T, et al. DNA methylation analysis in nonalcoholic fatty liver disease suggests distinct disease-specific and remodeling signatures after bariatric surgery. Cell Metab (2013) 18(2):296–302. doi:10.1016/j.cmet.2013.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Younossi ZM, Gorreta F, Ong JP, Schlauch K, Del Giacco L, Elariny H, et al. Hepatic gene expression in patients with obesity-related non-alcoholic steatohepatitis. Liver Int (2005) 25(4):760–71. doi:10.1111/j.1478-3231.2005.01117.x

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Reid DT, Reyes JL, McDonald BA, Vo T, Reimer RA, Eksteen B. Kupffer cells undergo fundamental changes during the development of experimental NASH and are critical in initiating liver damage and inflammation. PLoS One (2016) 11(7):e0159524. doi:10.1371/journal.pone.0159524

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: obesity, insulin resistance, inflammation, hepatocytes, Kupffer cells, immunometabolism

Citation: Mayoral Monibas R, Johnson AMF, Osborn O, Traves PG and Mahata SK (2016) Distinct Hepatic Macrophage Populations in Lean and Obese Mice. Front. Endocrinol. 7:152. doi: 10.3389/fendo.2016.00152

Received: 30 September 2016; Accepted: 22 November 2016;
Published: 06 December 2016

Edited by:

Timo Dirk Müller, Helmholtz Zentrum München, Germany

Reviewed by:

Zhihong Yang, University of Fribourg, Switzerland
Victor Costa Castro-Alves, University of São Paulo, Brazil

Copyright: © 2016 Mayoral Monibas, Johnson, Osborn, Traves and Mahata. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Rafael Mayoral Monibas, rafael.mayoral.monibas@merck.com;
Sushil K. Mahata, smahata@ucsd.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.