Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Feb 27, 2025; 17(2): 102328
Published online Feb 27, 2025. doi: 10.4254/wjh.v17.i2.102328
Role of triggering receptor expressed on myeloid cells 2 in the pathogenesis of non-alcoholic fatty liver disease
Li-Hui Zhang, Su-Tong Liu, Qing Zhao, Xiao-Yan Liu, Tong Liu, Qiang Zhang, Ming-Hao Liu, Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
Li-Hui Zhang, Su-Tong Liu, Ming-Hao Liu, Wen-Xia Zhao, Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
Wen-Xia Zhao, Department of Spleen, Stomach, Liver and Gallbladder Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
ORCID number: Wen-Xia Zhao (0000-0001-6666-9469).
Co-first authors: Li-Hui Zhang and Su-Tong Liu.
Author contributions: Zhang LH and Liu ST drafted the manuscript; Zhao Q and Liu XY contributed to literature search; Liu T and Zhang Q organized the references; Liu MH and Zhao WX reviewed and edited the manuscript; all authors have read and approved the final version to be published.
Supported by Henan Province's "Double First-Class" Creation of Scientific Research in Traditional Chinese Medicine, No. HSRP-DFCTCM-2023-7-23 and No. STG-ZYX02-202117; National Traditional Chinese Medicine Clinical Research Base Scientific Research Special Project, No. 2022JDZX098 and No. 2022JDZX114; National Natural Science Foundation of China, No. 82205086; and The 9th China Association for Science and Technology Young Talent Support Project, No. 2023QNRC001.
Conflict-of-interest statement: The authors declare that they have no competing interests.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Wen-Xia Zhao, MD, Chief Physician, Professor, Department of Spleen, Stomach, Liver and Gallbladder Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, No. 19 Renmin Road, Jinshui District, Zhengzhou 450000, Henan Province, China. zhao-wenxia@163.com
Received: October 15, 2024
Revised: January 4, 2025
Accepted: January 18, 2025
Published online: February 27, 2025
Processing time: 128 Days and 0.1 Hours

Abstract

Non-alcoholic fatty liver disease (NAFLD) is a progressive disease. Without effective interventions, NAFLD can gradually develop to non-alcoholic steatohepatitis, fatty liver fibrosis, liver cirrhosis and even hepatocellular carcinoma. It is still to investigate the precise molecular mechanism behind the pathophysiology of NAFLD. Triggering receptor expressed on myeloid cells 2 (TREM2) can sense tissue injury and mediate immune remodeling, thereby inducing phagocytosis, lipid metabolism, and metabolic transfer, promoting cell survival and combating inflammatory activation. NAFLD might develop as a result of TREM2's regulatory role. We here briefly summarize the biological characteristics of TREM2 and its functions in the disease progression of NAFLD. Moreover, we propose to broaden the therapeutic strategy for NAFLD by targeting TREM2.

Key Words: Triggering receptor expressed on myeloid cells 2; Non-alcoholic fatty liver disease; Macrophage; Lipid metabolism; Inflammation

Core Tip: Triggering receptor expressed on myeloid cells 2 (TREM2) is an immunoglobulin superfamily receptor located on the cell membrane. In the liver, it is mainly expressed on macrophages. TREM2 plays an important regulatory role in lipid metabolism, inflammation, fibrosis, and hepatocellular carcinoma, and is considered a potential therapeutic target for non-alcoholic fatty liver disease (NAFLD) (for example, macrophages with high expression of TREM2 can effectively clear apoptotic liver cells in a timely manner), thereby inhibiting chronic liver inflammation and non-alcoholic steatohepatitis lesions caused by obesity. This article highlights the most recent research developments on TREM2 in NAFLD, in an attempt to identify new pathways for NAFLD treatment.



INTRODUCTION

Non-alcoholic fatty liver disease (NAFLD) usually refers to hepatic steatosis that occurs without additional triggers (for example, medication, drinking alcohol, or certain genetic factors), symbolized by the formation of big fat droplets in liver cells due to an excess of triglyceride (TG)[1]. NAFLD patients are often accompanied by obesity, hyperlipidemia, diabetes mellitus type 2, as well as other metabolic disorders, which makes its prevention and treatment more complex[2]. According to current statistics, 30% of people worldwide have NAFLD, and the number is still rising[3]. In the upcoming decades, NAFLD may even emerge as the primary cause of end-stage liver disease, making it one of the most prevalent liver illnesses[4]. The range of NAFLD manifestations encompasses NAFLD, non-alcoholic steatohepatitis (NASH), as well as the cirrhosis and hepatocellular carcinoma (HCC) that can result from them[5]. NAFLD is histologically characterized by the presence of hepatic steatosis accounting for ≥ 5% of the liver, in the absence of any signs of hepatocyte injury. NASH is a more severe form of NAFLD, characterized by the presence of at least 5% hepatic steatosis along with inflammation accompanied by hepatocyte injury, including inflammatory reaction and ballooning degeneration, whether or not fibrosis is present. Over time, NASH has the potential to progress to cirrhosis and even cancer of the liver. In addition, NASH raises the risk of diabetes and cardiovascular disease[6]. The precise molecular mechanisms underlying the pathophysiology of NAFLD have not yet been thoroughly investigated, despite the fact that several fundamental and clinical investigations have been carried out. Resmetirom, the first medication in the world to treat adult NASH patients, is currently authorized for sale by the Food and Drug Administration[7]. Although encouraging, the results of the latest phase 3 clinical trial (MAESTRO-NASH) in patients with F1-F3 fibrosis and NASH have raised important questions about the comprehensive risks and advantages of Resmetirom[8,9]. Considering the need for long-term treatment, it is necessary to balance the effectiveness, safety, tolerability, and cost of drugs. For the diagnosis of NAFLD, as of right now, no particular biomarker exists for evaluating the progression and outcome of NAFLD, except for invasive liver biopsy. Therefore, the current understanding of NAFLD remains insufficient.

The immunoglobulin superfamily triggering receptor expressed on myeloid cells (TREM) 2 is found on the cell membrane, which undergoes a single transmembrane transition. The TREM2 gene was initially discovered by Swiss scientists Axel and Jess during cDNA cloning of dendritic cells (DCs) derived from mouse macrophages and monocytes[10]. TREM2 is expressed in osteoclasts and microglia under physiological settings and is crucial for tissue growth and functional preservation[11]. Zhou et al[12] reported the TREM2 gene is also expressed in certain macrophages of human adipose tissue, adrenal gland, and placental tissue, according to single-cell RNA transcriptome sequencing, but its role still needs further confirmation. In a diseased state, TREM2, as a membrane receptor, can perceive tissue damage and mediate immune remodeling, inducing phagocytosis, lipid metabolism, and metabolic transfer, promoting cell survival, and suppressing inflammatory activation[13]. Currently, TREM2 has been linked to both the onset and progression of Alzheimer's disease (AD) and other potential neurodegenerative disorders, obesity related to metabolic syndrome, NAFLD, atherosclerosis, tumors and other diseases. In the past few years, the biological function of TREM2 in liver associated illnesses has gradually been demonstrated. Non-parenchymal cells (NPCs) in the liver, including hepatic stellate cells (HSCs), liver sinusoidal endothelial cells, immature monocyte derived DCs, and Kupffer cells (KCs), express TREM2[14-17]. Numerous studies have demonstrated that TREM2 is a possible therapeutic target for NAFLD and regulates lipid metabolism, inflammation, fibrosis, and HCC. This article reviews the biological characteristics of TREM2 and the role it plays in NAFLD development. It is suggested that treatment strategies should be expanded for NAFLD by targeting TREM2.

STRUCTURE AND FUNCTION OF TREM2

The human TREM2 gene has five exons and is found on chromosome 6's broken arm 21.1 (Figure 1)[18]. The five axons on the TREM2 gene encode 693 base pairs[19]. Structurally, TREM2 consists of a signal peptide, a V-type immunoglobulin domain, an extracellular domain with a short stalk sequence, a transmembrane helix, and a cytoplasmic tail[20]. Among them, V-type immunoglobulin domain is used to identify the ligand of TREM2, and extracellular domain with a short stalk sequence is used to connect V-type immunoglobulin domain and transmembrane helix[21,22]. The cytoplasmic tail receives external signals from the transmembrane helix, which is anchored on the cell membrane[23,24].

Figure 1
Figure 1 Structure and function of triggering receptor expressed on myeloid cells 2. A: Triggering receptor expressed on myeloid cells (TREM) 2 consists of a signal peptide, an extracellular domain containing a V-type immunoglobulin domain, a short stalk sequence, a transmembrane helix, and a cytoplasmic tail; B: After binding to TREM2, ligands such as high-density lipoprotein, low-density lipoproteins, lipopolysaccharide, apolipoprotein, and other ligands bind to adaptor proteins DNAX-activating proteins (DAP) 12 and DAP10 to form a ligand-receptor complex, which triggers the phosphorylation of the immune receptor tyrosine-based activation motif in the cytoplasmic region of DAP12, recruiting and mediating spleen tyrosine kinase (Syk) phosphorylation, thereby activating downstream signaling pathways such as phosphatidylinositol 3-kinase, mitogen-activated protein kinase, extracellular regulating kinase, and c-Jun N-terminal kinase signaling pathways; C: During the early formation of the TREM2-DAP12 complex, the recruitment of Syk to DAP12 and the activation of downstream signaling can be blocked by Src homology 2-containing inositol-5'-phosphatase 1; D: TREM2 can also be cleaved by a disintegrin and metalloproteinase (ADAM) 10, ADAM17, and γ-secretase to release soluble TREM2. ADAM: A disintegrin and metalloproteinase; APOE: Apolipoprotein E; DAP: DNAX-activating proteins; ERK: Extracellular regulating kinase; HDL: High-density lipoprotein; ITAM: Immune receptor tyrosine-based activation motif; JNK: C-Jun N-terminal kinase; LDL: Low-density lipoproteins; LPS: Lipopolysaccharide; MAPK: Mitogen-activated protein kinase; PI3K: Phosphatidylinositol 3-kinase; STREM2: Soluble triggering receptor expressed on myeloid cells 2; Syk: Spleen tyrosine kinase; TREM2: Triggering receptor expressed on myeloid cells 2.

Transmembrane receptor TREM2 belongs to the immunoglobulin superfamily that requires binding to extracellular ligands to exert its physiological functions. The ligands of TREM2 include various free and bound anionic molecules on the plasma membrane and several apolipoproteins[25]. Through the positively charged residues in the transmembrane domain, TREM2 binds to ligands to form a receptor signaling complex that attaches to adaptor proteins: Signal transmission is mediated by DNAX-activating proteins (DAP) 12 and DAP10[26]. DAP12, often referred to as TYRO protein tyrosine kinase binding protein, is a signal transduction adaptor protein produced in cells involved in innate immunity responses. It possesses an immune receptor tyrosine-based activation motif (ITAM) structure in the cytosolic region[27]. Upon binding to ligands to form a ligand-receptor complex, TREM2 triggers phosphorylation of the ITAM in the cytosolic region of DAP12, recruiting and mediating phosphorylation of spleen tyrosine kinase (Syk), thereby downstream signaling pathways such phosphatidylinositol 3-kinase (PI3K), mitogen-activated protein kinase (MAPK), extracellular regulating kinase (ERK), and c-Jun N-terminal kinase (JNK) signaling pathways are activated[28]. In this process, Src homology 2-containing inositol-5'-phosphatase 1 (SHIP1) plays a facilitating role in the recruitment of PI3K to the TREM2-DAP12 complex[29]. Syk expression has been discovered to be positively connected with the severity of the disease, causing hepatocyte damage and exacerbating liver inflammation and fibrosis. It has been seen in both parenchymal (hepatocytes) and non-parenchymal (HSCs and KCs) cells in the liver[30]. In the early stage of TREM2-DAP12 complex formation, recruitment of Syk to DAP12 and activation of downstream signaling can be blocked by SHIP1, which contains inositol-5'-phosphatase 1[16]. By directly attaching its Src homology domain to the membrane proximal SPYQEL sequence inside the ITAM structure of DAP12, SHIP1 transforms the activating signaling complex into an inhibitory signaling complex, preventing Syk from signaling downstream and having anti-inflammatory and anti-fibrotic properties[31]. Kim et al[32] have revealed that a disintegrin and metalloproteinase (ADAM) 10, ADAM17, as well as gamma secretase can also cleave TREM2, releasing soluble TREM2 (sTREM2). The sTREM2 has been found in biological samples from people with inflammatory neurological disorders, including AD and amyotrophic lateral sclerosis, in several kinds of studies and the degree of sTREM2 is strongly connected with the severity of the condition[33,34]. Previous studies have shown that ADAM17 and ADAM10 can cleave human TREM2 at the H157-S158 peptide bond, releasing sTREM[35-38]. Experimental studies have shown that the level of sTREM2 dynamically reflects the infiltration of TREM2 + macrophages in the liver and can be used as a circulating biomarker to track the progression of NAFLD[39]. Clinical studies have shown that plasma sTREM2 is a better biomarker than conventional laboratory indicators for identifying distinct phases of NAFLD[40]. It is recommended that plasma sTREM2 be used as a non-invasive diagnostic marker for NAFLD.

TREM2 AND LIVER LIPID METABOLISM

Liver is the primary organ of the lipid metabolism in the human body, and plays a significant part in the metabolic processes of lipids including digestion, absorption, decomposition, synthesis, and transportation. Abnormal accumulation of lipids in liver cells is a pathological characteristic of NAFLD, and TG is the primary type of lipid accumulation in the liver of NAFLD patients[41]. TG deposition in the liver can be attributed to increased production of new fat in the liver, enhanced esterification of free fatty acids (FFA) (glycerol and FFA form TG through esterification), and reduced output of TG from very low density lipoprotein[42]. It has been demonstrated that insulin resistance (IR) is linked to the process of fat accumulation in the liver[43]. IR not only leads to dysfunction in the synthesis and transport of TG in liver cells, but also promotes the mobilization of peripheral fat and increases the concentration of FFA in plasma[44]. Excessive FFA can increase the production of lipotoxic substances in liver cells, which in turn promotes the progression of NAFLD to NASH[45,46]. There is mounting evidence that TREM2 regulates lipid metabolism (Figure 2)[47,48].

Figure 2
Figure 2 Regulatory role of triggering receptor expressed on myeloid cells 2 in liver lipid metabolism. On the one hand, macrophages lacking triggering receptor expressed on myeloid cells (TREM) 2 lead to the accumulation of hypertrophic adipocytes, causing systemic hypercholesterolemia. On the other hand, macrophages lacking TREM2 release exosomes, leading to mitochondrial fragmentation in hepatocytes and fatty acid oxidation dysfunction, accelerating the progression of non-alcoholic fatty liver disease (NAFLD). The deficiency of TREM2 may aggravate insulin resistance (IR) by increasing serum ceramide, leading to hepatic steatosis. The amount of soluble TREM2 in serum and IR are positively correlated. The increase of TREM1 and the decrease of TREM2 are involved in the pathological process of NAFLD. FAO: fatty acid oxidation; IR: insulin resistance; STREM2: Soluble triggering receptor expressed on myeloid cells 2; TREM: Triggering receptor expressed on myeloid cells 2.

Under the influence of metabolic disorders, obesity, and IR, abnormal formation of TG in liver cells results in hepatic steatosis, which is the first hit in the development of NAFLD[49]. One study used a high-fat diet (HFD) to induce a rat NAFLD model. HFD-induced NAFLD model rats exhibited severe metabolic dysfunction and hepatic steatosis, as evidenced by elevated plasma TG and total cholesterol (TC) levels and decreased TREM2 protein expression, in contrast to rats fed normal diet[50]. Knocking out the TREM2 gene in mice can inhibit the recruitment of macrophages to hypertrophic adipocytes and their clearance from hypertrophic adipocytes, leading to the accumulation of hypertrophic adipocytes, causing systemic hypercholesterolemia and inflammation[51]. TREM1 is a transmembrane receptor belonging to the immunoglobulin superfamily, just to TREM2[52,53]. NAFLD is closely associated with both an increase in TREM1 and a decrease in TREM2, and the pathological process of NAFLD involves an imbalance between TREM1 and TREM2[54]. Quercetin is one of the important flavonoids in nature, with high biological activity in vivo, and has the effects of improving lipid metabolism, anti-inflammatory, and anti-tumor[55-58]. According to studies, quercetin can lower plasma TC and TG levels, regulate the protein expression of TREM1 and TREM2, and alleviate HFD-induced NAFLD model rat liver lipid droplet accumulation[50]. However, whether TREM2 has an inhibitory effect on TG synthesis has not been further investigated.

The final product of lipid hydrolysis in the body is fatty acids, which are mainly oxidized and decomposed in mitochondria. TREM2 can protect the structure of mitochondria and ensure the oxidative decomposition of fatty acids[13]. Hepatocyte mitochondria and TREM2-expressing liver macrophages were discovered to have a metabolic coordination using in vivo and in vitro models of liver lipid excess[59]. TREM2-deficient macrophages can release exosomes rich in miR-106b-5p, which blocks downstream mitochondrial fusion protein 2 target molecules, leading to mitochondrial fragmentation in hepatocytes, fatty acid metabolism dysfunction (fatty acid oxidation dysfunction), and accelerated initial progression of NAFLD[60]. On the other hand, overexpression of TREM2 in liver macrophages improves the structure and energy supply of hepatocyte mitochondria, increases lipid catabolism, reduces de novo fatty acid synthesis, and alleviates NAFLD[61,62]. This suggests that TREM2 may protect the structure and energy supply of mitochondria by inhibiting the secretion of macrophage exosomes, ensuring the smooth oxidation of fatty acids, prevent hepatocytes from accumulating too much fatty acids, participating in regulating lipid metabolism, and alleviating NAFLD.

As the initiating factor of the "first hit" in the pathogenesis of NAFLD, IR and the onset of NAFLD are tightly associated. IR is the term used to describe the increased secretion of excessive insulin to induce hyperinsulinemia in order to maintain blood glucose stability when insulin's effectiveness in promoting glucose uptake and utilization is reduced for a variety of reasons[63]. In the state of IR, high concentrations of insulin promote the massive synthesis and storage of fat, inhibit the decomposition and oxidation of fat, and deposit a significant liver fat, which may contribute to the development of NAFLD[64]. Therefore, IR can cause fat to build up in liver cells, which can result in NAFLD liver steatosis[65]. TREM2 expression was considerably lower in obese patients with IR than in obese patients without IR and non-obese patients with IR, according to a clinical investigation involving 27 patients[66]. Serum sTREM2 levels and IR were positively correlated, and waist circumference and fat mass index rose when serum sTREM2 levels rose, according to a survey of 2742 Japanese community members over 40[67]. Under 13 weeks of HFD conditions, TREM2 knockout mice showed more severe IR and liver steatosis compared with wild-type (WT) mice[68]. Further research found that the mechanism by which TREM2 deficiency aggravates IR may be related to the increase in serum ceramide[69]. Ceramide can cause impaired insulin sensitivity, and targeted inhibition of ceramide synthesis can restore insulin tolerance and reverse liver steatosis in TREM2 knockout mice[70,71]. These results imply that TREM2 might be crucial to the IR-induced metabolism of liver lipids.

TREM2 AND CHRONIC INFLAMMATION

Reactive oxygen species (ROS), free radicals, endoplasmic reticulum stress, and other tissue damage can all contribute to liver injury and impaired cell function when there is an excessive buildup of lipids in the liver. Liver tissue injury can also induce immune response, leading to significant accumulation of monocyte-derived macrophages and resident macrophages, increased numbers of neutrophils and natural killer cells, infiltration of DCs, activation of liver inflammatory pathways, hence the transition from benign fatty degeneration to NASH. Therefore, chronic inflammation is an important histological marker of NAFLD.

In WT and TREM2 mice, researchers created models of acute and chronic liver injury. For in vitro tests, they separated primary liver cells from both mouse genotypes[72]. When exposed to acute and recurrent carbon tetrachloride and acetaminophen poisoning, mice deficient in TREM2 show increased levels of liver damage and inflammation, with TREM2 deficiency significantly associated with deteriorating survival. Consistent with its role in inflammation-related injury, when inflammation levels are high, TREM2 can block the production of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and ROS, while stimulating the production of interferon-β and IL-10, and alleviating excessive immune injury in the liver[72]. According to in vitro research, TREM2 + macrophages can help reduce inflammation following liver injury by blocking the generation of pro-inflammatory reactions brought on by lipoproteins containing lipopolysaccharide[73]. Liver macrophages expressing TREM2 exhibit inhibition of toll like receptor (TLR) 4-driven ERK and MAPK phosphorylation activation, leading to pro-inflammatory responses, as well as lipid peroxidation and cell death caused by ROS[61]. In addition, TREM2 can also inhibit the production of TLR4 dependent monocyte chemoattractant protein-1 in HSCs[16]. In summary, TREM2 has anti-inflammatory properties in chronic liver inflammation. In a multicenter proteomics study, the researchers characterized 4730 circulating proteins in NAFLD 306 patients with histological characteristics, and performed transcriptome analysis of paired liver tissues[74]. In order to identify high-risk steatohepatitis, they ultimately developed a non-invasive diagnostic logistic regression analysis model that included four proteins (TREM2, ADAMTSL2, AKR1B10, CFHR4), body mass index, and type 2 diabetes status. As a result, TREM2 might be crucial in regulating the inflammatory response in NAFLD (Figure 3).

Figure 3
Figure 3 Regulatory role of triggering receptor expressed on myeloid cells 2 in non-alcoholic fatty liver disease-related inflammatory injury. The liver macrophages expressing Triggering receptor expressed on myeloid cells 2 (TREM2) exhibit inhibition of the pro-inflammatory response caused by toll like receptor (TLR) 4-driven extracellular regulating kinase and mitogen-activated protein kinase phosphorylation activation, as well as reactive oxygen species-mediated lipid peroxidation and hepatocyte death. High expression of TREM2 ensures that macrophages can effectively clear apoptotic hepatocytes through efferocytosis in a timely manner, thereby inhibiting the transformation of non-alcoholic fatty liver disease into non-alcoholic steatohepatitis (NASH). TREM2 can induce macrophage differentiation towards M2 macrophages and inhibit inflammatory response. Tumor necrosis factor and interleukin-1β can induce a disintegrin and metalloproteinase 17 to cleave TREM2, leading to an increase in soluble TREM2 levels and aggravation of inflammation in NASH. TREM2 can also inhibit the production of TLR4-dependent monocyte chemoattractant protein-1 in hepatic stellate cells. The TREM2 receptor expressed in dendritic cells inhibits nuclear factor-kappa B through binding with DNAX-activating proteins 12, thereby alleviating liver inflammation. ADAM: A disintegrin and metalloproteinase; DAP: DNAX-activating proteins; STREM2: Soluble triggering receptor expressed on myeloid cells 2; TREM: Triggering receptor expressed on myeloid cells 2.

Although it is unknown if macrophages are friendly or hostile, they do contribute to liver inflammation as NAFLD progresses. The 90% of all macrophages in the human body are liver macrophages, which are extremely diverse and mostly comprise resident and infiltrating macrophages[75]. KCs, or liver-resident macrophages, are frequently seen in the liver's sinusoids. They are already established in the liver tissue throughout embryonic development and are derived from certain progenitor cells that were derived from the yolk sac[76]. In healthy liver tissue, KCs, as the sentinel cells of the liver, dominate the liver macrophages[77]. KCs can timely clean up damaged/dead cells to reduce tissue inflammation microenvironment and maintain liver homeostasis[78]. The three primary types of infiltrating macrophages are splenic, peritoneal, and monocyte-derived macrophages. Of these, the majority of infiltrating macrophages in the liver are monocyte-derived macrophages, which contribute to pathological diseases[79]. When the liver is affected by external factors and develops lesions, activated KCs can recruit a large number of monocytes in the blood to the liver and differentiate into CD11b+F4/80+ macrophages, known as classically activated M1 macrophages, which secrete pro-inflammatory factors such as TNF-α, IL-1β, and IL-6, as well as ROS, accelerating liver tissue injury[80]. These macrophages have the capacity to release a multitude of chemokines simultaneously, such as C-C chemokine ligand (CCL) 2, CCL3, and CCL5, which recruit monocyte macrophages and T lymphocytes in the blood circulation into the liver to exert their innate immunity and initiate adaptive immune regulation[81]. On the other hand, another classic type of liver macrophages, the alternative activated M2 macrophages, are crucial for tissue healing and the inhibition of inflammatory reactions[82]. When stimulated by cytokines such as IL-4 and IL-13, macrophages can polarize towards M2 type, which secretes a large amount of inhibitory inflammatory factors such as transforming growth factor (TGF)-β, IL-10, IL-4, and IL-13, while promoting the activation of arginase to induce the apoptosis of pro-inflammatory M1 type macrophages, accelerating tissue repair and remodeling[83]. Therefore, macrophages may have a dual role in the onset and progression of NAFLD. On the one hand, they may accelerate the progress of NAFLD by releasing inflammatory factors, on the other hand, they may slow down the progression of NAFLD by engulfing dead cells through their phagocytic function. Previous studies have demonstrated that monocyte-derived macrophages infiltration is markedly elevated in NAFLD patients' livers and is implicated in the progression and reversal of NAFLD disease[84]. TREM2 is mostly expressed in liver macrophages generated from monocytes, although it is hardly expressed in liver resident macrophages[85]. NPCs taken from the livers of both healthy and NASH mice were subjected to single-cell RNA sequencing by researchers. A characteristic of both animal and human NASH that is linked to disease severity is the elevated expression of TREM2, which they discovered to be present in NASH-associated macrophages (NAMs)[86]. Research shows that TREM2 can induce macrophage differentiation into M2 macrophages, increase the expression of M2 cytokines, and inhibit the expression of M1 cytokine[87]. This may explain the mechanism of the anti-inflammatory effect of TREM2 + macrophages in NAFLD. A recent study found that TREM2 expressed by macrophages is a key factor in maintaining immune homeostasis in the liver to stop NASH from developing[88]. The study found that high expression of TREM2 ensures that macrophages can effectively clear apoptotic hepatocytes (also known as "efferocytosis") in a timely manner, thereby inhibiting obesity-related NASH lesions and chronic liver inflammation. Defects in TREM2-dependent phagocytosis can lead to chronic liver inflammation and ultimately promote the transformation of NAFLD to NASH[88]. Fredrickson et al[89] used spatial transcriptomics to confirm that vertical sleeve gastrectomy (VSG) improved the metabolic status in the hepatic macrophage microenvironment in mouse and human MASH, and they further revealed by animal tests that TREM2 + macrophages directly mediated the benefits of VSG on MASH by attenuating hepatic inflammation and boosting efferocytosis of lipid-laden apoptotic hepatocytes, which directly mediated the mitigating effect of VSG on MASH. Cao et al[90] used bioinformatics and machine learning algorithms to screen five potential biomarkers for the diagnosis of NASH, including TREM2, which was subsequently validated by single-cell transcriptome and animal experiments to find that TREM2 was up-regulated in NASH, highlighting the great potential of TREM2 in the diagnosis of NASH. Further research found that in NASH mice, TNF and IL-1β caused the activity and expression of ADAM17, which played a role in splicing TREM2, leading in a notable surge in the level of TREM2 hydrolytic product sTREM2[91]. The increase in sTREM2 level means that TREM2 is hydrolyzed extensively, TREM2 cannot effectively exert anti-inflammatory effects, and NASH-related inflammatory reactions are exacerbated, revealing an important mechanism for the reduction of TREM2 in the progression of NAFLD. The hydrolytic product sTREM2 can be used as a non-invasive diagnostic biomarker to accurately exclude or diagnose NASH, reducing liver biopsy. A cross-sectional clinical study confirmed that plasma sTREM2 had good accuracy (area under the receiver operating characteristic curve = 0.92; 95%CI: 0.84-0.99, P < 0.01) for the diagnosis of NASH, with a sensitivity of 100% (95%CI: 0.76-1.0) and specificity of 86% (95%CI: 0.71-0.94)[39]. To further investigate the exclusion and diagnostic threshold of plasma sTREM2 for NASH, researchers collected 218 patients from three hospitals, including 48 patients in experimental group (9 patients with NASH, 18 patients with NAFLD, 11 obese patients without NAFLD, and 10 healthy controls), and 170 patients in validation group (all patients were confirmed to have increased liver stiffness by liver biopsy). The study ultimately determined that plasma sTREM2 levels ≤ 38 ng/mL were the optimal threshold for excluding NASH (sensitivity 90% and specificity 52%), and plasma sTREM2 levels ≥ 65 ng/mL were the optimal threshold for diagnosing NASH (specificity 89% and sensitivity 54%). As a non-invasive diagnostic biomarker, plasma sTREM2 can accurately exclude or diagnose NASH, reducing liver biopsy. Blocking macrophage TREM2 cleavage to restore its ability to clear apoptotic liver cells may be an effective method for preventing and treating NAFLD.

Currently acknowledged as the most efficient antigen presenting cells, DCs contribute significantly to the body's innate immune response, initiate adaptive immune responses, shape the type of immune response, and aid in the exertion of immune effects[92,93]. Signals in the liver microenvironment and the lipid content of DC cells can cause them to change from a tolerogenic to an immunogenic phenotype[94]. It is still unclear how DCs participate in the inflammatory process of NAFLD, as conflicting results have emerged from limited studies. Henning et al[95] have shown that DCs may minimize the inflammatory reaction of NASH by removing apoptotic bodies and necrotic material from the liver. According to certain research, liver DCs contribute to the development of illness in models of thioacetamide-induced liver fibrosis or major cellular (MCD)-induced NASH[96,97]. The immune regulatory ability of DCs is related to pattern recognition and immune regulation of DC surface receptors. DCs' expression of the TREM2 receptor contributes to reducing inflammatory reactions in the body by specifically pairing with DAP12 Ligands[98,99]. Research shows that in DCs with acute liver injury, TREM2 inhibits nuclear factor-kappa B (NF-κB) by binding with DAP12, thereby reducing liver inflammation[100].

TREM2 AND LIVER FIBROSIS

The primary cells in the liver that produce collagen are HSCs, which are quiescent, non-proliferative peri-sinusoidal cells that are situated between hepatocytes and sinusoidal endothelial cells in a healthy liver[87]. In the state of chronic liver injury, HSCs are in a sustained activated state, which disrupts the balance between the dissolution and deposition of extracellular matrix (ECM) in liver cells, resulting in progressive liver fibrosis. Liver fibrosis is one of the main factors contributing to the higher death rate from NAFLD[101-103]. If not intervened in time, liver fibrosis will eventually develop into liver cirrhosis and even HCC, and a series of serious issues like portal hypertension and liver failure, hepatic encephalopathy, etc. will occur, which seriously threatens the health of patients[104,105]. As an important immune cell in the liver, macrophages can regulate NAFLD-related liver fibrosis[106]. The study found that eliminating liver macrophages in mice might prevent liver fibrosis from developing in the early stages of fibrosis formation, while in the recovery stage of fibrosis, it increased the deposition of intrahepatic fibrosis and prevented the regression of liver fibrosis[107]. Why do these different effects occur? The reason is that in different stages of liver fibrosis, different subtypes of liver macrophages affect the appearance and regression of liver fibrosis pathology through different mechanisms. HSCs are the primary mediators of liver fibrosis, while the conversion of quiescent HSCs into myofibroblasts is the core of the pathogenesis of liver fibrosis. Macrophages regulate the activation of HSCs in a crucial way[108,109]. During the initial and progressive stages of NASH progression to liver fibrosis, injury-related pattern molecules and apoptotic bodies activate macrophages in the Disse space, causing their phenotype to shift toward M1 macrophages[110]. Activated macrophages can produce pro-fibrotic factors which include TGF-β and platelet-derived growth factor (PDGF), stimulate HSC, and promote their transformation into myofibroblasts, thus boosting the occurrence of liver fibrosis[111]. In addition, macrophages may result in inflammatory elements including TNFγ and IL-1β, which recruit extrahepatic inflammatory cells into the liver, further aggravating liver cell injury[112]. At the same time, inflammatory factors can activate HSCs via the pathway of NF-κB, maintain HSCs activity, and promote the development of liver fibrosis[113]. During the regression of liver fibrosis, the phenotype of liver macrophages is dominated by M2 macrophages, and they may have two ways of promoting anti-fibrosis: (1) Macrophages recruit and activate monocyte-derived macrophages and natural killer cells, leading to the apoptosis of activated HSCs, thereby exerting an anti-fibrotic effect[114]; and (2) Macrophages can phagocytize damaged hepatocytes, reduce inflammation, thus slowing the development of liver fibrosis[115].

The role of TREM2 in NAFLD-related liver fibrosis is unclear, as different studies have provided conflicting experimental results (Figure 4). According to research, male C57Bl6/J mice were given bone marrow from their littermates that were TREM2+/+ (TREM2+/+-tp, grey) and TREM2-/- (TREM2-/--tp, green) after being lethally irradiated. When mice were put on an MCD diet for four weeks following six weeks of recuperation, TREM2-/--bone marrow-chimeric animals displayed more pronounced fibrosis, indicating that TREM2 on macrophages has a protective function in NAFLD liver fibrosis. Further research showed that TREM2 could promote the reduction of mitochondrial membrane potential (MMP) 12 and increase the expression of tissue inhibitors of metalloproteinase 1, thereby exerting an anti-fibrotic effect[91]. Additionally, in vitro studies have demonstrated that TREM2 Loss can result in abnormalities in ECM remodeling and lipid processing, which can cause steatohepatitis and cell death, exacerbate fibrosis[91]. In NAFLD, plasma sTREM2 levels can be utilized as a biomarker to track the development of liver fibrosis. During the stages of liver fibrosis F0-F2, plasma sTREM2 levels increase proportionally with fibrosis stage, but no gradual increase was observed during the stages of fibrosis F3 and F4, indicating that plasma sTREM2 has a low ability to differentiate advanced liver fibrosis[40]. The increase in plasma sTREM2 means that TREM2 is cleaved. Does this imply that in the early phases of the development of liver fibrosis, TREM2 is cleaved extensively during the transition of macrophages to M1 macrophages, which in turn leads to the promotion of liver fibrosis by macrophages? In the late stages of liver fibrosis, the gradual increase in plasma sTREM2 does not mean that TREM2 cleavage is inhibited during the transition of macrophages to M2 macrophages, which in turn plays a role in inhibiting liver fibrosis?

Figure 4
Figure 4 Bidirectional regulation of triggering receptor expressed on myeloid cells 2 in non-alcoholic fatty liver disease-related liver fibrosis. Triggering receptor expressed on myeloid cells 2 (TREM2) can promote the reduction of mitochondrial membrane potential (MMP) 12 and increase the expression of tissue inhibitors of metalloproteinase 1, thereby exerting an anti-fibrotic effect. The level of plasma soluble TREM2 can be used as a biomarker to monitor the progression of liver fibrosis in non-alcoholic fatty liver disease. Conversely, studies have shown that CD9+TREM2+ non-alcoholic steatohepatitis-associated macrophages or scar-associated macrophages accumulate in the liver and promote liver fibrosis by enhancing the expression of MMP12, MMP13, and MMP14. MMP: Mitochondrial membrane potential; SAM: S-adenosyl methionine; STREM2: Soluble triggering receptor expressed on myeloid cells 2; TIMP1: Tissue inhibitors of metalloproteinase 1.

Other studies have revealed the role of TREM2 in promoting the formation of liver fibrosis. The researchers identified a TREM2+CD9+ macrophage subset that shares characteristics of both monocytes and KCs. They called this cell type CD9+TREM2+ NAMs or scar-associated macrophages. This cell type is amplified in fibrotic livers in humans and has a fibrogenic role[116]. TREM2 + macrophages build up in the liver as diet-induced NASH and drug-induced cirrhosis advance, and TREM2 can exacerbate liver fibrosis by upregulating the production of MMP12, MMP13, and MMP14[117]. Triiodothyronine (T3) can inhibit pro-fibrotic TREM2+CD9+ macrophages and participate in the inhibition of liver fibrosis progression[118]. A traditional Chinese medicine formula consisting of five herbs including Scutellaria, Sophora flavescens, Saposhnikovia divaricata, Citrus reticulata, and Panax quinquefolium can effectively inhibit the expansion of TREM2+CD9+ macrophage subsets in damaged livers, and remold the fibrotic niche by regulating ligand-receptor interactions (including TGF-β/epidermal growth factor receptor, PDGF-β/PDGFRα, and TNF superfamily 12/TNF receptor superfamily 12A signaling) to inhibit the progression of liver fibrosis[119]. Interestingly, in the experimental model of HCC in a fibrogenic environment, TREM2 promoted the incidence of fibrosis in liver, but interfered with the occurrence of liver tumors[17].

TREM2 AND HCC

Since the first report of the correlation between NAFLD and HCC in 1990[120], the incidence of NAFLD-related HCC has shown a significant upward trend, and in patients with NASH, HCC can occur directly without going through the cirrhosis stage[121-123]. Among NASH patients with cirrhosis, the annual incidence of HCC is 0.5%-2.6%, while the annual incidence of HCC in patients with non-cirrhosis NAFLD or NASH is 0.01%-0.13%[124]. The incidence of HCC is higher in patients with higher NASH degree or higher fibrosis grade[125]. In the presence of NAFLD/NASH, due to the effects of metabolism, oxidative stress, inflammation, immune response, and fibrosis, sustained destruction and compensatory proliferation of hepatocytes create an environment conducive to carcinogenesis[126]. Therefore, it is critical to treat NAFLD/NASH by comprehending the process by which it progresses to liver cancer.

In the acute diethylnitrosamine (DEN) model, TREM2 knockout mice display hepatocyte injury, inflammation, elevated oxidative stress, and increased liver tumor number. Studies conducted in vitro have demonstrated that the formation of human HCC spheroids was inhibited by conditioned media derived from human HSCs overexpressing TREM2[17], suggesting that TREM2 can protect the liver from HCC. Similarly, the researchers observed that TREM2 could also inhibit the growth of liver tumors in fibrosis-related HCC. The researchers observed in HSCs that overexpression of TREM2 could regulate the secretion of Wnt ligands and reduced the tumorigenicity of HCC. Another study showed that overexpression of TREM2 inhibited the progression of HCC by regulating the PI3K/protein kinase B (AKT)/β-catenin pathway[127]. In addition, during the development of advanced HCC, TREM2 in KCs and tumor-associated macrophages (TAMs) may counteract the initiation of liver tumors in HSCs by preventing TLR1 receptor-dependent responses[128]. According to the aforementioned studies, TREM2 protects against liver cancer development via a variety of mechanisms (Figure 5).

Figure 5
Figure 5 Bidirectional regulatory role of triggering receptor expressed on myeloid cells 2 in Non-alcoholic fatty liver disease-related hepatocellular carcinoma. Overexpressed triggering receptor expressed on myeloid cells 2 (TREM2) in hepatic stellate cells can regulate the secretion of Wnt ligands and protect the liver from hepatocellular carcinoma (HCC). Overexpressed TREM2 in HCC cells can inhibit HCC progression by regulating the phosphatidylinositol 3-kinase/protein kinase B/β-catenin pathway. During the development of end-stage HCC, TREM2 in Kupffer cells and tumor-associated macrophages may inhibit the occurrence of liver tumors by preventing toll like receptor-1 receptor-dependent responses. Another study revealed that TREM2 is a carcinogenic gene in the development of HCC. AKT: Protein kinase B; GSK3β: Glycogen synthase kinase-3β; HCC: Hepatocellular carcinoma; PI3K: Phosphatidylinositol 3-kinase; TAMs: Tumor-associated macrophages; TLR: Toll like receptor; TREM2: Triggering receptor expressed on myeloid cells 2.

However, some studies have revealed that TREM2 is a carcinogenic gene during the development of HCC[129]. The researchers used in situ injection of Hepa1-6 cells and spontaneous HCC based on NASH to construct two HCC models. In both HCC models, tumor growth in TREM2-WT mice was progressive, while tumor growth in TREM2-KO mice was considerably diminished. Knocking down TREM2 reconstitutes TAMs into an immune stimulatory state and enhances programmed death-1 immune checkpoint blockade's therapeutic impact in HCC[130]. Echinacoside (ECH), a phenylacetyl glucoside extracted from the medicinal herb Cistanche deserticola, significantly reduces the TREM2 protein levels in HepG2 cells and DEN-induced hepatoma cells[131]. Moreover, overexpression of TREM2 significantly reduced ECH-mediated proliferation inhibition and inactivation of the AKT signaling pathway, indicating that ECH exerts its anti-tumor activity by reducing TREM2 expression and the PI3K/AKT signaling pathway.

INTERACTION OF TREM2 WITH OTHER MOLECULAR PATHWAYS
In NAFLD

Ganguly et al[132] showed by bioinformatics analysis that in NASH-related macrophages, pathways like phagocytosis, anti-fibrotic, and enhanced lipid/lipoprotein handling were enriched in genes positively associated with elevated TREM2 expression. Among the lipid and lipoprotein handling-related pathways are involved in lipid metabolism, plasma lipoprotein assembly, remodeling, and clearance, glycosphingolipid metabolism, sphingolipid metabolism, low-density lipoproteins clearance, and the metabolism of fat-soluble vitamins. Phagocytosis pathway is involved in lysosomal vesicle biogenesis and production of ROS and reactive nitrogen species in phagocytes. The anti-fibrotic pathway involves the degradation of collagen and ECM, down-regulation of TGF-β signaling, and down-regulation of SMAD 3/4 activity. In addition, low TREM2 expression in NAMs was concentrated in metapathways associated with cell death, increased cytokine signaling, and inflammation. Among them, the inflammatory pathway involves TLR4 signaling, TLR9 signaling, co-stimulation of CD28 molecules, NF-κB activation, C-type lectin receptor, ERK signaling, and nucleotide-binding oligomerization domain (NOD) 1 and NOD 2 signaling. Enhanced cytokine signaling involves IL-1 signaling, IL signaling, TNF signaling pathways, and cytokine signaling in the immune system. Cell death pathways involve pyroptosis, apoptosis, and regulated necrosis. Yu et al[133] demonstrated that TREM2 deficiency increased inflammation and hepatic steatosis in mouse models. Through additional research, they have confirmed that TREM2 controls macrophage pyroptosis via the PI3K/AKT signaling pathway and is necessary for the macrophages' anti-inflammatory phenotypic transition. Furthermore, they have found that TREM2 additionally regulates the inflammatory reaction by affecting the expression of TGF-β1. These results imply that TREM2 is able to prevent or delay NAFLD progression by acting in correlation with other molecular pathways (Figure 6).

Figure 6
Figure 6 Interaction of triggering receptor expressed on myeloid cells 2 with other molecular pathways in non-alcoholic fatty liver disease. Triggering receptor expressed on myeloid cells 2 (TREM2) high-expressing macrophages have phagocytosic and anti-fibrotic functions, and improve lipid/lipoprotein processing. TREM2 Low-expressing macrophages are enriched in metapathways associated with inflammation, enhanced cytokine signaling, and cell death. AKT: Protein kinase B; ERK: Extracellular regulating kinase; IL: Interleukin; NF-κB: Nuclear factor-kappa B; NOD: Nucleotide-binding oligomerization domain; PI3K: Phosphatidylinositol 3-kinase; ROS: Reactive oxygen species; TGF: Transforming growth factor; TLR: Toll like receptor; TNF: Tumor necrosis factor.
CONCLUSION

Although previous studies have focused on elucidating the role of TREM2 in neurodegenerative diseases such as AD, researchers have begun to pay attention to its possible contribution to NAFLD.

In summary, TREM2 is involved in every facet of the pathophysiology of NAFLD. Currently, the mainstream view is that TREM2 has a protective effect on liver lipid metabolism, inflammation, liver fibrosis, and HCC. Nevertheless, we should also note that there is still some controversy over whether TREM2 plays a protective or harmful role in different animal models of liver fibrosis and HCC. Therefore, further basic research on the TREM2 pathway in liver fibrosis and HCC models during the natural course of NAFLD is needed. Due to its important regulatory role in disease, research on targeted TREM2 treatment has become a hot topic. At present, monoclonal antibodies against TREM2 that target the microglial inflammatory response have been applied in clinical phase I trials in AD patients[134,135], but potential therapeutic strategies for NAFLD are still being explored. In addition, TREM2 may benefit NAFLD progression through interactions with other molecular pathways. However, these summaries were mainly from correlation analyses through bioinformatics and were rarely validated experimentally. The mechanism by which TREM2 interacts with other molecular pathways to participate in the remission of NAFLD progression need to be explored through more studies in the future. Although some research teams have validated the possibility of sTREM2 as a diagnostic biomarker for NAFLD/NASH in the clinic, there is a lack of validation with multicenter and large-sample clinical data. In the future, there is a need to develop easy-to-operate and lower-cost sTREM2 detection kits so that they can be widely applied in clinical practice.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A, Grade C

Novelty: Grade A, Grade B

Creativity or Innovation: Grade B, Grade B

Scientific Significance: Grade B, Grade C

P-Reviewer: Liu H; Zhang P S-Editor: Luo ML L-Editor: A P-Editor: Zhao YQ

References
1.  Powell EE, Wong VW, Rinella M. Non-alcoholic fatty liver disease. Lancet. 2021;397:2212-2224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 461]  [Cited by in RCA: 1405]  [Article Influence: 351.3]  [Reference Citation Analysis (33)]
2.  Bugianesi E, Petta S. NAFLD/NASH. J Hepatol. 2022;77:549-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
3.  Younossi ZM, Golabi P, Paik JM, Henry A, Van Dongen C, Henry L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): a systematic review. Hepatology. 2023;77:1335-1347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in RCA: 935]  [Article Influence: 467.5]  [Reference Citation Analysis (2)]
4.  Cusi K, Isaacs S, Barb D, Basu R, Caprio S, Garvey WT, Kashyap S, Mechanick JI, Mouzaki M, Nadolsky K, Rinella ME, Vos MB, Younossi Z. American Association of Clinical Endocrinology Clinical Practice Guideline for the Diagnosis and Management of Nonalcoholic Fatty Liver Disease in Primary Care and Endocrinology Clinical Settings: Co-Sponsored by the American Association for the Study of Liver Diseases (AASLD). Endocr Pract. 2022;28:528-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in RCA: 482]  [Article Influence: 160.7]  [Reference Citation Analysis (1)]
5.  Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, Harrison SA, Brunt EM, Sanyal AJ. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology. 2018;67:328-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3544]  [Cited by in RCA: 4671]  [Article Influence: 667.3]  [Reference Citation Analysis (8)]
6.  Duell PB, Welty FK, Miller M, Chait A, Hammond G, Ahmad Z, Cohen DE, Horton JD, Pressman GS, Toth PP; American Heart Association Council on Arteriosclerosis, Thrombosis and Vascular Biology;  Council on Hypertension;  Council on the Kidney in Cardiovascular Disease;  Council on Lifestyle and Cardiometabolic Health;  and Council on Peripheral Vascular Disease. Nonalcoholic Fatty Liver Disease and Cardiovascular Risk: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol. 2022;42:e168-e185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in RCA: 279]  [Article Influence: 93.0]  [Reference Citation Analysis (0)]
7.  Kokkorakis M, Boutari C, Hill MA, Kotsis V, Loomba R, Sanyal AJ, Mantzoros CS. Resmetirom, the first approved drug for the management of metabolic dysfunction-associated steatohepatitis: Trials, opportunities, and challenges. Metabolism. 2024;154:155835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
8.  Harrison SA, Bedossa P, Guy CD, Schattenberg JM, Loomba R, Taub R, Labriola D, Moussa SE, Neff GW, Rinella ME, Anstee QM, Abdelmalek MF, Younossi Z, Baum SJ, Francque S, Charlton MR, Newsome PN, Lanthier N, Schiefke I, Mangia A, Pericàs JM, Patil R, Sanyal AJ, Noureddin M, Bansal MB, Alkhouri N, Castera L, Rudraraju M, Ratziu V; MAESTRO-NASH Investigators. A Phase 3, Randomized, Controlled Trial of Resmetirom in NASH with Liver Fibrosis. N Engl J Med. 2024;390:497-509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in RCA: 411]  [Article Influence: 411.0]  [Reference Citation Analysis (0)]
9.  The Lancet Gastroenterology Hepatology. Resmetirom for NASH: balancing promise and prudence. Lancet Gastroenterol Hepatol. 2024;9:273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
10.  Myers J, Kokkinos P, Nyelin E. Physical Activity, Cardiorespiratory Fitness, and the Metabolic Syndrome. Nutrients. 2019;11:1652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 325]  [Cited by in RCA: 304]  [Article Influence: 50.7]  [Reference Citation Analysis (0)]
11.  Qin Q, Teng Z, Liu C, Li Q, Yin Y, Tang Y. TREM2, microglia, and Alzheimer's disease. Mech Ageing Dev. 2021;195:111438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 94]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
12.  Zhou Y, Song WM, Andhey PS, Swain A, Levy T, Miller KR, Poliani PL, Cominelli M, Grover S, Gilfillan S, Cella M, Ulland TK, Zaitsev K, Miyashita A, Ikeuchi T, Sainouchi M, Kakita A, Bennett DA, Schneider JA, Nichols MR, Beausoleil SA, Ulrich JD, Holtzman DM, Artyomov MN, Colonna M. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer's disease. Nat Med. 2020;26:131-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 703]  [Cited by in RCA: 639]  [Article Influence: 127.8]  [Reference Citation Analysis (0)]
13.  Zhang K, Wang Y, Chen S, Mao J, Jin Y, Ye H, Zhang Y, Liu X, Gong C, Cheng X, Huang X, Hoeft A, Chen Q, Li X, Fang X. TREM2(hi) resident macrophages protect the septic heart by maintaining cardiomyocyte homeostasis. Nat Metab. 2023;5:129-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 64]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
14.  Gonçalves LA, Rodrigues-Duarte L, Rodo J, Vieira de Moraes L, Marques I, Penha-Gonçalves C. TREM2 governs Kupffer cell activation and explains belr1 genetic resistance to malaria liver stage infection. Proc Natl Acad Sci U S A. 2013;110:19531-19536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in RCA: 29]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
15.  Chen LC, Laskin JD, Gordon MK, Laskin DL. Regulation of TREM expression in hepatic macrophages and endothelial cells during acute endotoxemia. Exp Mol Pathol. 2008;84:145-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in RCA: 67]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
16.  Perugorria MJ, Esparza-Baquer A, Oakley F, Labiano I, Korosec A, Jais A, Mann J, Tiniakos D, Santos-Laso A, Arbelaiz A, Gawish R, Sampedro A, Fontanellas A, Hijona E, Jimenez-Agüero R, Esterbauer H, Stoiber D, Bujanda L, Banales JM, Knapp S, Sharif O, Mann DA. Non-parenchymal TREM-2 protects the liver from immune-mediated hepatocellular damage. Gut. 2019;68:533-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in RCA: 89]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
17.  Esparza-Baquer A, Labiano I, Sharif O, Agirre-Lizaso A, Oakley F, Rodrigues PM, Zhuravleva E, O'Rourke CJ, Hijona E, Jimenez-Agüero R, Riaño I, Landa A, La Casta A, Zaki MYW, Munoz-Garrido P, Azkargorta M, Elortza F, Vogel A, Schabbauer G, Aspichueta P, Andersen JB, Knapp S, Mann DA, Bujanda L, Banales JM, Perugorria MJ. TREM-2 defends the liver against hepatocellular carcinoma through multifactorial protective mechanisms. Gut. 2021;70:1345-1361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in RCA: 64]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
18.  Allcock RJ, Barrow AD, Forbes S, Beck S, Trowsdale J. The human TREM gene cluster at 6p21.1 encodes both activating and inhibitory single IgV domain receptors and includes NKp44. Eur J Immunol. 2003;33:567-577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in RCA: 122]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
19.  Ulland TK, Colonna M. TREM2 - a key player in microglial biology and Alzheimer disease. Nat Rev Neurol. 2018;14:667-675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in RCA: 247]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
20.  Huang W, Huang J, Huang N, Luo Y. The role of TREM2 in Alzheimer's disease: from the perspective of Tau. Front Cell Dev Biol. 2023;11:1280257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
21.  Molgora M, Liu YA, Colonna M, Cella M. TREM2: A new player in the tumor microenvironment. Semin Immunol. 2023;67:101739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
22.  Han J, Wang M, Ren M, Lou H. Contributions of triggering-receptor-expressed-on-myeloid-cells-2 to neurological diseases. Int J Neurosci. 2017;127:368-375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
23.  Kober DL, Alexander-Brett JM, Karch CM, Cruchaga C, Colonna M, Holtzman MJ, Brett TJ. Neurodegenerative disease mutations in TREM2 reveal a functional surface and distinct loss-of-function mechanisms. Elife. 2016;5:e20391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in RCA: 138]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
24.  Kawabori M, Kacimi R, Kauppinen T, Calosing C, Kim JY, Hsieh CL, Nakamura MC, Yenari MA. Triggering receptor expressed on myeloid cells 2 (TREM2) deficiency attenuates phagocytic activities of microglia and exacerbates ischemic damage in experimental stroke. J Neurosci. 2015;35:3384-3396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in RCA: 262]  [Article Influence: 26.2]  [Reference Citation Analysis (0)]
25.  Hakola HP. Neuropsychiatric and genetic aspects of a new hereditary disease characterized by progressive dementia and lipomembranous polycystic osteodysplasia. Acta Psychiatr Scand Suppl. 1972;232:1-173.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Schlepckow K, Morenas-Rodríguez E, Hong S, Haass C. Stimulation of TREM2 with agonistic antibodies-an emerging therapeutic option for Alzheimer's disease. Lancet Neurol. 2023;22:1048-1060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
27.  Konishi H, Kiyama H. Non-pathological roles of microglial TREM2/DAP12: TREM2/DAP12 regulates the physiological functions of microglia from development to aging. Neurochem Int. 2020;141:104878.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
28.  Deczkowska A, Weiner A, Amit I. The Physiology, Pathology, and Potential Therapeutic Applications of the TREM2 Signaling Pathway. Cell. 2020;181:1207-1217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in RCA: 143]  [Article Influence: 28.6]  [Reference Citation Analysis (1)]
29.  Magno L, Bunney TD, Mead E, Svensson F, Bictash MN. TREM2/PLCγ2 signalling in immune cells: function, structural insight, and potential therapeutic modulation. Mol Neurodegener. 2021;16:22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
30.  Kurniawan DW, Storm G, Prakash J, Bansal R. Role of spleen tyrosine kinase in liver diseases. World J Gastroenterol. 2020;26:1005-1019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 14]  [Cited by in RCA: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
31.  Kober DL, Brett TJ. TREM2-Ligand Interactions in Health and Disease. J Mol Biol. 2017;429:1607-1629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in RCA: 164]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
32.  Kim SH, Lee KY, Chang K. The Protective Role of TREM2 in the Heterogenous Population of Macrophages during Post-Myocardial Infarction Inflammation. Int J Mol Sci. 2023;24:5556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
33.  Paloneva J, Manninen T, Christman G, Hovanes K, Mandelin J, Adolfsson R, Bianchin M, Bird T, Miranda R, Salmaggi A, Tranebjaerg L, Konttinen Y, Peltonen L. Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am J Hum Genet. 2002;71:656-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 509]  [Cited by in RCA: 501]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
34.  Piccio L, Buonsanti C, Cella M, Tassi I, Schmidt RE, Fenoglio C, Rinker J 2nd, Naismith RT, Panina-Bordignon P, Passini N, Galimberti D, Scarpini E, Colonna M, Cross AH. Identification of soluble TREM-2 in the cerebrospinal fluid and its association with multiple sclerosis and CNS inflammation. Brain. 2008;131:3081-3091.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in RCA: 222]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
35.  Wunderlich P, Glebov K, Kemmerling N, Tien NT, Neumann H, Walter J. Sequential proteolytic processing of the triggering receptor expressed on myeloid cells-2 (TREM2) protein by ectodomain shedding and γ-secretase-dependent intramembranous cleavage. J Biol Chem. 2013;288:33027-33036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in RCA: 219]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
36.  Lichtenthaler SF, Lemberg MK, Fluhrer R. Proteolytic ectodomain shedding of membrane proteins in mammals-hardware, concepts, and recent developments. EMBO J. 2018;37:e99456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in RCA: 189]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
37.  Feuerbach D, Schindler P, Barske C, Joller S, Beng-Louka E, Worringer KA, Kommineni S, Kaykas A, Ho DJ, Ye C, Welzenbach K, Elain G, Klein L, Brzak I, Mir AK, Farady CJ, Aichholz R, Popp S, George N, Neumann U. ADAM17 is the main sheddase for the generation of human triggering receptor expressed in myeloid cells (hTREM2) ectodomain and cleaves TREM2 after Histidine 157. Neurosci Lett. 2017;660:109-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in RCA: 79]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
38.  Schlepckow K, Kleinberger G, Fukumori A, Feederle R, Lichtenthaler SF, Steiner H, Haass C. An Alzheimer-associated TREM2 variant occurs at the ADAM cleavage site and affects shedding and phagocytic function. EMBO Mol Med. 2017;9:1356-1365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in RCA: 161]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
39.  Kothari V, Savard C, Tang J, Lee SP, Subramanian S, Wang S, den Hartigh LJ, Bornfeldt KE, Ioannou GN. sTREM2 is a plasma biomarker for human NASH and promotes hepatocyte lipid accumulation. Hepatol Commun. 2023;7:e0265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
40.  Indira Chandran V, Wernberg CW, Lauridsen MM, Skytthe MK, Bendixen SM, Larsen FT, Hansen CD, Grønkjær LL, Siersbæk MS, Caterino TD, Detlefsen S, Møller HJ, Grøntved L, Ravnskjaer K, Moestrup SK, Thiele MS, Krag A, Graversen JH. Circulating TREM2 as a noninvasive diagnostic biomarker for NASH in patients with elevated liver stiffness. Hepatology. 2023;77:558-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
41.  McGlinchey AJ, Govaere O, Geng D, Ratziu V, Allison M, Bousier J, Petta S, de Oliviera C, Bugianesi E, Schattenberg JM, Daly AK, Hyötyläinen T, Anstee QM, Orešič M. Metabolic signatures across the full spectrum of non-alcoholic fatty liver disease. JHEP Rep. 2022;4:100477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 38]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
42.  Han SK, Baik SK, Kim MY. Non-alcoholic fatty liver disease: Definition and subtypes. Clin Mol Hepatol. 2023;29:S5-S16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in RCA: 73]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
43.  Tanase DM, Gosav EM, Costea CF, Ciocoiu M, Lacatusu CM, Maranduca MA, Ouatu A, Floria M. The Intricate Relationship between Type 2 Diabetes Mellitus (T2DM), Insulin Resistance (IR), and Nonalcoholic Fatty Liver Disease (NAFLD). J Diabetes Res. 2020;2020:3920196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in RCA: 261]  [Article Influence: 52.2]  [Reference Citation Analysis (0)]
44.  Pafili K, Roden M. Nonalcoholic fatty liver disease (NAFLD) from pathogenesis to treatment concepts in humans. Mol Metab. 2021;50:101122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in RCA: 164]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
45.  Branković M, Jovanović I, Dukić M, Radonjić T, Oprić S, Klašnja S, Zdravković M. Lipotoxicity as the Leading Cause of Non-Alcoholic Steatohepatitis. Int J Mol Sci. 2022;23:5146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
46.  Wasilewska N, Lebensztejn DM. Non-alcoholic fatty liver disease and lipotoxicity. Clin Exp Hepatol. 2021;7:1-6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
47.  Li RY, Qin Q, Yang HC, Wang YY, Mi YX, Yin YS, Wang M, Yu CJ, Tang Y. TREM2 in the pathogenesis of AD: a lipid metabolism regulator and potential metabolic therapeutic target. Mol Neurodegener. 2022;17:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 43]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
48.  Damisah EC, Rai A, Grutzendler J. TREM2: Modulator of Lipid Metabolism in Microglia. Neuron. 2020;105:759-761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
49.  Guo X, Yin X, Liu Z, Wang J. Non-Alcoholic Fatty Liver Disease (NAFLD) Pathogenesis and Natural Products for Prevention and Treatment. Int J Mol Sci. 2022;23:15489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 110]  [Reference Citation Analysis (1)]
50.  Gao XR, Chen Z, Fang K, Xu JX, Ge JF. Protective effect of quercetin against the metabolic dysfunction of glucose and lipids and its associated learning and memory impairments in NAFLD rats. Lipids Health Dis. 2021;20:164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
51.  Jaitin DA, Adlung L, Thaiss CA, Weiner A, Li B, Descamps H, Lundgren P, Bleriot C, Liu Z, Deczkowska A, Keren-Shaul H, David E, Zmora N, Eldar SM, Lubezky N, Shibolet O, Hill DA, Lazar MA, Colonna M, Ginhoux F, Shapiro H, Elinav E, Amit I. Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner. Cell. 2019;178:686-698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 582]  [Cited by in RCA: 751]  [Article Influence: 125.2]  [Reference Citation Analysis (0)]
52.  Siskind S, Brenner M, Wang P. TREM-1 Modulation Strategies for Sepsis. Front Immunol. 2022;13:907387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
53.  Lemarié J, Gibot S. Soluble Triggering Receptor Expressed on Myeloid Cells-1: Diagnosis or Prognosis? Crit Care Clin. 2020;36:41-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
54.  Dou ZF, Guo YR, Liu JC, Li N, Chai B, Li XQ, Fu R, Wang XJ. Ameliorative effects of glycine in an experimental nonalcoholic steatohepatitis and its correlation between TREM-1 and TREM-2. Am J Transl Res. 2016;8:284-297.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Hosseini A, Razavi BM, Banach M, Hosseinzadeh H. Quercetin and metabolic syndrome: A review. Phytother Res. 2021;35:5352-5364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 173]  [Article Influence: 43.3]  [Reference Citation Analysis (0)]
56.  Han X, Xu T, Fang Q, Zhang H, Yue L, Hu G, Sun L. Quercetin hinders microglial activation to alleviate neurotoxicity via the interplay between NLRP3 inflammasome and mitophagy. Redox Biol. 2021;44:102010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in RCA: 249]  [Article Influence: 62.3]  [Reference Citation Analysis (0)]
57.  Sethi G, Rath P, Chauhan A, Ranjan A, Choudhary R, Ramniwas S, Sak K, Aggarwal D, Rani I, Tuli HS. Apoptotic Mechanisms of Quercetin in Liver Cancer: Recent Trends and Advancements. Pharmaceutics. 2023;15:712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in RCA: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
58.  Reyes-Farias M, Carrasco-Pozo C. The Anti-Cancer Effect of Quercetin: Molecular Implications in Cancer Metabolism. Int J Mol Sci. 2019;20:3177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in RCA: 368]  [Article Influence: 61.3]  [Reference Citation Analysis (0)]
59.  Coelho I, Duarte N, Macedo MP, Penha-Gonçalves C. Insights into Macrophage/Monocyte-Endothelial Cell Crosstalk in the Liver: A Role for Trem-2. J Clin Med. 2021;10:1248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
60.  Yu T, Fu H, Sun JJ, Ding DR, Wang H. miR-106b-5p upregulation is associated with microglial activation and inflammation in the mouse hippocampus following status epilepticus. Exp Brain Res. 2021;239:3315-3325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
61.  Hou J, Zhang J, Cui P, Zhou Y, Liu C, Wu X, Ji Y, Wang S, Cheng B, Ye H, Shu L, Zhang K, Wang D, Xu J, Shu Q, Colonna M, Fang X. TREM2 sustains macrophage-hepatocyte metabolic coordination in nonalcoholic fatty liver disease and sepsis. J Clin Invest. 2021;131:e135197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in RCA: 120]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
62.  Ji PX, Chen YX, Ni XX, Miao Q, Hua J. Effect of triggering receptor expressed on myeloid cells 2-associated alterations on lipid metabolism in macrophages in the development of non-alcoholic fatty liver disease. J Gastroenterol Hepatol. 2024;39:369-380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
63.  Li M, Chi X, Wang Y, Setrerrahmane S, Xie W, Xu H. Trends in insulin resistance: insights into mechanisms and therapeutic strategy. Signal Transduct Target Ther. 2022;7:216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in RCA: 244]  [Article Influence: 81.3]  [Reference Citation Analysis (0)]
64.  Khan RS, Bril F, Cusi K, Newsome PN. Modulation of Insulin Resistance in Nonalcoholic Fatty Liver Disease. Hepatology. 2019;70:711-724.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in RCA: 266]  [Article Influence: 44.3]  [Reference Citation Analysis (0)]
65.  Kuchay MS, Choudhary NS, Mishra SK. Pathophysiological mechanisms underlying MAFLD. Diabetes Metab Syndr. 2020;14:1875-1887.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in RCA: 90]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
66.  Subramanian S, Pallati PK, Rai V, Sharma P, Agrawal DK, Nandipati KC. Increased Expression of Triggering Receptor Expressed on Myeloid Cells-1 in the Population with Obesity and Insulin Resistance. Obesity (Silver Spring). 2019;27:513-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
67.  Tanaka M, Honda T, Yamakage H, Hata J, Yoshida D, Hirakawa Y, Shibata M, Inoue T, Kusakabe T, Satoh-Asahara N, Ninomiya T. A potential novel pathological implication of serum soluble triggering receptor expressed on myeloid cell 2 in insulin resistance in a general Japanese population: The Hisayama study. Diabetes Res Clin Pract. 2018;146:225-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 12]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
68.  Liu C, Li P, Li H, Wang S, Ding L, Wang H, Ye H, Jin Y, Hou J, Fang X, Shu Q. TREM2 regulates obesity-induced insulin resistance via adipose tissue remodeling in mice of high-fat feeding. J Transl Med. 2019;17:300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 39]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
69.  Webster KL, Becker L. Surplus Ceramides: An Added Twist in the Tale of TREM2 and Insulin Resistance. Diabetes. 2021;70:1926-1928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
70.  Sharif O, Brunner JS, Korosec A, Martins R, Jais A, Snijder B, Vogel A, Caldera M, Hladik A, Lakovits K, Saluzzo S, Boehm B, Gorki AD, Mesteri I, Lindroos-Christensen J, Tillmann K, Stoiber D, Menche J, Schabbauer G, Bilban M, Superti-Furga G, Esterbauer H, Knapp S. Beneficial Metabolic Effects of TREM2 in Obesity Are Uncoupled From Its Expression on Macrophages. Diabetes. 2021;70:2042-2057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in RCA: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
71.  Chaurasia B, Tippetts TS, Mayoral Monibas R, Liu J, Li Y, Wang L, Wilkerson JL, Sweeney CR, Pereira RF, Sumida DH, Maschek JA, Cox JE, Kaddai V, Lancaster GI, Siddique MM, Poss A, Pearson M, Satapati S, Zhou H, McLaren DG, Previs SF, Chen Y, Qian Y, Petrov A, Wu M, Shen X, Yao J, Nunes CN, Howard AD, Wang L, Erion MD, Rutter J, Holland WL, Kelley DE, Summers SA. Targeting a ceramide double bond improves insulin resistance and hepatic steatosis. Science. 2019;365:386-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in RCA: 307]  [Article Influence: 51.2]  [Reference Citation Analysis (0)]
72.  Dabla A, Liang YC, Rajabalee N, Irwin C, Moonen CGJ, Willis JV, Berton S, Sun J. TREM2 Promotes Immune Evasion by Mycobacterium tuberculosis in Human Macrophages. mBio. 2022;13:e0145622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 18]  [Reference Citation Analysis (0)]
73.  Liebold I, Meyer S, Heine M, Kuhl A, Witt J, Eissing L, Fischer AW, Koop AC, Kluwe J, Wiesch JSZ, Wehmeyer M, Knippschild U, Scheja L, Heeren J, Bosurgi L, Worthmann A. TREM2 Regulates the Removal of Apoptotic Cells and Inflammatory Processes during the Progression of NAFLD. Cells. 2023;12:341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
74.  Govaere O, Hasoon M, Alexander L, Cockell S, Tiniakos D, Ekstedt M, Schattenberg JM, Boursier J, Bugianesi E, Ratziu V; LITMUS Investigators, Daly AK, Anstee QM. A proteo-transcriptomic map of non-alcoholic fatty liver disease signatures. Nat Metab. 2023;5:572-578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in RCA: 53]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
75.  Dou L, Shi X, He X, Gao Y. Macrophage Phenotype and Function in Liver Disorder. Front Immunol. 2019;10:3112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in RCA: 100]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
76.  Li W, Chang N, Li L. Heterogeneity and Function of Kupffer Cells in Liver Injury. Front Immunol. 2022;13:940867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 24]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
77.  Chen J, Deng X, Liu Y, Tan Q, Huang G, Che Q, Guo J, Su Z. Kupffer Cells in Non-alcoholic Fatty Liver Disease: Friend or Foe? Int J Biol Sci. 2020;16:2367-2378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in RCA: 60]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
78.  Li W, Yang Y, Yang L, Chang N, Li L. Monocyte-derived Kupffer cells dominate in the Kupffer cell pool during liver injury. Cell Rep. 2023;42:113164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
79.  Guilliams M, Scott CL. Liver macrophages in health and disease. Immunity. 2022;55:1515-1529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 143]  [Article Influence: 47.7]  [Reference Citation Analysis (0)]
80.  Yang Y, Sheng J, Sheng Y, Wang J, Zhou X, Li W, Kong Y. Lapachol treats non-alcoholic fatty liver disease by modulating the M1 polarization of Kupffer cells via PKM2. Int Immunopharmacol. 2023;120:110380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
81.  Wallace SJ, Tacke F, Schwabe RF, Henderson NC. Understanding the cellular interactome of non-alcoholic fatty liver disease. JHEP Rep. 2022;4:100524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in RCA: 45]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
82.  Videla LA, Valenzuela R, Del Campo A, Zúñiga-Hernández J. Omega-3 Lipid Mediators: Modulation of the M1/M2 Macrophage Phenotype and Its Protective Role in Chronic Liver Diseases. Int J Mol Sci. 2023;24:15528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
83.  Xu L, Chen Y, Nagashimada M, Ni Y, Zhuge F, Chen G, Li H, Pan T, Yamashita T, Mukaida N, Kaneko S, Ota T, Nagata N. CC chemokine ligand 3 deficiency ameliorates diet-induced steatohepatitis by regulating liver macrophage recruitment and M1/M2 status in mice. Metabolism. 2021;125:154914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in RCA: 34]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
84.  Barreby E, Chen P, Aouadi M. Macrophage functional diversity in NAFLD - more than inflammation. Nat Rev Endocrinol. 2022;18:461-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in RCA: 105]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
85.  Daemen S, Gainullina A, Kalugotla G, He L, Chan MM, Beals JW, Liss KH, Klein S, Feldstein AE, Finck BN, Artyomov MN, Schilling JD. Dynamic Shifts in the Composition of Resident and Recruited Macrophages Influence Tissue Remodeling in NASH. Cell Rep. 2021;34:108626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in RCA: 192]  [Article Influence: 48.0]  [Reference Citation Analysis (0)]
86.  Xiong X, Kuang H, Ansari S, Liu T, Gong J, Wang S, Zhao XY, Ji Y, Li C, Guo L, Zhou L, Chen Z, Leon-Mimila P, Chung MT, Kurabayashi K, Opp J, Campos-Pérez F, Villamil-Ramírez H, Canizales-Quinteros S, Lyons R, Lumeng CN, Zhou B, Qi L, Huertas-Vazquez A, Lusis AJ, Xu XZS, Li S, Yu Y, Li JZ, Lin JD. Landscape of Intercellular Crosstalk in Healthy and NASH Liver Revealed by Single-Cell Secretome Gene Analysis. Mol Cell. 2019;75:644-660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in RCA: 486]  [Article Influence: 97.2]  [Reference Citation Analysis (0)]
87.  Zhu D, Huang M, Shen P, Zhang B, Chen G, Chen J, Duan L, Duan Y. TREM2 expression promotes liver and peritoneal M2 macrophage polarization in mice infected with Schistosoma japonicum. J Cell Mol Med. 2023;27:2261-2269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
88.  Wang X, He Q, Zhou C, Xu Y, Liu D, Fujiwara N, Kubota N, Click A, Henderson P, Vancil J, Marquez CA, Gunasekaran G, Schwartz ME, Tabrizian P, Sarpel U, Fiel MI, Diao Y, Sun B, Hoshida Y, Liang S, Zhong Z. Prolonged hypernutrition impairs TREM2-dependent efferocytosis to license chronic liver inflammation and NASH development. Immunity. 2023;56:58-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in RCA: 75]  [Article Influence: 37.5]  [Reference Citation Analysis (0)]
89.  Fredrickson G, Florczak K, Barrow F, Mahmud S, Dietsche K, Wang H, Parthiban P, Hakeem A, Almutlaq R, Adeyi O, Herman A, Bartolomucci A, Staley C, Dong X, Jahansouz C, Williams JW, Mashek DG, Ikramuddin S, Revelo XS. TREM2 macrophages mediate the beneficial effects of bariatric surgery against MASH. Hepatology. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
90.  Cao C, Liu W, Guo X, Weng S, Chen Y, Luo Y, Wang S, Zhu B, Liu Y, Peng D. Identification and validation of efferocytosis-related biomarkers for the diagnosis of metabolic dysfunction-associated steatohepatitis based on bioinformatics analysis and machine learning. Front Immunol. 2024;15:1460431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
91.  Hendrikx T, Porsch F, Kiss MG, Rajcic D, Papac-Miličević N, Hoebinger C, Goederle L, Hladik A, Shaw LE, Horstmann H, Knapp S, Derdak S, Bilban M, Heintz L, Krawczyk M, Paternostro R, Trauner M, Farlik M, Wolf D, Binder CJ. Soluble TREM2 levels reflect the recruitment and expansion of TREM2(+) macrophages that localize to fibrotic areas and limit NASH. J Hepatol. 2022;77:1373-1385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 88]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
92.  Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira da Costa M, Reis e Sousa C. Dendritic Cells Revisited. Annu Rev Immunol. 2021;39:131-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in RCA: 390]  [Article Influence: 97.5]  [Reference Citation Analysis (0)]
93.  Gardner A, de Mingo Pulido Á, Ruffell B. Dendritic Cells and Their Role in Immunotherapy. Front Immunol. 2020;11:924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in RCA: 303]  [Article Influence: 60.6]  [Reference Citation Analysis (0)]
94.  Nati M, Chung KJ, Chavakis T. The Role of Innate Immune Cells in Nonalcoholic Fatty Liver Disease. J Innate Immun. 2022;14:31-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 36]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
95.  Henning JR, Graffeo CS, Rehman A, Fallon NC, Zambirinis CP, Ochi A, Barilla R, Jamal M, Deutsch M, Greco S, Ego-Osuala M, Bin-Saeed U, Rao RS, Badar S, Quesada JP, Acehan D, Miller G. Dendritic cells limit fibroinflammatory injury in nonalcoholic steatohepatitis in mice. Hepatology. 2013;58:589-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in RCA: 139]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
96.  Schuster S, Cabrera D, Arrese M, Feldstein AE. Triggering and resolution of inflammation in NASH. Nat Rev Gastroenterol Hepatol. 2018;15:349-364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in RCA: 591]  [Article Influence: 84.4]  [Reference Citation Analysis (0)]
97.  Nati M, Haddad D, Birkenfeld AL, Koch CA, Chavakis T, Chatzigeorgiou A. The role of immune cells in metabolism-related liver inflammation and development of non-alcoholic steatohepatitis (NASH). Rev Endocr Metab Disord. 2016;17:29-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in RCA: 99]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
98.  Ito H, Hamerman JA. TREM-2, triggering receptor expressed on myeloid cell-2, negatively regulates TLR responses in dendritic cells. Eur J Immunol. 2012;42:176-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in RCA: 125]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
99.  Bouchon A, Hernández-Munain C, Cella M, Colonna M. A DAP12-mediated pathway regulates expression of CC chemokine receptor 7 and maturation of human dendritic cells. J Exp Med. 2001;194:1111-1122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in RCA: 362]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
100.  Nakao T, Ono Y, Dai H, Nakano R, Perez-Gutierrez A, Camirand G, Huang H, Geller DA, Thomson AW. DNAX Activating Protein of 12 kDa/Triggering Receptor Expressed on Myeloid Cells 2 Expression by Mouse and Human Liver Dendritic Cells: Functional Implications and Regulation of Liver Ischemia-Reperfusion Injury. Hepatology. 2019;70:696-710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in RCA: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
101.  Diehl AM, Day C. Cause, Pathogenesis, and Treatment of Nonalcoholic Steatohepatitis. N Engl J Med. 2017;377:2063-2072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 671]  [Cited by in RCA: 864]  [Article Influence: 108.0]  [Reference Citation Analysis (0)]
102.  Angulo P, Kleiner DE, Dam-Larsen S, Adams LA, Bjornsson ES, Charatcharoenwitthaya P, Mills PR, Keach JC, Lafferty HD, Stahler A, Haflidadottir S, Bendtsen F. Liver Fibrosis, but No Other Histologic Features, Is Associated With Long-term Outcomes of Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology. 2015;149:389-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2137]  [Cited by in RCA: 2113]  [Article Influence: 211.3]  [Reference Citation Analysis (0)]
103.  Vilar-Gomez E, Calzadilla-Bertot L, Wai-Sun Wong V, Castellanos M, Aller-de la Fuente R, Metwally M, Eslam M, Gonzalez-Fabian L, Alvarez-Quiñones Sanz M, Conde-Martin AF, De Boer B, McLeod D, Hung Chan AW, Chalasani N, George J, Adams LA, Romero-Gomez M. Fibrosis Severity as a Determinant of Cause-Specific Mortality in Patients With Advanced Nonalcoholic Fatty Liver Disease: A Multi-National Cohort Study. Gastroenterology. 2018;155:443-457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 390]  [Cited by in RCA: 561]  [Article Influence: 80.1]  [Reference Citation Analysis (0)]
104.  Khan S, Saxena R. Regression of Hepatic Fibrosis and Evolution of Cirrhosis: A Concise Review. Adv Anat Pathol. 2021;28:408-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
105.  Friedman SL. Hepatic Fibrosis and Cancer: The Silent Threats of Metabolic Syndrome. Diabetes Metab J. 2024;48:161-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
106.  Xia S, Huang Y, Zhang Y, Zhang M, Zhao K, Han P, Tian D, Liao J, Liu J. Role of macrophage-to-myofibroblast transition in chronic liver injury and liver fibrosis. Eur J Med Res. 2023;28:502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
107.  Duffield JS, Forbes SJ, Constandinou CM, Clay S, Partolina M, Vuthoori S, Wu S, Lang R, Iredale JP. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest. 2005;115:56-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in RCA: 718]  [Article Influence: 35.9]  [Reference Citation Analysis (0)]
108.  Zhang X, Zeng Y, Zhao L, Xu Q, Miao D, Yu F. Targeting Hepatic Stellate Cell Death to Reverse Hepatic Fibrosis. Curr Drug Targets. 2023;24:568-583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
109.  Cheng D, Chai J, Wang H, Fu L, Peng S, Ni X. Hepatic macrophages: Key players in the development and progression of liver fibrosis. Liver Int. 2021;41:2279-2294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 97]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
110.  Jiang K, Lu S, Li D, Liu M, Jin H, Lei B, Wang S, Long K, He S, Zhong F. Blockade of C5aR1 alleviates liver inflammation and fibrosis in a mouse model of NASH by regulating TLR4 signaling and macrophage polarization. J Gastroenterol. 2023;58:894-907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
111.  Luo Y, Guo J, Jia W, Wu M, Yin F, Niu G, Shih DQ, Targan SR, Zhang X. TNF-Like Ligand 1 Aberrance Aggravates Nonalcoholic Steatohepatitis via M1 Macrophage Polarization. Oxid Med Cell Longev. 2021;2021:3877617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
112.  Yang Y, Han CY, Guan QB, Ruan SL. [Interleukin-17-mediated inflammation promotes nonalcoholic fatty liver disease in mice with regulation of M1-type macrophage polarization]. Zhonghua Gan Zang Bing Za Zhi. 2018;26:916-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
113.  Zhou D, Huang W, Wei J, Zhang J, Liu Z, Ji R, Ge S, Xiao M, Fan Y, Lu C. RelB promotes liver fibrosis via inducing the release of injury-associated inflammatory cytokines. J Cell Mol Med. 2020;24:6008-6014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
114.  Feng M, Ding J, Wang M, Zhang J, Zhu X, Guan W. Kupffer-derived matrix metalloproteinase-9 contributes to liver fibrosis resolution. Int J Biol Sci. 2018;14:1033-1040.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in RCA: 68]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
115.  Shi H, Wang X, Li F, Gerlach BD, Yurdagul A Jr, Moore MP, Zeldin S, Zhang H, Cai B, Zheng Z, Valenti L, Tabas I. CD47-SIRPα axis blockade in NASH promotes necroptotic hepatocyte clearance by liver macrophages and decreases hepatic fibrosis. Sci Transl Med. 2022;14:eabp8309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in RCA: 32]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
116.  Ramachandran P, Dobie R, Wilson-Kanamori JR, Dora EF, Henderson BEP, Luu NT, Portman JR, Matchett KP, Brice M, Marwick JA, Taylor RS, Efremova M, Vento-Tormo R, Carragher NO, Kendall TJ, Fallowfield JA, Harrison EM, Mole DJ, Wigmore SJ, Newsome PN, Weston CJ, Iredale JP, Tacke F, Pollard JW, Ponting CP, Marioni JC, Teichmann SA, Henderson NC. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature. 2019;575:512-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 868]  [Cited by in RCA: 973]  [Article Influence: 162.2]  [Reference Citation Analysis (0)]
117.  Lee KJ, An S, Kim MY, Kim SM, Jeong WI, Ko HJ, Yang YM, Noh M, Han YH. Hepatic TREM2(+) macrophages express matrix metalloproteinases to control fibrotic scar formation. Immunol Cell Biol. 2023;101:216-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
118.  He W, Huang C, Wang L, Su W, Wang S, Huang P, Zhang X, Huang Y, Zhao Y, Lin M, Shi X, Li X. The correlation between triiodothyronine and the severity of liver fibrosis. BMC Endocr Disord. 2022;22:313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
119.  Liu Z, Xiang H, Xiang D, Xiao S, Xiang H, Xiao J, Ren H, Hu P, Liu H, Peng M. Revealing potential anti-fibrotic mechanism of Ganxianfang formula based on RNA sequence. Chin Med. 2022;17:23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
120.  Powell EE, Cooksley WG, Hanson R, Searle J, Halliday JW, Powell LW. The natural history of nonalcoholic steatohepatitis: a follow-up study of forty-two patients for up to 21 years. Hepatology. 1990;11:74-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1029]  [Cited by in RCA: 969]  [Article Influence: 27.7]  [Reference Citation Analysis (0)]
121.  Stine JG, Wentworth BJ, Zimmet A, Rinella ME, Loomba R, Caldwell SH, Argo CK. Systematic review with meta-analysis: risk of hepatocellular carcinoma in non-alcoholic steatohepatitis without cirrhosis compared to other liver diseases. Aliment Pharmacol Ther. 2018;48:696-703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in RCA: 215]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
122.  Mittal S, El-Serag HB, Sada YH, Kanwal F, Duan Z, Temple S, May SB, Kramer JR, Richardson PA, Davila JA. Hepatocellular Carcinoma in the Absence of Cirrhosis in United States Veterans is Associated With Nonalcoholic Fatty Liver Disease. Clin Gastroenterol Hepatol. 2016;14:124-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 373]  [Cited by in RCA: 454]  [Article Influence: 50.4]  [Reference Citation Analysis (1)]
123.  Paradis V, Zalinski S, Chelbi E, Guedj N, Degos F, Vilgrain V, Bedossa P, Belghiti J. Hepatocellular carcinomas in patients with metabolic syndrome often develop without significant liver fibrosis: a pathological analysis. Hepatology. 2009;49:851-859.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 416]  [Cited by in RCA: 398]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
124.  Huang DQ, El-Serag HB, Loomba R. Global epidemiology of NAFLD-related HCC: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol. 2021;18:223-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 840]  [Cited by in RCA: 1049]  [Article Influence: 262.3]  [Reference Citation Analysis (0)]
125.  Kanwal F, Kramer JR, Mapakshi S, Natarajan Y, Chayanupatkul M, Richardson PA, Li L, Desiderio R, Thrift AP, Asch SM, Chu J, El-Serag HB. Risk of Hepatocellular Cancer in Patients With Non-Alcoholic Fatty Liver Disease. Gastroenterology. 2018;155:1828-1837.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in RCA: 525]  [Article Influence: 75.0]  [Reference Citation Analysis (0)]
126.  Llovet JM, Willoughby CE, Singal AG, Greten TF, Heikenwälder M, El-Serag HB, Finn RS, Friedman SL. Nonalcoholic steatohepatitis-related hepatocellular carcinoma: pathogenesis and treatment. Nat Rev Gastroenterol Hepatol. 2023;20:487-503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in RCA: 122]  [Reference Citation Analysis (0)]
127.  Tang W, Lv B, Yang B, Chen Y, Yuan F, Ma L, Chen S, Zhang S, Xia J. TREM2 acts as a tumor suppressor in hepatocellular carcinoma by targeting the PI3K/Akt/β-catenin pathway. Oncogenesis. 2019;8:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in RCA: 47]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
128.  Cubero FJ. Keeping the liver fit with TREM2 during hepatic carcinogenesis. Gut. 2021;70:1210-1211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
129.  Tan J, Fan W, Liu T, Zhu B, Liu Y, Wang S, Wu J, Liu J, Zou F, Wei J, Liu L, Zhang X, Zhuang J, Wang Y, Lin H, Huang X, Chen S, Kuang M, Li J. TREM2(+) macrophages suppress CD8(+) T-cell infiltration after transarterial chemoembolisation in hepatocellular carcinoma. J Hepatol. 2023;79:126-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 62]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
130.  Wang Q, Zheng K, Tan D, Liang G. TREM2 knockdown improves the therapeutic effect of PD-1 blockade in hepatocellular carcinoma. Biochem Biophys Res Commun. 2022;636:140-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
131.  Ye Y, Song Y, Zhuang J, Wang G, Ni J, Xia W. Anticancer effects of echinacoside in hepatocellular carcinoma mouse model and HepG2 cells. J Cell Physiol. 2019;234:1880-1888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
132.  Ganguly S, Rosenthal SB, Ishizuka K, Troutman TD, Rohm TV, Khader N, Aleman-Muench G, Sano Y, Archilei S, Soroosh P, Olefsky JM, Feldstein AE, Kisseleva T, Loomba R, Glass CK, Brenner DA, Dhar D. Lipid-associated macrophages' promotion of fibrosis resolution during MASH regression requires TREM2. Proc Natl Acad Sci U S A. 2024;121:e2405746121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
133.  Yu W, Zhang Y, Sun L, Huang W, Li X, Xia N, Chen X, Wikana LP, Xiao Y, Chen M, Han S, Wang Z, Pu L. Myeloid Trem2 ameliorates the progression of metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. Metabolism. 2024;155:155911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
134.  Wang S, Mustafa M, Yuede CM, Salazar SV, Kong P, Long H, Ward M, Siddiqui O, Paul R, Gilfillan S, Ibrahim A, Rhinn H, Tassi I, Rosenthal A, Schwabe T, Colonna M. Anti-human TREM2 induces microglia proliferation and reduces pathology in an Alzheimer's disease model. J Exp Med. 2020;217:e20200785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in RCA: 266]  [Article Influence: 66.5]  [Reference Citation Analysis (0)]
135.  Hampel H, Caraci F, Cuello AC, Caruso G, Nisticò R, Corbo M, Baldacci F, Toschi N, Garaci F, Chiesa PA, Verdooner SR, Akman-Anderson L, Hernández F, Ávila J, Emanuele E, Valenzuela PL, Lucía A, Watling M, Imbimbo BP, Vergallo A, Lista S. A Path Toward Precision Medicine for Neuroinflammatory Mechanisms in Alzheimer's Disease. Front Immunol. 2020;11:456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in RCA: 203]  [Article Influence: 40.6]  [Reference Citation Analysis (0)]