TREM2

Last updated
TREM2
Identifiers
Aliases TREM2 , TREM-2, Trem2a, Trem2b, Trem2c, triggering receptor expressed on myeloid cells 2, PLOSL2
External IDs OMIM: 605086; MGI: 1913150; HomoloGene: 10352; GeneCards: TREM2; OMA:TREM2 - orthologs
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_001271821
NM_018965

NM_001272078
NM_031254

RefSeq (protein)

NP_001258750
NP_061838

NP_001259007
NP_112544

Location (UCSC) Chr 6: 41.16 – 41.16 Mb Chr 17: 48.65 – 48.66 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Triggering receptor expressed on myeloid cells 2(TREM2) is a protein that in humans is encoded by the TREM2 gene. [5] [6] [7] TREM2 is expressed on macrophages, immature monocyte-derived dendritic cells, osteoclasts, and microglia, [8] which are immune cells in the central nervous system. [9] In the liver, TREM2 is expressed by several cell types, including macrophages, that respond to injury. [10] In the intestine, TREM2 is expressed by myeloid-derived dendritic cells and macrophage. [11] TREM2 is overexpressed in many tumor types and has anti-inflammatory activities. It might therefore be a good therapeutic target.

Contents

Gene

The TREM2 gene lies on the sixth chromosome in humans, specifically in location 6p21.1. The gene has 5 coding exon regions. [12] [13] Alternative splicing of the TREM2 mRNA transcript leads to different isoforms of the protein being produced upon translation. [12] Specifically, TREM2 mRNA has 3 different isoforms containing 3 consistent exons, and 2 that vary between the isoforms. [14] TREM2 mRNA is most highly expressed in brain, lungs, adrenal glands, placenta, gall bladder, and colon. [12] The functions of TREM2 have been studied in mice with disruption or mutation of the mouse ortholog, Trem2 . [15] TREM2 orthologs are also present in rat, dog, Rhesus monkey, macaque, chimpanzee, and other animals. [16]

Protein

The TREM2 receptor with the ADAM10 and ADAM17 enzymes that create the soluble TREM2 fragment. Created with BioRender.com STREM2-2.jpg
The TREM2 receptor with the ADAM10 and ADAM17 enzymes that create the soluble TREM2 fragment. Created with BioRender.com

The TREM2 receptor is a transmembrane protein that is made up of an extracellular region (also referred to as the ectodomain), the membrane-traversing segment, and an intracellular component. [17] The extracellular component of TREM2 can bind different anionic ligands, specifically glycoproteins and lipids. [18] [19] This ectodomain component includes an Ig-like V-type domain, where ligands bind the receptor. [20] The TREM2 ectodomain is modified after protein translation; these modifications affect its affinity for different ligands. [14] The intracellular component of TREM2 does not have any signaling ability on its own; rather, it signals via the DNAX activator proteins 10 and 12 (DAP10 and DAP12). A single TREM2 molecule can interact with DAP10 and DAP12 at the same time. [19]

Part of the ectodomain of TREM2 can be processed by enzymes (ADAM10, ADAM17) and released as a soluble version, called soluble TREM2 (sTREM2). [14] This protein fragment is released into the serum and cerebral spinal fluid (CSF), and might serve as a biomarker for neurodegenerative and other disorders, but further studies are needed. [14]

Function

TREM2 structure as identified with X-ray crystallography. Image available through RCSB PDB. 5eli assembly-1.jpg
TREM2 structure as identified with X-ray crystallography. Image available through RCSB PDB.

The TREM2 protein is found in immune cells termed myeloid cells, which include macrophages, granulocytes, monocytes, and dendritic cells. [22] Monocyte-, macrophage-, and neutrophil-mediated inflammatory responses can be stimulated through G protein-linked 7-transmembrane receptors (e.g., FPR1), Fc receptors, CD14, toll like receptors (such as TLR4), and cytokine receptors (e.g., IFNGR1). [23] [24] Engagement of these receptors can also prime myeloid cells to respond to other stimuli. Myeloid cells express receptors belonging to the immunoglobulin (Ig) superfamily, such as TREM2, or to the C-type lectin superfamily. [23] [25]

On myeloid cells, TREM2 binds anionic molecules, free and bound to plasma membrane, including bacterial products, DNA, lipoproteins, phospholipids, glycoproteins, DNA, and bacterial fragments. [13] [18] [19] [26] [27] TREM2 binding of ligand results in phosphorylation at 2 tyrosines in the immunoreceptor tyrosine-based activation motif (ITAM) of DAP12 by SRC tyrosine kinases. [19] Spleen tyrosine kinase (Syk) interacts with these phosphorylation sites and activates the phosphatidylinositol-3 kinase (PI3K) signaling pathway, as well as other signaling molecules such as mTOR, MAPK, and ERK. [19] [28] Association of TREM2 with DAP10 also activates the PI3K signaling pathway, [29] leading to expression of transcription factors that include AP1, NF-κB, and NFAT. [28] The PI3K signaling pathway also increases intracellular calcium content, which activates calcium-dependent kinases. [28] [29] TREM2 activation also affects expression of GAL1, GAL3, IL1RN, and progranulin, which modulate the immune response. [19]

TREM2 is expressed by microglia [30] and osteoclasts, and is involved in development and/or maintenance of brain and bone. [19] In mice, TREM2 is involved in synaptic pruning, a process of shaping neuronal circuitry by microglia- and astrocyte-mediated removal of excessive synapses via phagocytosis. [14] [31] [32] In brain, the highest levels of TREM2 are found in hippocampus, white matter, and the spinal cord, and levels of TREM2 increase with age in humans and mice. [30] TREM2 is also expressed by macrophages of adipose tissue, adrenal gland, and placenta. [19]

Immunosuppressive tumor-associated macrophages (TAMs) have been characterized by expression of TREM2. [33] TREM2 signaling leads to downregulated transcription of genes that promote inflammation (Tnf, Il1b, and Nos2), [34] as well as release of cytokines that prevent activation of anti-tumor CD8+ T cells. [35] TREM2+ immunosuppressive TAMs correlate with the level of exhausted T cells in the human tumor microenvironment (TME). [36] A TREM2+ TAM-rich TME therefore appears to be immune suppressive and might promote resistance to cancer therapies, such as checkpoint inhibitors.

TREM2 signaling can antagonize TLR expression and signaling, resulting in reduced production of inflammatory cytokines by cultured mouse macrophages. [19] Conversely, TREM2 expression is reduced following inflammatory signaling induction by lipopolysaccharide (a TLR4 ligand) or interferon gamma (IFNG). [37] The neuroprotective effects of TREM2 involve not only production of anti-inflammatory cytokines, but also clearance of abnormal proteins and phagocytosis of apoptotic neurons. [14] [34]

In contrast to anti-inflammatory effects in brain and cancer, TREM2 signaling has been reported to contribute to intestinal inflammation and development of inflammatory bowel diseases (IBD). [11] sTREM2 is believed to negatively regulate TREM2 signaling by acting as decoy receptors. [38] sTREM might therefore have pro-inflammatory effects. [19] sTREM2 has been indicated in activating signaling pathways such as PI3K and ERK through an unidentified receptor. [39] Levels of sTREM2 are increased in CSF of patients with Alzheimer's disease, and correlate with the CSF levels of disease biomarkers, such as t-tau and p-tau. [40]

Association with diseases

TREM2 signaling has been associated with pathogenesis of several diseases. Variants of in the DAP12 (TYROBP) or TREM2 genes have been associated with polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL or Nasu–Hakola disease). [41]

Mutations in TREM2 associated with Alzheimer's disease lead to decreased binding affinity for ligands, resulting in reduced microglial responses to amyloid-beta plaques. Created with BioRender.com TREM2 Alzheimer's.jpg
Mutations in TREM2 associated with Alzheimer's disease lead to decreased binding affinity for ligands, resulting in reduced microglial responses to amyloid-beta plaques. Created with BioRender.com

Alzheimer's disease

Variants of TREM2 have been associated with neurodegenerative disorders, including Alzheimer's disease. [42] TREM2 is involved in the microglial response to the amyloid plaques that are characteristic of AD. Loss of TREM2 function reduces the responses of microglia to plaques, which then appear to take on a more toxic state. [42] Expression of TREM2 is associated with that of CD33. [43] [44] [45]

Zhong et al. reported that, in mice, stereotactic injection of sTREM2 or adeno-associated virus-mediated activation of sTREM2 reduced the amyloid plaque load and reduced functional memory deficits. [46] Moreover, sTREM2 stimulated microglial proliferation and homing toward amyloid plaques where amyloid-β uptake and degradation was increased. Interestingly, these effects were specifically mediated by microglia. Level of sTREM2 in the CSF might be a biomarker for Alzheimer's disease and the associated inflammatory response. [47] [48] [49]

Cancer

Although TREM2 expression is low in most normal tissues, it is overexpressed in many human tumor types. [50] An analysis of levels of TREM2 mRNA in 33 cancer tissues from The Cancer Genome Atlas (TCGA) indicate higher levels of expression in tumor vs normal tissues in 18 cancer types, including head and neck squamous cell carcinoma, colon adenocarcinoma, and glioblastoma, as well as gynecologic, liver, gastric, kidney, breast, bladder, and esophageal cancers. [50] High expression of TREM2 was associated with shorter survival times of patients with ovarian cancer, gastric cancer, lower-grade glioma, hepatocellular carcinoma, or renal clear cell carcinoma. [50] Tumor infiltration by TREM2+, APOE +, C1q + macrophage was reported to be a biomarker for recurrence of clear-cell renal carcinoma. [51] [52] TREM2+ macrophages from human tumors also express CD68, CD163, CSF1R, and nuclear MAFB. [53]

IBD

TREM2 expressed by human monocyte dendritic cells [11] in the intestine. [54] Expression of TREM2 is limited to inflamed sections of intestine and contribute to IBD development. [11] TREM2 is associated with increased production of inflammatory cytokines and changes in the gut microbiota. [11]

Liver disease

One feature of liver disease is the initiation of an inflammatory process, leading to fibrosis and steatohepatitis. In mouse models of nonalcoholic steatohepatitis (NASH), disease development was associated with liver infiltration by monocyte-derived macrophages and increased expression of Trem2 and Cd9. [55] Mice with disruption of Trem2 had more severe liver damage following administration of carbon tetrachloride or acetaminophen, compared to mice without gene disruption. [56] The authors of this study found that TREM2 is expressed by Kupfer cells and hepatic stellate cells, indicating that TREM2 might downregulate inflammation. [56] Expression was also increased in liver tissues from patients with cirrhosis. [56] Compared with non-tumor liver tissue, TREM2 expression was increased in tumors from mice and patients with hepatocellular carcinoma (HCC). [57] This study also showed that disruption of Trem2 promoted tumor development and exacerbated liver damage and inflammation. In liver tumors, TREM2 was expressed by tumor-infiltrating macrophages (TAMs). TREM2 might therefore promote the resolution of inflammation during hepatic injury, ultimately preventing parenchymal cell death.

Mutations in the TREM2 or TYROBP genes (encodes DAP12) can lead to development of PLOSL. This disease is characterized by dysfunctional microglia, bone cysts and fractures, frontal lobe syndrome, and dementia. Created with BioRender.com TREM2 NHD-2.jpg
Mutations in the TREM2 or TYROBP genes (encodes DAP12) can lead to development of PLOSL. This disease is characterized by dysfunctional microglia, bone cysts and fractures, frontal lobe syndrome, and dementia. Created with BioRender.com

PLOSL or Nasu–Hakola disease

PLOSL or Nasu–Hakola disease is a neurodegenerative disorder characterized by bone cysts, dementia, and early death and is associated with variants in the TYROBP gene (encodes DAP12 protein) and TREM2 gene. [41] Bone cysts in patients with PLOSL contain fat in lieu of bone marrow. [28] In this disease, the main cell type in the brain that is affected is the microglia, where TREM2 is expressed. [58] Several recessive, inactivating mutations in TREM2 and TYROBP (encodes DAP12 protein) have been identified that can cause PLOSL. [20] [59] [60] The mutations prevent association between TREM2 and DAP12 or expression of shorter, non-functional forms of TREM2. [28] [60] Loss of function of TREM2 signaling increases the inflammatory responses of microglia, [58] reducing clearance of dead neurons and promoting inflammation and even formation of amyloid plaques. [58]

Stroke

During ischemic stroke, microglia respond to the area of insult. TREM2 appears to reduce the inflammatory response induced by TLR signaling and promote microglial migration, survival, and regeneration. [61] [62]

Other diseases

TREM2 has also been linked to additional disorders such as ALS, Parkinson's disease, and more dementia related conditions. [20]

Therapeutic targeting of TREM2

TREM2 is a good therapeutic target for several diseases, including cancer and liver and neurodegenerative diseases. Several companies are developing agents to target TREM2. However, TREM2 is likely to have distinct roles in the pathogenesis of these disorders, so therapeutic agents in development employ different approaches to modify TREM2 activity.

Neurodegenerative diseases

In the brain, TREM2 is expressed on microglia that regulate clearance of neuronal debris. [19] Binding of apolipoproteins, such as ApoE, to TREM2 promotes phagocytosis of apoptotic neurons or the uptake of amyloid beta by microglia. [63] Variants of TREM2 that encode proteins with reduced affinity for ligands have been associated with Alzheimer’s disease. [64]

Targeting sTREM2

A potential mechanism of intervention could be targeting the enzymes that cleave the ectodomain, adjusting the rate at which sTREM2 is released. In rodents, a potential therapeutic using this mechanism was used against AD pathology, and the rodents had smaller plaques than controls. [19]

Related Research Articles

<span class="mw-page-title-main">Macrophage</span> Type of white blood cell

Macrophages are a type of white blood cell of the innate immune system that engulf and digest pathogens, such as cancer cells, microbes, cellular debris, and foreign substances, which do not have proteins that are specific to healthy body cells on their surface. This process is called phagocytosis, which acts to defend the host against infection and injury.

<span class="mw-page-title-main">Tumor necrosis factor</span> Protein

Tumor necrosis factor ('TNF), also known as cachexin or cachectin, is a cytokine and a member of the TNF superfamily, which comprises various transmembrane proteins with a homologous TNF domain. Initially referred to as TNFα, this cytokine was the first to be identified as an adipokine, secreted by adipose tissue. TNF signaling occurs through two receptors: TNFR1 and TNFR2. TNFR1 is widely expressed across most cell types and is typically associated with pro-inflammatory and apoptotic signaling. In contrast, TNFR2 is mainly found on endothelial, epithelial, and immune cells, where it mediates anti-inflammatory responses and promotes cell proliferation.

<span class="mw-page-title-main">Microglia</span> Glial cell located throughout the brain and spinal cord

Microglia are a type of neuroglia located throughout the brain and spinal cord. Microglia account for about 10-15% of cells found within the brain. As the resident macrophage cells, they act as the first and main form of active immune defense in the central nervous system (CNS). Microglia originate in the yolk sac under a tightly regulated molecular process. These cells are distributed in large non-overlapping regions throughout the CNS. Microglia are key cells in overall brain maintenance—they are constantly scavenging the CNS for plaques, damaged or unnecessary neurons and synapses, and infectious agents. Since these processes must be efficient to prevent potentially fatal damage, microglia are extremely sensitive to even small pathological changes in the CNS. This sensitivity is achieved in part by the presence of unique potassium channels that respond to even small changes in extracellular potassium. Recent evidence shows that microglia are also key players in the sustainment of normal brain functions under healthy conditions. Microglia also constantly monitor neuronal functions through direct somatic contacts and exert neuroprotective effects when needed.

<span class="mw-page-title-main">Integrin alpha M</span> Mammalian protein found in Homo sapiens

Integrin alpha M (ITGAM) is one protein subunit that forms heterodimeric integrin alpha-M beta-2 (αMβ2) molecule, also known as macrophage-1 antigen (Mac-1) or complement receptor 3 (CR3). ITGAM is also known as CR3A, and cluster of differentiation molecule 11B (CD11B). The second chain of αMβ2 is the common integrin β2 subunit known as CD18, and integrin αMβ2 thus belongs to the β2 subfamily integrins.

<span class="mw-page-title-main">Interleukin 19</span> Protein-coding gene in the species Homo sapiens

Interleukin 19 (IL-19) is an immunosuppressive protein that belongs to the IL-10 cytokine subfamily.

<span class="mw-page-title-main">CD33</span> Mammalian protein found in Homo sapiens

CD33 or Siglec-3 is a transmembrane receptor expressed on cells of myeloid lineage. It is usually considered myeloid-specific, but it can also be found on some lymphoid cells.

<span class="mw-page-title-main">CX3C motif chemokine receptor 1</span> Protein-coding gene in the species Homo sapiens

CX3C motif chemokine receptor 1 (CX3CR1), also known as the fractalkine receptor or G-protein coupled receptor 13 (GPR13), is a transmembrane protein of the G protein-coupled receptor 1 (GPCR1) family and the only known member of the CX3C chemokine receptor subfamily.

<span class="mw-page-title-main">Toll-like receptor 4</span> Cell surface receptor found in humans

Toll-like receptor 4 (TLR4), also designated as CD284, is a key activator of the innate immune response and plays a central role in the fight against bacterial infections. TLR4 is a transmembrane protein of approximately 95 kDa that is encoded by the TLR4 gene.

<span class="mw-page-title-main">P2RX7</span> Protein-coding gene in the species Homo sapiens

P2X purinoceptor 7 is a protein that in humans is encoded by the P2RX7 gene.

<span class="mw-page-title-main">TYROBP</span> Protein-coding gene in the species Homo sapiens

TYRO protein tyrosine kinase-binding protein is an adapter protein that in humans is encoded by the TYROBP gene.

<span class="mw-page-title-main">C5AR2</span> Protein-coding gene in the species Homo sapiens

Complement component 5a receptor 2 is a protein of the complement system that in humans is encoded by the C5AR2 gene. It is highly expressed in the blood and spleen, predominantly by myeloid cells.

<span class="mw-page-title-main">GPR84</span> Protein-coding gene in the species Homo sapiens

Probable G-protein coupled receptor 84 is a protein that in humans is encoded by the GPR84 gene.

<span class="mw-page-title-main">Hydroxycarboxylic acid receptor 2</span> Protein-coding gene in the species Homo sapiens

Hydroxycarboxylic acid receptor 2 (HCA2), also known as GPR109A and niacin receptor 1 (NIACR1), is a protein which in humans is encoded (its formation is directed) by the HCAR2 gene and in rodents by the Hcar2 gene. The human HCAR2 gene is located on the long (i.e., "q") arm of chromosome 12 at position 24.31 (notated as 12q24.31). Like the two other hydroxycarboxylic acid receptors, HCA1 and HCA3, HCA2 is a G protein-coupled receptor (GPCR) located on the surface membrane of cells. HCA2 binds and thereby is activated by D-β-hydroxybutyric acid (hereafter termed β-hydroxybutyric acid), butyric acid, and niacin (also known as nicotinic acid). β-Hydroxybutyric and butyric acids are regarded as the endogenous agents that activate HCA2. Under normal conditions, niacin's blood levels are too low to do so: it is given as a drug in high doses in order to reach levels that activate HCA2.

<span class="mw-page-title-main">Colony stimulating factor 1 receptor</span> Protein found in humans

Colony stimulating factor 1 receptor (CSF1R), also known as macrophage colony-stimulating factor receptor (M-CSFR), and CD115, is a cell-surface protein encoded by the human CSF1R gene. CSF1R is a receptor that can be activated by two ligands: colony stimulating factor 1 (CSF-1) and interleukin-34 (IL-34). CSF1R is highly expressed in myeloid cells, and CSF1R signaling is necessary for the survival, proliferation, and differentiation of many myeloid cell types in vivo and in vitro. CSF1R signaling is involved in many diseases and is targeted in therapies for cancer, neurodegeneration, and inflammatory bone diseases.

<span class="mw-page-title-main">Granulin</span> Protein-coding gene in humans

Granulin is a protein that in humans is encoded by the GRN gene. Each granulin protein is cleaved from the precursor progranulin, a 593 amino-acid-long and 68.5 kDa protein. While the function of progranulin and granulin have yet to be determined, both forms of the protein have been implicated in development, inflammation, cell proliferation and protein homeostasis. The 2006 discovery of the GRN mutation in a population of patients with frontotemporal dementia has spurred much research in uncovering the function and involvement in disease of progranulin in the body. While there is a growing body of research on progranulin's role in the body, studies on specific granulin residues are still limited.

<span class="mw-page-title-main">TREM1</span> Protein-coding gene in the species Homo sapiens

Triggering receptor expressed on myeloid cells 1 (TREM1) is an immunoglobulin (Ig) superfamily transmembrane protein that, in humans, is encoded by the TREM1 gene. TREM1 is constitutively expressed on the surface of peripheral blood monocytes and neutrophils, and upregulated by toll-like receptor (TLR) ligands; activation of TREM1 amplifies immune responses.

<span class="mw-page-title-main">MARCO</span> Protein-coding gene in the species Homo sapiens

Macrophage receptor with collagenous structure (MARCO) is a protein that in humans is encoded by the MARCO gene. MARCO is a class A scavenger receptor that is found on particular subsets of macrophages. Scavenger receptors are pattern recognition receptors (PRRs) found most commonly on immune cells. Their defining feature is that they bind to polyanions and modified forms of a type of cholesterol called low-density lipoprotein (LDL). MARCO is able to bind and phagocytose these ligands and pathogen-associated molecular patterns (PAMPs), leading to the clearance of pathogens and cell signaling events that lead to inflammation. As part of the innate immune system, MARCO clears, or scavenges, pathogens, which leads to inflammatory responses. The scavenger receptor cysteine-rich (SRCR) domain at the end of the extracellular side of MARCO binds ligands to activate the subsequent immune responses. MARCO expression on macrophages has been associated with tumor development and also with Alzheimer's disease, via decreased responses of cells when ligands bind to MARCO.

<span class="mw-page-title-main">Peptidoglycan recognition protein 1</span> Protein-coding gene in the species Homo sapiens

Peptidoglycan recognition protein 1, PGLYRP1, also known as TAG7, is an antibacterial and pro-inflammatory innate immunity protein that in humans is encoded by the PGLYRP1 gene.

Microglia are the primary immune cells of the central nervous system, similar to peripheral macrophages. They respond to pathogens and injury by changing morphology and migrating to the site of infection/injury, where they destroy pathogens and remove damaged cells.

Melanie Greter is a Swiss neuroimmunologist and a Swiss National Science Foundation Professor in the Institute of Experimental Immunology at the University of Zurich. Greter explores the ontogeny and function of microglia and border-associated macrophages of the central nervous system to understand how they maintain homeostasis and contribute to brain-related diseases.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000095970 Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000023992 Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. Bouchon A, Dietrich J, Colonna M (May 2000). "Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes". Journal of Immunology. 164 (10): 4991–4995. doi: 10.4049/jimmunol.164.10.4991 . PMID   10799849.
  6. Paloneva J, Manninen T, Christman G, Hovanes K, Mandelin J, Adolfsson R, et al. (September 2002). "Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype". American Journal of Human Genetics. 71 (3): 656–662. doi:10.1086/342259. PMC   379202 . PMID   12080485.
  7. "Entrez Gene: TREM2 triggering receptor expressed on myeloid cells 2".
  8. Rodríguez-Gómez JA, Kavanagh E, Engskog-Vlachos P, Engskog MK, Herrera AJ, Espinosa-Oliva AM, et al. (July 2020). "Microglia: Agents of the CNS Pro-Inflammatory Response". Cells. 9 (7): E1717. doi: 10.3390/cells9071717 . PMC   7407646 . PMID   32709045.
  9. Masuda T, Sankowski R, Staszewski O, Prinz M (February 2020). "Microglia Heterogeneity in the Single-Cell Era". Cell Reports. 30 (5): 1271–1281. doi: 10.1016/j.celrep.2020.01.010 . PMID   32023447. S2CID   211047418.
  10. Sun H, Feng J, Tang L (December 2020). "Function of TREM1 and TREM2 in Liver-Related Diseases". Cells. 9 (12): 2626. doi: 10.3390/cells9122626 . PMC   7762355 . PMID   33297569.
  11. 1 2 3 4 5 Genua M, Rutella S, Correale C, Danese S (October 2014). "The triggering receptor expressed on myeloid cells (TREM) in inflammatory bowel disease pathogenesis". Journal of Translational Medicine. 12: 293. doi: 10.1186/s12967-014-0293-z . PMC   4231187 . PMID   25347935.
  12. 1 2 3 "TREM2 triggering receptor expressed on myeloid cells 2 [Homo sapiens (human)] - Gene - NCBI". www.ncbi.nlm.nih.gov. Retrieved 2021-11-02.
  13. 1 2 Natale G, Biagioni F, Busceti CL, Gambardella S, Limanaqi F, Fornai F (September 2019). "TREM Receptors Connecting Bowel Inflammation to Neurodegenerative Disorders". Cells. 8 (10): E1124. doi: 10.3390/cells8101124 . PMC   6829526 . PMID   31546668.
  14. 1 2 3 4 5 6 Yang J, Fu Z, Zhang X, Xiong M, Meng L, Zhang Z (July 2020). "TREM2 ectodomain and its soluble form in Alzheimer's disease". Journal of Neuroinflammation. 17 (1): 204. doi: 10.1186/s12974-020-01878-2 . PMC   7341574 . PMID   32635934.
  15. "Jackson Laboratory Search- TREM2". Jackson Laboratory.
  16. Smith JR, Hayman GT, Wang SJ, Laulederkind SJ, Hoffman MJ, Kaldunski ML, et al. (January 2020). "The Year of the Rat: The Rat Genome Database at 20: a multi-species knowledgebase and analysis platform". Nucleic Acids Research. 48 (D1): D731–D742. doi:10.1093/nar/gkz1041. PMC   7145519 . PMID   31713623.
  17. Kulkarni B, Kumar D, Cruz-Martins N, Sellamuthu S (October 2021). "Role of TREM2 in Alzheimer's Disease: A Long Road Ahead". Molecular Neurobiology. 58 (10): 5239–5252. doi:10.1007/s12035-021-02477-9. PMID   34275100. S2CID   236090999.
  18. 1 2 Hamerman JA, Pottle J, Ni M, He Y, Zhang ZY, Buckner JH (January 2016). "Negative regulation of TLR signaling in myeloid cells--implications for autoimmune diseases". Immunological Reviews. 269 (1): 212–227. doi:10.1111/imr.12381. PMC   4703580 . PMID   26683155.
  19. 1 2 3 4 5 6 7 8 9 10 11 12 Deczkowska A, Weiner A, Amit I (June 2020). "The Physiology, Pathology, and Potential Therapeutic Applications of the TREM2 Signaling Pathway". Cell. 181 (6): 1207–1217. doi: 10.1016/j.cell.2020.05.003 . PMID   32531244. S2CID   219572314.
  20. 1 2 3 Dardiotis E, Siokas V, Pantazi E, Dardioti M, Rikos D, Xiromerisiou G, et al. (May 2017). "A novel mutation in TREM2 gene causing Nasu-Hakola disease and review of the literature". Neurobiology of Aging. 53: 194.e13–194.e22. doi:10.1016/j.neurobiolaging.2017.01.015. PMID   28214109. S2CID   22618858.
  21. Kober DL, Alexander-Brett JM, Karch CM, Cruchaga C, Colonna M, Holtzman MJ, Brett TJ (December 2016). "Neurodegenerative disease mutations in TREM2 reveal a functional surface and distinct loss-of-function mechanisms". eLife. 5: e20391. doi: 10.7554/eLife.20391 . PMC   5173322 . PMID   27995897.
  22. De Kleer I, Willems F, Lambrecht B, Goriely S (2014). "Ontogeny of myeloid cells". Frontiers in Immunology. 5: 423. doi: 10.3389/fimmu.2014.00423 . PMC   4153297 . PMID   25232355.
  23. 1 2 Futosi K, Fodor S, Mócsai A (November 2013). "Neutrophil cell surface receptors and their intracellular signal transduction pathways". International Immunopharmacology. 17 (3): 638–650. doi:10.1016/j.intimp.2013.06.034. PMC   3827506 . PMID   23994464.
  24. Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, et al. (January 2018). "Inflammatory responses and inflammation-associated diseases in organs". Oncotarget. 9 (6): 7204–7218. doi:10.18632/oncotarget.23208. PMC   5805548 . PMID   29467962.
  25. Sancho D, Reis e Sousa C (February 2013). "Sensing of cell death by myeloid C-type lectin receptors". Current Opinion in Immunology. 25 (1): 46–52. doi:10.1016/j.coi.2012.12.007. PMC   4480265 . PMID   23332826.
  26. Kober DL, Brett TJ (June 2017). "TREM2-Ligand Interactions in Health and Disease". Journal of Molecular Biology. 429 (11): 1607–1629. doi:10.1016/j.jmb.2017.04.004. PMC   5485854 . PMID   28432014.
  27. Kober DL, Brett TJ (June 2017). "TREM2-Ligand Interactions in Health and Disease". Journal of Molecular Biology. 429 (11): 1607–1629. doi:10.1016/j.jmb.2017.04.004. PMC   5485854 . PMID   28432014.
  28. 1 2 3 4 5 Xing J, Titus AR, Humphrey MB (2015). "The TREM2-DAP12 signaling pathway in Nasu-Hakola disease: a molecular genetics perspective". Research and Reports in Biochemistry. 5: 89–100. doi: 10.2147/RRBC.S58057 . PMC   4605443 . PMID   26478868.
  29. 1 2 Qiu H, Shao Z, Wen X, Jiang J, Ma Q, Wang Y, et al. (2021). "TREM2: Keeping Pace With Immune Checkpoint Inhibitors in Cancer Immunotherapy". Frontiers in Immunology. 12: 716710. doi: 10.3389/fimmu.2021.716710 . PMC   8446424 . PMID   34539652.
  30. 1 2 Gratuze M, Leyns CE, Holtzman DM (December 2018). "New insights into the role of TREM2 in Alzheimer's disease". Molecular Neurodegeneration. 13 (1): 66. doi: 10.1186/s13024-018-0298-9 . PMC   6302500 . PMID   30572908.
  31. Hong S, Dissing-Olesen L, Stevens B (February 2016). "New insights on the role of microglia in synaptic pruning in health and disease". Current Opinion in Neurobiology. 36: 128–134. doi:10.1016/j.conb.2015.12.004. PMC   5479435 . PMID   26745839.
  32. Filipello F, Morini R, Corradini I, Zerbi V, Canzi A, Michalski B, et al. (May 2018). "The Microglial Innate Immune Receptor TREM2 Is Required for Synapse Elimination and Normal Brain Connectivity". Immunity. 48 (5): 979–991.e8. doi: 10.1016/j.immuni.2018.04.016 . PMID   29752066. S2CID   21700781.
  33. Molgora M, Colonna M (June 2021). "Turning enemies into allies-reprogramming tumor-associated macrophages for cancer therapy". Med. 2 (6): 666–681. doi:10.1016/j.medj.2021.05.001. PMC   8238417 . PMID   34189494.
  34. 1 2 Takahashi K, Rochford CD, Neumann H (February 2005). "Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2". The Journal of Experimental Medicine. 201 (4): 647–657. doi:10.1084/jem.20041611. PMC   2213053 . PMID   15728241.
  35. Molgora M, Esaulova E, Vermi W, Hou J, Chen Y, Luo J, et al. (August 2020). "TREM2 Modulation Remodels the Tumor Myeloid Landscape Enhancing Anti-PD-1 Immunotherapy". Cell. 182 (4): 886–900.e17. doi:10.1016/j.cell.2020.07.013. PMC   7485282 . PMID   32783918.
  36. Binnewies M, Pollack JL, Rudolph J, Dash S, Abushawish M, Lee T, et al. (October 2021). "Targeting TREM2 on tumor-associated macrophages enhances immunotherapy". Cell Reports. 37 (3): 109844. doi: 10.1016/j.celrep.2021.109844 . PMID   34686340. S2CID   239472808.
  37. Gao X, Dong Y, Liu Z, Niu B (March 2013). "Silencing of triggering receptor expressed on myeloid cells-2 enhances the inflammatory responses of alveolar macrophages to lipopolysaccharide". Molecular Medicine Reports. 7 (3): 921–926. doi: 10.3892/mmr.2013.1268 . PMID   23314916.
  38. Piccio L, Buonsanti C, Cella M, Tassi I, Schmidt RE, Fenoglio C, et al. (November 2008). "Identification of soluble TREM-2 in the cerebrospinal fluid and its association with multiple sclerosis and CNS inflammation". Brain. 131 (Pt 11): 3081–3091. doi:10.1093/brain/awn217. PMC   2577803 . PMID   18790823.
  39. Konishi H, Kiyama H (2018). "Microglial TREM2/DAP12 Signaling: A Double-Edged Sword in Neural Diseases". Frontiers in Cellular Neuroscience. 12: 206. doi: 10.3389/fncel.2018.00206 . PMC   6087757 . PMID   30127720.
  40. Yang J, Fu Z, Zhang X, Xiong M, Meng L, Zhang Z (July 2020). "TREM2 ectodomain and its soluble form in Alzheimer's disease". Journal of Neuroinflammation. 17 (1): 204. doi: 10.1186/s12974-020-01878-2 . PMC   7341574 . PMID   32635934.
  41. 1 2 Bianchin MM, Capella HM, Chaves DL, Steindel M, Grisard EC, Ganev GG, et al. (February 2004). "Nasu-Hakola disease (polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy--PLOSL): a dementia associated with bone cystic lesions. From clinical to genetic and molecular aspects". Cellular and Molecular Neurobiology. 24 (1): 1–24. doi:10.1023/b:cemn.0000012721.08168.ee. PMID   15049507. S2CID   7089775.
  42. 1 2 Shi Y, Holtzman DM (December 2018). "Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight". Nature Reviews. Immunology. 18 (12): 759–772. doi:10.1038/s41577-018-0051-1. PMC   6425488 . PMID   30140051.
  43. Griciuc A, Patel S, Federico AN, Choi SH, Innes BJ, Oram MK, et al. (September 2019). "TREM2 Acts Downstream of CD33 in Modulating Microglial Pathology in Alzheimer's Disease". Neuron. 103 (5): 820–835.e7. doi:10.1016/j.neuron.2019.06.010. PMC   6728215 . PMID   31301936.
  44. Chan G, White CC, Winn PA, Cimpean M, Replogle JM, Glick LR, et al. (November 2015). "CD33 modulates TREM2: convergence of Alzheimer loci". Nature Neuroscience. 18 (11): 1556–1558. doi:10.1038/nn.4126. PMC   4682915 . PMID   26414614.
  45. Stetka B (2022-01-30). "How a hyperactive cell in the brain might trigger Alzheimer's disease". NPR. Retrieved 2022-02-17.
  46. Zhong L, Xu Y, Zhuo R, Wang T, Wang K, Huang R, et al. (March 2019). "Soluble TREM2 ameliorates pathological phenotypes by modulating microglial functions in an Alzheimer's disease model". Nature Communications. 10 (1): 1365. Bibcode:2019NatCo..10.1365Z. doi:10.1038/s41467-019-09118-9. PMC   6433910 . PMID   30911003.
  47. Liu D, Cao B, Zhao Y, Huang H, McIntyre RS, Rosenblat JD, Zhou H (November 2018). "Soluble TREM2 changes during the clinical course of Alzheimer's disease: A meta-analysis". Neuroscience Letters. 686: 10–16. doi:10.1016/j.neulet.2018.08.038. PMID   30171911. S2CID   52146124.
  48. Rauchmann BS, Schneider-Axmann T, Alexopoulos P, Perneczky R (February 2019). "CSF soluble TREM2 as a measure of immune response along the Alzheimer's disease continuum". Neurobiology of Aging. 74: 182–190. doi:10.1016/j.neurobiolaging.2018.10.022. PMC   6331262 . PMID   30458365.
  49. Piccio L, Deming Y, Del-Águila JL, Ghezzi L, Holtzman DM, Fagan AM, et al. (June 2016). "Cerebrospinal fluid soluble TREM2 is higher in Alzheimer disease and associated with mutation status". Acta Neuropathologica. 131 (6): 925–933. doi:10.1007/s00401-016-1533-5. PMC   4867123 . PMID   26754641.
  50. 1 2 3 Cheng X, Wang X, Nie K, Cheng L, Zhang Z, Hu Y, Peng W (2021). "Systematic Pan-Cancer Analysis Identifies TREM2 as an Immunological and Prognostic Biomarker". Frontiers in Immunology. 12: 646523. doi: 10.3389/fimmu.2021.646523 . PMC   7925850 . PMID   33679809.
  51. Binnewies M, Pollack JL, Rudolph J, Dash S, Abushawish M, Lee T, et al. (October 2021). "Targeting TREM2 on tumor-associated macrophages enhances immunotherapy". Cell Reports. 37 (3): 109844. doi: 10.1016/j.celrep.2021.109844 . PMID   34686340. S2CID   239472808.
  52. Obradovic A, Chowdhury N, Haake SM, Ager C, Wang V, Vlahos L, et al. (May 2021). "Single-cell protein activity analysis identifies recurrence-associated renal tumor macrophages". Cell. 184 (11): 2988–3005.e16. doi:10.1016/j.cell.2021.04.038. PMC   8479759 . PMID   34019793.
  53. Molgora M, Esaulova E, Vermi W, Hou J, Chen Y, Luo J, et al. (August 2020). "TREM2 Modulation Remodels the Tumor Myeloid Landscape Enhancing Anti-PD-1 Immunotherapy". Cell. 182 (4): 886–900.e17. doi:10.1016/j.cell.2020.07.013. PMC   7485282 . PMID   32783918.
  54. Stagg AJ (2018). "Intestinal Dendritic Cells in Health and Gut Inflammation". Frontiers in Immunology. 9: 2883. doi: 10.3389/fimmu.2018.02883 . PMC   6291504 . PMID   30574151.
  55. Coelho I, Duarte N, Macedo MP, Penha-Gonçalves C (March 2021). "Insights into Macrophage/Monocyte-Endothelial Cell Crosstalk in the Liver: A Role for Trem-2". Journal of Clinical Medicine. 10 (6): 1248. doi: 10.3390/jcm10061248 . PMC   8002813 . PMID   33802948.
  56. 1 2 3 Perugorria MJ, Esparza-Baquer A, Oakley F, Labiano I, Korosec A, Jais A, et al. (March 2019). "Non-parenchymal TREM-2 protects the liver from immune-mediated hepatocellular damage". Gut. 68 (3): 533–546. doi:10.1136/gutjnl-2017-314107. PMC   6580759 . PMID   29374630.
  57. Esparza-Baquer A, Labiano I, Sharif O, Agirre-Lizaso A, Oakley F, Rodrigues PM, et al. (July 2021). "TREM-2 defends the liver against hepatocellular carcinoma through multifactorial protective mechanisms". Gut. 70 (7): 1345–1361. doi:10.1136/gutjnl-2019-319227. PMC   8223629 . PMID   32907830.
  58. 1 2 3 Mecca C, Giambanco I, Donato R, Arcuri C (January 2018). "Microglia and Aging: The Role of the TREM2-DAP12 and CX3CL1-CX3CR1 Axes". International Journal of Molecular Sciences. 19 (1): E318. doi: 10.3390/ijms19010318 . PMC   5796261 . PMID   29361745.
  59. Paloneva J, Kestilä M, Wu J, Salminen A, Böhling T, Ruotsalainen V, et al. (July 2000). "Loss-of-function mutations in TYROBP (DAP12) result in a presenile dementia with bone cysts". Nature Genetics. 25 (3): 357–361. doi:10.1038/77153. PMID   10888890. S2CID   9243117.
  60. 1 2 Walter J (February 2016). "The Triggering Receptor Expressed on Myeloid Cells 2: A Molecular Link of Neuroinflammation and Neurodegenerative Diseases". The Journal of Biological Chemistry. 291 (9): 4334–4341. doi: 10.1074/jbc.R115.704981 . PMC   4813462 . PMID   26694609.
  61. Gervois P, Lambrichts I (2019). "The Emerging Role of Triggering Receptor Expressed on Myeloid Cells 2 as a Target for Immunomodulation in Ischemic Stroke". Frontiers in Immunology. 10: 1668. doi: 10.3389/fimmu.2019.01668 . PMC   6650572 . PMID   31379859.
  62. Ulrich JD, Holtzman DM (April 2016). "TREM2 Function in Alzheimer's Disease and Neurodegeneration". ACS Chemical Neuroscience. 7 (4): 420–427. doi:10.1021/acschemneuro.5b00313. PMID   26854967.
  63. Atagi Y, Liu CC, Painter MM, Chen XF, Verbeeck C, Zheng H, et al. (October 2015). "Apolipoprotein E Is a Ligand for Triggering Receptor Expressed on Myeloid Cells 2 (TREM2)". The Journal of Biological Chemistry. 290 (43): 26043–26050. doi: 10.1074/jbc.M115.679043 . PMC   4646257 . PMID   26374899.
  64. Guerreiro R, Hardy J (October 2013). "TREM2 and neurodegenerative disease". The New England Journal of Medicine. 369 (16): 1569–1570. doi:10.1056/NEJMc1306509. PMC   3980568 . PMID   24143816.