eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
1/2023
vol. 48
 
Share:
Share:
Review paper

Molecular regulatory mechanism of LILRB4 in the immune response

Haiyin Liu
1, 2, 3
,
Jun Yang
1, 2
,
Jing Zhang
2, 3
,
Peiyue Zhang
1, 2, 3
,
Mengting Zhang
1, 2, 3
,
Chaojun Yang
2, 3
,
Li Liu
2, 3
,
Cuiyuan Huang
3
,
Wei Wang
2, 3
,
Yuhong Zhai
1, 2, 3
,
Jian Yang
1, 2

1.
Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
2.
Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China
3.
HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
Cent Eur J Immunol 2023; 48 (1): 43-47
Online publish date: 2023/02/23
Article file
- Molecular regulatory.pdf  [0.14 MB]
Get citation
 
PlumX metrics:
 

Introduction

Immune response (IR) refers to the process whereby immune cells recognize, activate, proliferate, differentiate antigen molecules, produce immune substances and generate specific immune effects under stimulation of antigens. The immune response can be activated or inhibited in the body, which are balanced by substances ensuring immune responses to pathogens in the immune system, such as leukocyte immunoglobulin-like receptors (LILRs). Leukocyte immunoglobulin-like receptor B4 (LILRB4) belongs to the LILR family, which is widely distributed on immune cell membranes. LILRB4 is a kind of inhibitory receptor that plays a key role in immune checkpoint pathways and participates in regulating multiple immune diseases [1].

The structure of LILRB4

LILRB4 is a member of the leukocyte immunoglobulin-like receptor (LIR) family, which is found in a gene cluster in chromosomal region 19q13.4 [2, 3]. According to the role of intracellular motifs, immunoglobulin-like transcripts (ILTs) were divided into the activated receptor LILRA and the inhibitory receptor LILRB. The activating receptor LILRA includes six receptors, LILRA1-LILRA6, while the inhibitory receptor includes LILRB1-LILRB5 [4, 5]. Inhibitory LILR members have a long cytoplasmic tail containing different sets of immunoreceptor tyrosine-based inhibitory motifs that recruit phosphatases and thus contribute to downstream inhibitory signaling pathways [6, 7]. Activating members on the other hand have truncated cytoplasmic regions and are thought to associate with activating adaptor proteins via a positively charged amino acid in the transmembrane domain (e.g. LILRA2 and LILRA4 both associate with the γ chain of FcεRI) [8, 9]. LILRBs contain two or four extracellular immunoglobulin domains, a transmembrane domain, and two to four cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIMs) [10]. Interestingly, LILRB4 is somewhat unusual as whereas most family members contain four immunoglobulin-like domains in their extracellular region (designated D1, D2, D3, and D4) LILRB4 is one of two members that only possess two immunoglobulin-like domains (the other one is the closely related activating receptor LILRA5) [11].

The function of LILRB4

The immune system is regulated by LILRB4, which is produced on immune cells and may bind to MHC class I molecules on antigen-presenting cells as well as other ligands such as integrins αVβ3, apolipoprotein E, and fibronectin [12-15]. Furthermore, it can also function in antigen capture and presentation. Previous studies have indicated that ILT3-Fc acts through BCL6 and is a potent immunosuppressive agent for reversing the onset of allo- or possibly autoimmune attacks against pancreatic islets [16]. It is thought to control inflammatory responses and cytotoxicity, which is helpful to focus the immune response and limit autoreactivity. Chang et al. found that ILT3-Fc inhibits T cell activation and induces the generation of Ts targeting multiple inflammatory miRNA pathways [17]. Simultaneously, LILRB4 can regulate marginal zone B cells and antibody production [18]. In cardiomyocytes, LILRB4 can regulate apoptosis and mediate myocardial hypertrophy [19].

Regulators of LILRB4 expression

LILRB4 has been proven to play an important role in the immune system. There are three main substances that can regulate LILRB4 expression: immunosuppressants, immunomodulators and anti-inflammatory drugs (Fig. 1). Penna et al. detected a significant increase in the expression of ILT3 after treatment with 1,25(OH)2D3, and proved that 1,25(OH)2D3 can induce the production of regulatory T cells through ILT3-dependent and non-ILT3-dependent pathways, while ILT3-dependent pathways are necessary for the induction of CD4+Foxp3+ regulatory T cells [20] (Table 1). Rochat et al. found that vitamin D supplementation during pregnancy was associated with increased ILT3 gene expression in a prospective trial [21]. However, Waschbisch et al. found that interferon-β alone or in combination with vitamin D could induce upregulation of ILT3 in vitro [22]. Interferon treatment led to a significant increase in monocyte ILT3 in vitro, and dihydroxyvitamin D3 also enhanced ILT3 expression. Švajger et al. found similar synergies between interferon (IFN)-γ and 1,25(OH)2D3 [23]. These results suggest that vitamin D receptor (VDR) agonists, alone or in combination with other immunomodulators, could up-regulate LILRB4 and contribute to DC tolerance. Some members of the interferon family also upregulate LILRB4 expression. Inui et al. found interleukin (IL)-2 to be an effective inducer of B4+CD38+ cells, and IFN-α is the main inducer of B4+CD38+ cells. The IFN-α pathway is involved in the pathogenesis of systemic lupus erythematosus (SLE) and IFN-α induces LILRB4 expression through plasmacytoid dendritic cells (PDC) and monocytoid dendritic cells (MDCs) [24]. After interferon treatment for 48 h, ILT3 expression was increased on freshly isolated purified monocytes and on immature myeloid dendritic cells derived from blood monocytes of multiple sclerosis (MS) patients and controls [25]. When ILT3 was blocked by anti- ILT3 antibodies, mitogen-driven proliferation of CD4+ T cells was increased several times in IFN-β treated relapsing-remitting MS (RRMS) patients and healthy controls, which may be involved in the inhibition of T cell activation in vivo. Immunosuppressants can also up-regulate LILRB4 expression in kidney transplant patients with chronic allogeneic nephropathy [26]. The infiltration of renal tubule interstitium and the level of ILT3 in endothelial cells were significantly increased, leading to reduction of CD40 in BDCA2+ cells by rapamycin and increase of Treg number, as well as expansion of the CD8+CD28+ T cell population. It was found that the RNA and protein levels of ILT3 and ILT4 in NKL cells increased 12, 24 and 36 h after cyclosporin A (CsA) treatment, and the killing activity of NKL cells towards tumor cells decreased significantly after CsA injection (15 mg/l) for 36 h. Thus, CsA up-regulated the expression of ILT3 and ILT4 in NKL cells, and affected its killing effect on tumor cells with different human leukocyte antigen G (HLA-G) expression and NKL cell proliferation [27]. Svajger et al. found that resveratrol could induce ILT3 expression, resulting in differentiation of dendritic cells (DCs) derived from human peripheral blood mononuclear cells (PBMCs) [28]. HLA-G exerts its inhibitory functions via interaction with ILT2, ILT4, and KIR2DL4, which are differentially expressed by NK, T, and antigen-presenting cells. ILT2, ILT3, ILT4, and KIR2DL4 expression is up-regulated by HLA-G in antigen-presenting cells, NK cells, and T cells, which may not need antigenic costimulation, possibly before the immune response [29]. In addition to immunosuppressants, some anti-inflammatory drugs can also regulate LILRB4 expression. Buckland et al. found that aspirin can induce the production of tolerant DCs, which may be related to the significant expression of ILT-3. Aspirin inhibits nuclear factor κB (NF-κB) translocation to the nucleus and induces antigen-specific Foxp3 positive regulatory T cells. Another nonsteroidal anti-inflammatory drug, niflumic acid (NFA), also upregulates ILT3 expression and participates in the development of immune tolerance in human monocyte derived DCs [30]. Brenk et al. [31] isocytes from peripheral blood of healthy subjects and cultured them under normal (30 µM) and low (5 µM) Trp (DCs+Trp and DCslow-TRP) conditions. The expression of ILT3 and ILT4 increased significantly in the DCslow-TRP group. The addition of anti-ILT3 mab partially restored the stimulation activity of DCslow-Trp on T cells, but had no effect on the stimulation of DC+Trp on T cells. These findings suggest that DCs with low tryptophan can induce high expression of inhibitory receptors ILT3 and ILT4, and increase inhibitory CD4, CD25 and Foxp3T cells in an ILT3-dependent manner, and weaken the stimulation ability of CD4+ T cells, thus leading to the immune tolerance of DCs. Interleukin 10 could inhibit endothelial dependent T cell costimulation by up-regulating ILT3/4 in human vascular endothelial cells [32]. In summary, LILRB4 has a variety of ligands, the discovery of which will provide a new therapeutic target for LILRB4 related diseases.

Table 1

Regulators of LILRB4 expression

RegulatorsCell distributionEffect
1,25(OH)2D3 [20]Regulatory T cellsUpregulation
Interferon β [25]MonocyteUpregulation
Interleukin 2 [24]B4+CD38+ cellsUpregulation
Interferon α [24]B4+CD38+ cellsUpregulation
Rapamycin [26]CD8+CD28+ T cellUpregulation
Cyclosporin A [27]NKL cellUpregulation
Resveratrol [28]Dendritic cellsUpregulation
Human leukocyte antigen G [29]NK cells, T cellsUpregulation
Aspirin [30]Dendritic cellsUpregulation
Tryptophan [31]Dendritic cellsDownregulation
Interleukin 10 [32]Human vascular endothelial cellUpregulation
Fig. 1

Upstream and downstream structure diagram of LILRB4

/f/fulltexts/CEJOI/50171/CEJI-48-50171-g001_min.jpg

LILRB4/SHP-1 or SHP-2

LILRB4 has three ITIMs. The ITIMs of LILRB4 can recruit src homology 2 (SH2) domain-containing phosphatase 1 (SHP-1) and SHP-2 from the cytosol, leading to the activation of SHPs and the subsequent inhibition of various downstream signaling pathways [33]. Li et al. reported that ILT3 promotes tumor motility and angiogenesis via recruitment of SHP2/SHIP1 and subsequent activation of the ERK1/2 signaling pathway [34]. By interaction with its ligand ApoE, ILT3 induced tumor cell migration and invasion as well as tumor angiogenesis via activation of the SHP-2/SHIP1-ERK1/2 signaling pathway, which subsequently promoted EMT and the expression of VEGF-A, leading to non-small cell lung cancer (NSCLC) metastasis. Truong et al. demonstrated that the chicken LILRB4-5 genes activate the JAK/STAT signaling pathway, which plays a key role in multiple cytokine-induced responses [35]. In addition, LILRB4 can recruit SHP1 to inhibit TRAF6 ubiquitination and subsequently inactivate NF-κB and MAPK cascades [36]. Blocking TRAF6 ubiquitination to inactivate downstream MAPK and NF-κB signaling largely explained the inhibitory effect of LILRB4 on nonalcoholic fatty liver disease (NAFLD) progression. NF-κB plays a key role in the cellular inflammatory response and immune response. LILRB4 regulated NF-κB by recruiting SHP-1 or SHP-2 in turn affects atherosclerosis and myocardial hypertrophy [37, 38]. In leukemia, inhibition of NF-κB signaling reversed T cell suppression and reduced AML cell infiltration in a LILRB4-dependent manner. Urokinase-type plasminogen activator receptor (UPAR), an NF-κB target, is known to promote cancer invasion, metastasis, survival and angiogenesis [39]. The urokinase-type plasminogen activator (uPA) system is a biomarker and therapeutic target in human malignancies. It was found that ARG1 is a key downstream effector of LILRB4/NF-κB/uPAR signaling. LILRB4/SHP-2/NF-κB/uPAR-ARG1 could suppress immune activity and supports leukemia migration [40].

Relationship with disease

As an immune checkpoint, LILRB4 is of great significance in the treatment of autoimmune diseases [41]. Jensen et al. revealed that ILT3 loss of function polymorphism was associated with the increase of inflammatory cytokine level in SLE [42]. In addition to SLE, LILRB4 is also associated with a variety of immune diseases, such as Kawasaki disease [43] and allergic diseases [18]. LILRB4 is also closely related to diseases of other systems. By inhibiting NF-κB signaling pathway, LILRB4 can attenuate cardiac hypertrophy and atherosclerosis [44-46]. The maturation and migration of pulmonary DCs to lymph nodes in response to Ag and the innate immune stimulus is associated with upregulation of LILRB4. LILRB4 could attenuate the ability of DCs to elicit pathologic Th2 pulmonary inflammation [47]. Myeloid-derived suppressor cells (MDSC) accumulate in the tumor microenvironment (TME); they are engaged in tumor-associated immunosuppression, and govern tumor growth and metastasis. It was shown that LILRB4 can facilitate tumor invasion and migration by controlling MDSC and preventing the release of miRNAs from the miR-1 family, hence facilitating MDSC-mediated tumor metastasis [48]. ILT3 expressed on M-MDSCs can induce immunosuppression in cancer, and antagonism of ILT3 may be useful to reverse the immunosuppressive function of M-MDSCs and enhance the efficacy of immune checkpoint inhibitors [49]. Sharma et al. also revealed that LILRB4 strongly suppresses tumor immunity in TME [50]. The above evidence indicates that LILRB4 may be an effective target for cancer treatment. LILRB4 may also be a potential drug for the treatment of autoimmunity and graft rejection [51]. For example, LILRB4 is an effective immunomodulator for inhibiting allograft rejection in islet allograft [52]. Furthermore, it is presumed that LILRB4 also participates in regulating central nervous system immune surveillance. Recombinant human ILT3.Fc protein can bind to murine immune cells and further inhibit the release of proinflammatory cytokines, which suggests the feasibility of ILT3 in treatment of multiple sclerosis [53].

Conclusion and perspectives

LILRB4 was first found on the immune cell membrane and gradually came into people’s vision. Current studies prove that LILRB4 not only acts on tumors and immune diseases, but also diseases of multiple systems, which indicates that LILRB4 has great potential in disease treatment. The understanding of the LILRB4 extracellular ligand and downstream signal pathway can deepen the understanding of LILRB4 in physiology and pathology, and help LILRB4 become a new biomarker of disease. However, it is still necessary to study LILRB4 further, especially at the cellular and molecular levels.

Funding

This work was supported by National Natural Science Foundation of China (82070372, 82170418, 82271618), Hubei Province’s Outstanding Medical Academic Leader Program; Natural Science Foundation of Hubei Province (2022CFA015), Educational Commission of Hubei Province (D20221205); Science and Technology Innovation Platform of Hubei Province (2022DCC014); Key Research and Development Program of Hubei Province (2022BCE001).

Notes

[1] Conflicts of interest The authors declare no conflict of interest.

References

1 

Cella M, Dohring C, Samaridis J, et al. (1997): A novel inhibitory receptor (ILT3) expressed on monocytes, macrophages, and dendritic cells involved in antigen processing. J Exp Med 185: 1743-1751.

2 

Gleissner CA, Dengler TJ (2009): Induction of ILT expression on nonprofessional antigen presenting cells: Clinical applications. Hum Immunol 70: 357-359.

3 

Kabalak G, Dobberstein SB, Matthias T, et al. (2009): Association of immunoglobulin-like transcript 6 deficiency with Sjögren’s syndrome. Arthritis Rheum 60: 2923-2925.

4 

van der Touw W, Chen HM, Pan PY, et al. (2017): LILRB receptor-mediated regulation of myeloid cell maturation and function. Cancer Immunol Immunother 66: 1079-1087.

5 

Li Z, Deng M, Huang F, et al. (2020): LILRB4 ITIMs mediate the T cell suppression and infiltration of acute myeloid leukemia cells. Cell Mol Immunol 17: 272-282.

6 

Bellon T, Kitzig F, Sayos J, et al. (2002): Mutational analysis of immunoreceptor tyrosine-based inhibition motifs of the Ig-like transcript 2 (CD85j) leukocyte receptor. J Immunol 168: 3351-3359.

7 

Sayos J, Martinez-Barriocanal A, Kitzig F, et al. (2004): Recruitment of C-terminal Src kinase by the leukocyte inhibitory receptor CD85j. Biochem Biophys Res Commun 324: 640-647.

8 

Cao W, Rosen DB, Ito T, et al. (2006): Plasmacytoid dendritic cell-specific receptor ILT7-Fc epsilonRI gamma inhibits Toll-like receptor-induced interferon production. J Exp Med 203: 1399-1405.

9 

Nakajima H, Samaridis J, Angman L, et al. (1999): Human myeloid cells express an activating ILT receptor (ILT1) that associates with Fc receptor gamma-chain. J Immunol 162: 5-8.

10 

Park M, Liu RW, An H, et al. (2017): A dual positive and negative regulation of monocyte activation by leukocyte Ig-like receptor B4 depends on the position of the tyrosine residues in its ITIMs. Innate Immun 23: 381-391.

11 

Borges L, Kubin M, Kuhlman T (2003): LIR9, an immunoglobulin-superfamily-activating receptor, is expressed as a transmembrane and as a secreted molecule. Blood 101: 1484-1486.

12 

Cheng H, Mohammed F, Nam G, et al. (2011): Crystal structure of leukocyte Ig-like receptor LILRB4 (ILT3/LIR-5/CD85k): a myeloid inhibitory receptor involved in immune tolerance. J Biol Chem 286: 18013-18025.

13 

Su MT, Inui M, Wong YL, et al. (2021): Blockade of checkpoint ILT3/LILRB4/gp49B binding to fibronectin ameliorates autoimmune disease in BXSB/Yaa mice. Int Immunol 33: 447-458.

14 

Deng M, Gui X, Kim J, et al. (2018): LILRB4 signalling in leukaemia cells mediates T cell suppression and tumour infiltration. Nature 562: 605-609.

15 

Castells MC, Klickstein LB, Hassani K, et al. (2001): gp49B1-alpha(v)beta3 interaction inhibits antigen-induced mast cell activation. Nat Immunol 2: 436-442.

16 

Chang CC, Vlad G, D’Agati VD, et al. (2010): BCL6 is required for differentiation of Ig-like transcript 3-Fc-induced CD8+ T suppressor cells. J Immunol 185: 5714-5722.

17 

Chang CC, Zhang QY, Liu Z, et al. (2012): Downregulation of inflammatory microRNAs by Ig-like transcript 3 is essential for the differentiation of human CD8(+) T suppressor cells. J Immunol 188: 3042-3052.

18 

Fukao S, Haniuda K, Nojima T, et al. (2014): gp49B-mediated negative regulation of antibody production by memory and marginal zone B cells. J Immunol 193: 635-644.

19 

Li Q, Wei G, Tao T (2019): Leukocyte immunoglobulin-like receptor B4 (LILRB4) negatively mediates the pathological cardiac hypertrophy by suppressing fibrosis, inflammation and apoptosis via the activation of NF-κB signaling. Biochem Biophys Res Commun 509: 16-23.

20 

Penna G, Roncari A, Amuchastegui S, et al. (2005): Expression of the inhibitory receptor ILT3 on dendritic cells is dispensable for induction of CD4+Foxp3+ regulatory T cells by 1,25-dihydroxyvitamin D3. Blood 106: 3490-3497.

21 

Rochat MK, Ege MJ, Plabst D, et al. (2010): Maternal vitamin D intake during pregnancy increases gene expression of ILT3 and ILT4 in cord blood. Clin Exp Allergy 40: 786-794.

22 

Waschbisch A, Sanderson N, Krumbholz M, et al. (2014): Interferon beta and vitamin D synergize to induce immunoregulatory receptors on peripheral blood monocytes of multiple sclerosis patients. PLoS One 9: e115488.

23 

Švajger U, Rožman PJ (2019): Synergistic effects of interferon-γ and vitamin D3 signaling in induction of ILT-3highPDL-1high tolerogenic dendritic cells. Front Immunol 10: 2627.

24 

Inui M, Sugahara-Tobinai A, Fujii H, et al. (2016): Tolerogenic immunoreceptor ILT3/LILRB4 paradoxically marks pathogenic auto-antibody-producing plasmablasts and plasma cells in non-treated SLE. Int Immunol 28: 597-604.

25 

Jensen MA, Yanowitch RN, Reder AT, et al. (2010): Immunoglobulin-like transcript 3, an inhibitor of T cell activation, is reduced on blood monocytes during multiple sclerosis relapses and is induced by interferon beta-1b. Mult Scler 16: 30-38.

26 

Stallone G, Pontrelli P, Infante B, et al. (2014): Rapamycin induces ILT3(high)ILT4(high) dendritic cells promoting a new immunoregulatory pathway. Kidney Int 85: 888-897.

27 

Si YQ, Bian XK, Lu N, et al. (2012): Cyclosporine induces up-regulation of immunoglobulin-like transcripts 3 and 4 expression on and activity of NKL cells. Transplant Proc 44: 1407-1411.

28 

Svajger U, Obermajer N, Jeras M (2010): Dendritic cells treated with resveratrol during differentiation from monocytes gain substantial tolerogenic properties upon activation. Immunology 129: 525-535.

29 

Lemaoult J, Zafaranloo K, Le Danff C, et al. (2005): HLA-G up-regulates ILT2, ILT3, ILT4, and KIR2DL4 in antigen presenting cells, NK cells, and T cells. FASEB J 19: 662-664.

30 

Buckland M, Jago CB, Fazekasova H, et al. (2006): Aspirin-treated human DCs up-regulate ILT-3 and induce hyporesponsiveness and regulatory activity in responder T cells. Am J Transplant 6: 2046-2059.

31 

Brenk M, Scheler M, Koch S, et al. (2009): Tryptophan deprivation induces inhibitory receptors ILT3 and ILT4 on dendritic cells favoring the induction of human CD4+CD25+ Foxp3+ T regulatory cells. J Immunol 183: 145-154.

32 

Gleissner CA, Zastrow A, Klingenberg R, et al. (2007): IL-10 inhibits endothelium-dependent T cell costimulation by up-regulation of ILT3/4 in human vascular endothelial cells. Eur J Immunol 37: 177-192.

33 

Kuroiwa A, Yamashita Y, Inui M, et al. (1998): Association of tyrosine phosphatases SHP-1 and SHP-2, inositol 5-phosphatase SHIP with gp49B1, and chromosomal assignment of the gene. J Biol Chem 273: 1070-1074.

34 

Li J, Gao A, Zhang F, et al. (2021): ILT3 promotes tumor cell motility and angiogenesis in non-small cell lung cancer. Cancer Lett 501: 263-276.

35 

Truong AD, Hong Y, Tran H, et al. (2019): Characterization and functional analyses of novel chicken leukocyte immunoglobulin-like receptor subfamily B members 4 and 5. Poult Sci 98: 6989-7002.

36 

Lu Y, Jiang Z, Dai H, et al. (2018): Hepatic leukocyte immunoglobulin-like receptor B4 (LILRB4) attenuates nonalcoholic fatty liver disease via SHP1-TRAF6 pathway. Hepatology 67: 1303-1319.

37 

Jiang Z, Qin JJ, Zhang Y, et al. (2017): LILRB4 deficiency aggravates the development of atherosclerosis and plaque instability by increasing the macrophage inflammatory response via NF-κB signaling. Clin Sci (Lond) 131: 2275-2288.

38 

Zhou H, Li N, Yuan Y, et al. (2020): Leukocyte immunoglobulin-like receptor B4 protects against cardiac hypertrophy via SHP-2-dependent inhibition of the NF-κB pathway. J Mol Med (Berl) 98: 691-705.

39 

Wang Y, Dang J, Wang H, et al. (2000): Identification of a novel nuclear factor-kappaB sequence involved in expression of urokinase-type plasminogen activator receptor. Eur J Biochem 267: 3248-3254.

40 

Deng M, Gui X, Kim J, et al. (2018): LILRB4 signalling in leukaemia cells mediates T cell suppression and tumour infiltration. Nature 562: 605-609.

41 

Huang C, Zhu H X, Yao Y, et al. (2019): Immune checkpoint molecules. Possible future therapeutic implications in autoimmune diseases. J Autoimmun 104: 102333.

42 

Jensen MA, Patterson KC, Kumar AA, et al. (2013): Functional genetic polymorphisms in ILT3 are associated with decreased surface expression on dendritic cells and increased serum cytokines in lupus patients. Ann Rheum Dis 72: 596-601.

43 

Sugahara-Tobinai A, Inui M, Metoki T, et al. (2019): Augmented ILT3/LILRB4 expression of peripheral blood antibody secreting cells in the acute phase of Kawasaki disease. Pediatr Infect Dis J 38: 431-438.

44 

Zhou H, Li N, Yuan Y, et al. (2020): Leukocyte immunoglobulin-like receptor B4 protects against cardiac hypertrophy via SHP-2-dependent inhibition of the NF-κB pathway. J Mol Med (Berl) 98: 691-705.

45 

Li Q, Wei G, Tao T (2019): Leukocyte immunoglobulin-like receptor B4 (LILRB4) negatively mediates the pathological cardiac hypertrophy by suppressing fibrosis, inflammation and apoptosis via the activation of NF-κB signaling. Biochem Biophys Res Commun 509: 16-23.

46 

Jiang Z, Qin JJ, Zhang Y, et al. (2017): LILRB4 deficiency aggravates the development of atherosclerosis and plaque instability by increasing the macrophage inflammatory response via NF-kappaB signaling. Clin Sci (Lond) 131: 2275-2288.

47 

Breslow RG, Rao JJ, Xing W, et al. (2010): Inhibition of Th2 adaptive immune responses and pulmonary inflammation by leukocyte Ig-like receptor B4 on dendritic cells. J Immunol 184: 1003-1013.

48 

Su MT, Kumata S, Endo S, et al. (2022): LILRB4 promotes tumor metastasis by regulating MDSCs and inhibiting miR-1 family miRNAs. Oncoimmunology 11: 2060907.

49 

Singh L, Muise ES, Bhattacharya A, et al. (2021): ILT3 (LILRB4) Promotes the immunosuppressive function of tumor-educated human monocytic myeloid-derived suppressor cells. Mol Cancer Res 19: 702-716.

50 

Sharma N, Atolagbe OT, Ge Z, et al. (2021): LILRB4 suppresses immunity in solid tumors and is a potential target for immunotherapy. J Exp Med 218: e20201811.

51 

Vlad G, Suciu-Foca N (2012): Induction of antigen-specific human T suppressor cells by membrane and soluble ILT3. Exp Mol Pathol 93: 294-301.

52 

Vlad G, D’Agati VD, Zhang QY, et al. (2008): Immunoglobulin-like transcript 3-Fc suppresses T-cell responses to allogeneic human islet transplants in hu-NOD/SCID mice. Diabetes 57: 1878-1886.

53 

Xu Z, Lin CC, Ho S, et al. (2021): Suppression of experimental autoimmune encephalomyelitis by ILT3.Fc. J Immunol 206: 554-565.

Copyright: © 2023 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.