Open access peer-reviewed chapter

Toll-Like Receptors and Natural Killer Cells

Written By

Carmen Maldonado-Bernal and David Sánchez-Herrera

Submitted: 20 September 2018 Reviewed: 17 April 2019 Published: 31 May 2019

DOI: 10.5772/intechopen.86393

From the Edited Volume

Toll-like Receptors

Edited by Nima Rezaei

Chapter metrics overview

1,462 Chapter Downloads

View Full Metrics

Abstract

Natural killer (NK) cells represent a heterogeneous subpopulation of lymphocytes of the innate immune system with a powerful antitumor activity, a function given by a complex collection of receptors. They act synergistically to recognize, regulate, or amplify the response according to the microenvironment, thus highlighting Toll-like receptors (TLRs), a type of receptors that allows sensing evolutionarily molecules conserved of pathogens known as pathogen-associated molecular patterns (PAMPs) and/or damage-associated molecular patterns (DAMPs). Those TLRs are essential to start the immune response. There is little information about the different subpopulations that form NK cells as well as their expression profile of innate immune response receptors in hematological cancers.

Keywords

  • Toll-like receptors
  • natural killer cells
  • innate immunity
  • pathogen-associated molecular patterns
  • damage-associated molecular patterns

1. Introduction

Natural killer (NK) cells represent a highly specialized subpopulation of lymphocytes that are part of the innate immune system, whose functions vary according to the microenvironment. NK cells are involved in the early defense against foreign cells or own cells subjected to some stress (bacterial infection, viral or tumor transformation) through the recruitment of neutrophils, macrophages, dendritic cells, or B/T lymphocytes. They induce an effective adaptive response; regulate, directly or indirectly, the activity of antigen-presenting cells (APCs); and activate T lymphocytes through the natural cytotoxic activity that characterizes them or through the production of cytokines and chemokines that generate an inflammatory environment [1, 2].

NK cells play an important role in the surveillance and suppression of tumor cells; despite the significant advances that have been reached in the last decades, it is still unknown if there is a direct relationship among the population dynamics, functionality, and the phenotype of these cells. Its role in the establishment and development of malignant hematological disorders such as acute lymphoblastic leukemia (ALL), a disease characterized by the uncontrolled proliferation of B or T lymphoid precursors, is still unknown.

Advertisement

2. Overview of natural killer cells and toll-like receptors

2.1 NK cells

NK cells, although they are larger and present granules in their cytoplasm, morphologically are indistinguishable from the other lymphocytes. According to different authors, they comprise from 5 to 15% [3], 20% [2, 4], or even 25% [5] of total peripheral blood mononuclear cells and are derived from a CD34+ hematopoietic progenitor, as are dendritic cells (DC) and B and T lymphocytes [6].

NK cells are phenotypically defined by the expression of CD56 (neural cell adhesion molecule (NCAM) and CD16a (also known as Fcγ-RIIIA), but not CD3 and CD19, which are molecules of T and B lymphocytes, respectively [7, 8]. For a long time, they were considered as the only population of non-B or T lymphocytes. It is currently accepted that NK cells are within a subgroup of the so-called innate lymphoid cells (ILCs), whose subpopulations are differentiated according to the immunophenotype, the profile of cytokines they produce, and the transcription factors they possess [9, 10].

The NK cells were classified in group 1 of the ILCs (ILC1) due to their ability to produce INF-γ, but not cytokines such as IL-4, IL-5, IL-9, IL-13, IL-17, or IL-22, characteristic of the ILC2 and ILC3 groups, respectively [9, 11, 12]. Even within the same subgroup, they differ by having cytotoxic capacity and selectively expressing the eomesodermin (EOMES) transcription factor of other ILC non-NK that also produce INF-γ [13].

NK cells maintain a pro-inflammatory environment through the release of cytokines and recruit cells of the immune system to combat infectious agents through the release of chemokines [14] and regulate the activity of dendritic cells or activated lymphocytes [1]; they are in charge of antitumor surveillance and tolerance to healthy own cells, which conditions the rejection of transplants [15] among other functions.

In recent years, it has been reported that in addition to NK cytotoxic or regulatory NK cells, there are memory NK cells [16, 17] and NK cooperators, which secrete Th1-type cytokines (NK1) and Th2 (NK2) [18]. Even, NK cells are similar to antigen-presenting cells [19, 20], although this is in controversy.

2.2 Population diversification of NK cells and their role in the immune response

According to what was compiled by Huntington et al., NK cell precursors (NKPs) originate mainly in the bone marrow from hematopoietic precursor cells (HSCs), although they can also do so in organs such as the thymus from early lymphoid progenitor (ELP). These NKPs can mature into competent NK cells in the bone marrow or other organs, such as the liver, spleen, thymus, and lymphoid nodules, to finally enter to the circulation [21]. They migrate to other sites such as the lung, liver, mucous membranes and skin, uterus, pancreas, joints, and central nervous system (CNS), where they can exhibit unique characteristics ranging from the increase or decrease of the expression of activation receptors or effector cytotoxic molecules to the modulation of resident immune cells (Figure 1) [22].

Figure 1.

Origin and distribution of NK cells in humans. The precursors of NK cell precursor (NKP) can originate from hematopoietic progenitor cells (HSC) in the bone marrow or from early lymphoid progenitor (ELP). NK cells mature mainly in the bone marrow, although the immature NKP and NK (iNK) can recirculate among the liver, spleen, and lymphoid nodes as alternative maturation sites. Mature NK cells (mNK) that leave the bone marrow reach different organs through blood circulation where they reside and modify their phenotypic and functional characteristics.

According to the expression of CD56 (NCAM), NK cells can be divided into two subpopulations: NKDim and NKBright. However, according to the relative expression of the CD56 and CD16 (FcγRIIIa) markers, their functionality, and their distribution in peripheral blood or lymphoid organs, it is possible to differentiate five subpopulations of mature NK cells (Figure 2).

Figure 2.

Subpopulations of NK cells in peripheral blood based on the relative expression of CD56 and CD16. (1) CD56Bright CD16Neg, recognized for its immunoregulatory activity, represents between 50 and 70% of the CD56Bright population; (2) CD56Bright CD16Dim represent between 30 and 50% of the CD56Bright population; (3) CD56Dim CD16Neg; (4) CD56Dim CD16Bright is recognized for its cytotoxic activity; and (5) CD56Neg CD16Bright whose function is still unknown. Modified from [23].

The NKBright populations, also known as NK regulators, comprise about 10% of total peripheral blood NK cells. From these, about 50–70% have a [CD56High CD16Neg] phenotype and 30–50% a [CD56High/CD16Low] phenotype [23]; they are mainly characterized by their poor cytotoxic capacity and their high capacity to secrete several types of post-activation cytokines, mainly INF-γ but also TNF-β, IL-5, IL-10, and IL-13 [7, 8] and constitutively some chemokines such as MIP-1α, MIP-1β, and RANTES [24]. They also have the ability to proliferate in culture when exposed to low doses of IL-2 (picomoles) compared to the cytotoxic NK cells, which does not show an evident proliferation under the same conditions [25]. The NKBright cells [CD56High CD16Neg] are assigned an exclusively regulatory function, since their cytotoxic activity is poor and their genetic profile is directed mainly to the production of cytokines and not to the cytotoxic activity, in comparison with the NKDim, whereas the NKBright subpopulation [CD56High CD16Low] is considered more as a transition phenotype [26]; because despite having the same characteristics of the previous phenotype, it has a lower rate of cell division when stimulated with IL-2 and does not change its activity even with ligands for c-KIT [27]. It exhibits cytotoxic activity [28], and it has also been seen that it represents the highest percentage of NK cells in circulation in bone marrow transplants, until its normalization around the fourth month [29, 30]. NKBright cells are usually not found in peripheral blood, bone marrow, and spleen as they are mainly distributed in secondary lymphoid nodules (parafollicular zone of T cells) [31] and tonsils [32].

On the other hand, the NKDim population represents around 90% of peripheral blood NK cells; they have a phenotype [CD56Low CD16Neg] whose main function has not been well established [23] and a main phenotype [CD56Low CD16High] that exhibits potent cytotoxic activity. Although they are generally poor producers of cytokines [7, 8, 33], they tend to predominate in the spleen, peripheral blood, and bone marrow [32, 34].

It is important to clarify that the behavior of each subpopulation, in terms of post-activation secretion of cytokines and chemokines, will depend largely on the stimuli they receive either by recognizing target cells (tumor or transformed) or responding to exogenous cytokines.

Compared with NKDim cells, NKBright cells produce more TNF-α and INF-γ after activation with PMA/ionomycin [35] or exogenous cytokines such as IL-12, IL-15, or IL-18, alone or in combination [8].

On the contrary, when it comes to a response to the recognition of target cells, NKDim cells significantly increase their production of cytokines and chemokines compared to NKBright, such as MCP-1 (CCL2), IL-8 (CXCL8), IP-10 (CXCL10), soluble IL-2Rα (CD25), GM-CSF, and IL-5 and low levels of IL-1β, IL-6, IL-7, IL-10, IL-12p40, IFN-α, and MIG (CXCL9). In addition, it increases the production of chemokines such as MIP-1α (CCL3), MIP-1β (CCL4), and RANTES (CCL5) that produce constitutively [24].

The cytotoxic activity exhibited by NK cells does not require prior sensitization to kill their target cells, since it is not dependent on the presentation of a specific antigen as in the case of CD8+ T cells [7, 36] and can be mediated through membranolitic and/or apoptotic mechanisms (Figure 3).

Figure 3.

Recognition and elimination of abnormal cells by NK cells. NK cells possess the ability to discriminate normal cells of tumor or transformed cells by detecting alterations at the HLA-I level; the target cells are eliminated by membranolitic and/or apoptotic mechanisms.

The membranolitic mechanisms include the production of perforins, enzymes that when integrated into the cell membrane form a pore that allows water to enter and cause osmotic lysis [37]. In the past, it was believed that both NKBright and NKDim cells had similar levels of perforins [38]; however, more recent studies by flow cytometry indicate that NKDim cells have at least 10 times more perforins than NKBright ones [35].

Regarding apoptotic mechanisms, these can induce the death of the target cell through complex mechanisms that involve both death-inducing proteins and specific ligand-receptor interactions through one of the following routes:

2.3 Granzyme pathway

Granzymes are proteins capable of activating the apoptosis program [39] following two mechanisms. The first does not depend on the activity of caspase proteins and is mediated mainly by granzyme A, and this fraction is single-stranded DNA (ssDNA) and interferes in the repair of genetic material without producing cell lysis [40, 41], while the second promotes the activity of caspase proteins and is mediated mainly by granzyme B [42].

The NK cell presents granzymes A, B, K, and M; NKDim cells present a high expression of granzymes A and B, whereas NKBright cells mainly express granzyme K [35, 43]. There are reports that NK cells express almost exclusively granzyme M; this enzyme is capable of mediating cell death independent of the activation of caspase proteins and in the presence of perforins, without fractionating DNA or producing changes in mitochondria [44].

In mice, deficient in granzymes A/B and/or perforins, it has been seen that there is uncontrolled growth of solid tumors, which suggests that these enzymes play an important role in the immunosurveillance of tumor cells mediated by NK cells [45].

2.4 NK cell ligand binding pathway to the cell death receptors expressed by the target cell

They include Fas ligands [FasL (CD95L)-Fas (CD95)] and/or the ligand that induces apoptosis related to tumor necrosis factor α (TRAIL) [46, 47].

2.5 Antibody-dependent cellular cytotoxicity (ADCC)

NK cells express FcγRIIC/CD32c [48] and FcγRIIIA/CD16a [34]. These receptors interact with opsonized target cells, through the Fc regions of the antibodies, which combined with cellular antigens that cause the death of the target cell [49]. To through of mechanisms that involve the release of cytotoxic granules (perforin-granzyme), or by stimulation of apoptosis through of TNF-related apoptosis-inducing ligand (TRAIL) and/or by release of pro-inflammatory cytokines that promote the activity of other cells [50].

NKDim cells with [CD56Low CD16High] phenotype direct this mechanism in comparison with NKBright cells. Although it has been seen that the subpopulation with [CD56High CD16Low] phenotype exhibits low cytotoxic activity [51], NKDim cells [CD56Low CD16Neg] show a higher antitumor activity against cell lines (natural cytotoxicity) than other subpopulations [52]. This is supported by other studies where it is reported that NKDim cells [CD56Low CD16High] lose the expression of CD16 and increase the expression of CD107a (a degranulation marker), through a disintegrin and a metalloprotease-17 (ADAM-17), to become [CD56Low CD16Neg] with high cytotoxic capacity [53].

The role of the [CD56Neg CD16High] subpopulation is still not clearly defined. It is known that it is found in a low frequency in healthy individuals. It does not express surface molecules of other lymphoid lineages and that in chronic viral diseases, such as the human immunodeficiency virus (HIV). It presents changes in the level of expression of their activity receptors, characterized by the increase in the expression of inhibitory receptors and the decrease of natural cytotoxicity receptors (NCRs), together with other effector molecules that are hardly observed in healthy people [54, 55, 56].

It is considered that the [CD56Neg CD16High] subpopulation is dysfunctional in terms of its lytic and antiviral activity, although it retains the ability to produce pro-inflammatory chemokines [54, 55, 56].

Zulu et al. demonstrated that the HIV induces the expansion of the negative CD56 population of NK cells through the upregulation of NKG2C receptors and the negative regulation of Siglec-7, NKG2A, and CD57 receptors [57].

2.6 Receptors of the NK cells

NK cells have signals through a wide variety of receptors that allow them to respond to different types of stimuli and grant great flexibility when exercising their effector and/or cytotoxic function.

The function of the NK cell is given by a complex collection of receptors that act in a synergistic way to recognize, regulate, or amplify the response according to the microenvironment. Thus highlighting the pattern recognition receptors (PRRs), such as Toll-like receptors or natural cytotoxicity receptors, and inhibitory killing receptors (iNKRs), such as receptors that are activated during early response to pathogens, cells transformed by virus or tumor cells [58].

PRRs are a family of innate immune response receptors that recognize evolutionarily conserved microbial products whose activation favors the production of pro-inflammatory cytokines. Within the PRR group, the TLRs are the most studied, although they are not the only ones; there are also the NOD-like receptors (NLRs) and the retinoid acid-inducible gene I (RIG-I)-like receptors (RLRs) [59].

NK cells express innate immune response receptors, such as NOD2, NLRP3, TLR3, TLR7, and TLR9, and promote the production of inflammatory cytokines and chemokines that are capable of amplifying the immune response [60]. The modulation of these cells through their innate immune response receptors, mainly via TLR, has gained interest and represents a promising therapeutic alternative against conditions such as cancer. Since there have been studies for a long time that support the possibility of its use, it has been observed that when ODNs (ligands of TLR9) are intraperitoneally administered in lymphoma murine models, an effective elimination of tumor cells occurs in 80% of cases [61].

2.7 Toll-like receptors and their role in NK cells

Toll-like receptors are among the most important group of pattern recognition receptors, since they orchestrate a wide variety of activities related to the immune response.

These receptors recognize a wide variety of molecules evolutionarily conserved, associated with microorganisms, such as lipopolysaccharides, lipoproteins, mycolic acids, non-methylated DNA, and double-stranded RNA, generically known as pathogen-associated molecular patterns [62, 63, 64]. TLRs also recognize endogenous molecules called damage-associated molecular patterns, which originate from damaged cells [65] or are products of altered metabolism of transformed cells in conditions such as cancer [66, 67] and autoimmune diseases [67, 68, 69, 70] or associated with chronic inflammation [71, 72]. They play an important role in the evolution of these conditions.

2.8 Overview of the toll-like receptors

Structurally, TLRs are type I integral glycoproteins that present an extracellular domain with leucine-rich repeats (LRRs) that are responsible for binding and discriminating ligands (PAMPs or DAMPS) present in the cellular microenvironment. They have a transmembrane domain and an intracellular Toll/interleukin (IL)-1 receptor (TIR) domain that triggers the signaling cascade via MyD88/TRIF and is highly conserved among each subfamily of TLRs [73].

There are 13 TLRs described in mammals, and 10 are found at the protein level in humans and differ according to their cellular localization and to the different PAMPs/DAMPs to which they respond. TLR11 in humans is a pseudogene, so it is not expressed [74].

The TLRs that are found mainly in the cell membrane are TLRs 1, 2, 4, 5, and 6. They sense structural components of bacteria, fungi, helminthes, or protozoa, whereas TLRs that are mainly found in intracellular compartments, such as TLRs 3, 7, 8, and 9, sense nucleic acids of viral and/or bacterial origin [73, 75] (Figure 4).

Figure 4.

Toll-like receptors and their ligands. TLRs are transmembrane proteins of a glycoprotein nature that possess the ability to sense highly conserved microorganism molecules known as pathogen-associated molecular patterns (PAMPs), such as flagellin, LPS, or genetic material (ssDNA, ssRNA, dsDNA, dsRNA). In humans, 10 of the 13 TLRs present in mammals have been detected [84].

The stimulation of the TLRs is capable of initiating an immune response to various stimuli on its own, as well as of controlling the adaptive response through the inflammatory process with the production of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α), chemokines (IL-8, MCP-1) [76]; defensins [77]; type I interferons [78]; co-stimulation and MHC molecules [79]. The union between the different types of immune responses through the TLRs takes as a classic example the dendritic cells. They inspect their environment through the TLRs [80]; and once they detect a ligand (bacterial product, viral or stress protein), increase the expression of co-stimulatory molecules capable of stimulating T naive cells [81] and polarizes the adaptive immune response toward Th1 or Th2 profiles [82].

The cellular response through direct stimulation with TLR ligands will depend largely on the type of lineage in question. The information that is reported about activity and expression in NK cells is relatively new, and it is more associated with innate antibacterial or antiviral immune response [83], but not in cancer.

2.9 Expression of TLRs in NK cells

The expression profile of TLRs in NK cells was initially limited to the detection of mRNA. However, the results do not always reflect the expression of the protein since it is difficult to identify the receptors in the NK cell.

NK cells express most of the human TLRs reported to date, although the detection and level of mRNA expression of each receptor vary depending on the author. There are authors who indicate that NK cells express high levels of TLR1, followed by TLR2, TLR3, TLR5, TLR6, and low levels of TLR4 [85, 86]. Other authors agree that TLR2 and TLR3 have greater expression, followed by TLR5 and TLR6; in those studies, TLR1 mRNA was not quantified [87, 88, 89]. On the other hand, TLR7 [85, 89, 90] and TLR10 [83, 86] present levels so low that they are practically undetectable.

There is a controversy about whether or not NK cells express TLR8 [86, 87, 89] and TLR9 [86, 88], although some authors point out that the cells constitutively express mRNA of all TLRs [85, 91, 92].

NK cells have higher levels of TLR3 mRNA than any other peripheral blood mononuclear cell, such as monocytes, B and T lymphocytes, or plasmacytoid dendritic cells [85].

Through techniques such as flow cytometry, Western blot, and immunoprecipitation, it is possible to know that NK cells of healthy people have a defined TLR expression profile (Table 1) and the expression of receptors is independent of their activation state [58].

TLR mRNA1 Protein Detection method
Presence
1 Very high Yes Flow cytometry [97]
Western blot [97]
2 High/moderate Yes3 Direct activation of the TLR2-/MyD88-dependent pathway [96]
Flow cytometry [95, 96, 97]
Western blot
3 High/moderate Yes Flow cytometry [93, 94]
Western blot [94]
4 Low Yes4 Flow cytometry [94, 95, 99]
5 High/moderate Yes5 S/R
6 High/moderate Yes Flow cytometry and Western blot [97]
7 Very low/undetectable Yes Flow cytometry [93, 94]
Western blot [94]
8 Low2 Yes Flow cytometry [94]
Western blot [90, 94]
9 Low2 Yes Flow cytometry [93, 95, 100]
Western blot [100]
10 Very low/undetectable N/R N/R

Table 1.

Expression of TLRs in human natural killer cells.

The levels of relative expression are given according to what was reported by [85, 91] and refer to the comparison of expression among the 10 TLRs.


There is a controversy whether or not they express mRNA of these TLRs, since some reports indicate that it was not possible to detect it.


In previous studies, TLR2 could not be detected by flow cytometry or by immunoprecipitation.


More recent studies indicate that it is expressed mainly as intracellular [95, 99].


No reports were found indicating the presence of TLR5; however it is inferred that it is present as it responds specifically to flagellin [88, 101, 102], a molecule that it is only recognized through this receptor.


N/R, not reported.

In addition, there are variations in the level of TLR expression within the same subpopulations of NK cells. It is accepted that both NKBright and NKDim exhibit a similar mRNA profile of TLRs, although it is not always reflected at the protein level and there is a great controversy regarding the distribution and presence of some of these receptors in both subpopulations, especially TLR2, TLR4, and TLR3.

TLR2 and TLR4 are mainly distributed on the cell surface, whereas TLR3 is generally found in intracellular vesicles [75]; however, it has been seen that in NK cells, TLR3 is expressed both within [93] and on the cell surface [94]. There are publications reporting that TLR2 and TLR4 exhibit a marked intracellular distribution [95], although other authors indicate otherwise [96, 97].

The relative amount of some TLRs may vary according to the phenotype (Dim or Bright), although expression levels appear to be higher in cells with regulatory phenotype [89], which suggests that the type of response could be conditioned to promote a cytotoxic or immunomodulatory response when using one ligand or another in TLR activation assays (Table 2).

TLR Cellular localization Population distribution
1 Extracellular [97] NKBright > NKDim [97]
2 Extracellular [96, 97] and intracellular [95] NKBright > NKDim [97]
3 Intracellular [93] and extracellular1 [94] N/R
4 Extracellular [94] and intracellular2 [95, 99] NKBright < NKDim [99]
NKBright = NKDim [95]
5 N/R N/R
6 Extracellular [97] NKBright > NKDim [97]
7 Intracellular [93, 94] N/R
8 Intracellular [94] N/R4
9 Mainly intracellular3 [93, 95, 100] NKBright = NKDim5 [95]
10 N/R N/R

Table 2.

Localization and differential distribution of TLRs in human natural killer cells.

It was found that both, in cell lines (NKL, NK92, and YT) and in NK of peripheral blood, TLR3 is expressed on the surface [94].


Studies that are more recent indicate that it is mainly expressed as intracellular [95, 99].


TLR9 expression exists in plasma membrane, but it is quite low compared to intracellular expression.


There are no studies that determine whether there is differential expression, although it has been seen that NKBright cells are better activated with ssRNA40 than NKDim cells, suggesting that the latter have a lower expression of TLR8.


In other studies, it seems that NKDim cells express more TLR9 than NKBright cells and its expression conditions the response to ligands of this TLR [100].


N/R, not reported.

It has been seen that NKBright cells express more TLR1, TLR2, and TLR6 than NKDim [97], although other studies report that less than 1% of total NK cells express these three receptors [98].

NKDim cells can express, under normal conditions, more TLR4 than NKBright cells [99] although other authors seem to find no differences in the expression in TLR2, TLR4 [95], and TLR9 [95, 100].

There is no information about whether there is differential expression of TLRs 3, 5, 7, or 8, and the distribution pattern of TLR5 is not known. However, it is inferred that NK cells express it, since they respond to flagellin and there are several studies that demonstrate it [88, 101, 102]. To date there are no reports about the presence, distribution, or role of TLR10 in NK cells.

The therapeutic use of TLR ligands in the modulation of NK cells against cancer, especially in malignant hematological disorders such as leukemia, is an interesting alternative for the treatment of this type of diseases, since there are reports that reveal their therapeutic use as potential antitumor agents and as adjuvants in vaccines and other therapeutic modalities [103]. It is currently the subject of an extensive review by several research groups [104, 105].

Advertisement

3. Conclusion

In this chapter, we included the overview of NK cells, their population diversification and role in the immune response, and their expression and role of TLRs.

Advertisement

Conflict of interest

The authors declare that there is no conflict of interest.

Advertisement

Acknowledgment

We thank doctors Jacques Zimmer, MD, PhD, and Sankar Ghosh, PhD, for allowing us to use some of their figures, Figures 2 and 4, in this chapter of the book.

References

  1. 1. LongEO KHS, Liu D, Peterson ME, Rajagopalan S. Controlling NK cell responses: Integration of signals for activation and inhibition. Annual Review of Immunology. 2013;31:227-258. DOI: 10.1146/annurev-immunol-020711-075005
  2. 2. Campbell KS, Hasegawa J. Natural killer cell biology: An update and future directions. The Journal of Allergy and Clinical Immunology. 2013;132(3):536-544. DOI: 10.1016/j.jaci.2013.07.006
  3. 3. Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer-cell subsets. Trends in Immunology. 2001;22(11):633-640
  4. 4. Rojas-Pandales F, Bolaños N, Mercado M, Gonzále JM, Cuéllar A, Cifuentes-Rojas C. Reference values of the natural killer cells (NK and NKT) on blood donors in Bogota. Acta médica colombiana. 2007;32(3):124-128
  5. 5. Pittari G, Filippini P, Gentilcore G, Grivel JC, Rutella S. Revving up natural killer cells and cytokine-induced killer cells against hematological malignancies. Frontiers in Immunology. 2015;6:230. DOI: 10.3389/fimmu.2015.00230
  6. 6. Galy A, Travis M, Cen D, Chen B. Human T, B, natural killer, and dendritic cells arise from a common bone marrow progenitor cell subset. Immunity. 1995;3(4):459-473
  7. 7. Caliguri MA. Human natural killer cells. Blood. 2008;112(3):461-469. DOI: 10.1182/blood-2007-09-077438
  8. 8. Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, Ghayur T, et al. Human natural killer cells: A unique innate immunoregulatory role for the CD56 bright subset. Blood. 2001;97:3146-3151
  9. 9. Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells—How did we miss them? Nature Reviews Immunology. 2013;13:75. DOI: 10.1038/nri3349
  10. 10. Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. Innate lymphoid cells—A proposal for uniform nomenclature. Nature Reviews Immunology. 2013;13:145-149. DOI: 10.1038/nri3365
  11. 11. Artis D, Spits H. The biology of innate lymphoid cells. Nature. 2015;517(7534):293-301. DOI: 10.1038/nature14189
  12. 12. Hazenberg MD, Spits H. Human innate lymphoid cells. Blood. 2014;124(5):700-709. DOI: 10.1182/blood-2013-11-427781
  13. 13. Gordon SM, Chaix J, Rupp LJ, Wu J, Madera S, Sun JC, et al. The transcription factors T-bet and Eomes control key checkpoints of natural killer cell maturation. Immunity. 2012;36(1):55-67. DOI: 10.1016/j.immuni.2011.11.016
  14. 14. Souza-Fonseca-Guimaraes F, Adib-Conquy M, Cavaillon JM. Natural killer (NK) cells in antibacterial innate immunity: Angels or devils? Molecular Medicine. 2012;18(1):270-285. DOI: 10.2119/molmed.2011.00201
  15. 15. Benichou G, Yamada Y, Aoyama A, Madsen JC. Natural killer cells in rejection and tolerance of solid organ allografts. Current Opinion in Organ Transplantation. 2011;16(1):47. DOI: 10.1097/MOT.0b013e32834254cf
  16. 16. Sun JC, Beilke JN, Lanier LL. Adaptive immune features of natural killer cells. Nature. 2009;457(7229):557. DOI: 10.1038/nature07665
  17. 17. O’Sullivan TE, Sun JC, Lanier LL. Natural killer cell memory. Immunity. 2015;43(4):634-645
  18. 18. Kimura MY, Nakayama T. Differentiation of NK1 and NK2 cells. Critical Reviews in Immunology. 2005;25(5):361-374. PMID: 16167886
  19. 19. Spits H, Lanier LL. Natural killer or dendritic: What’s in a name? Immunity. 2007;26(1):11-16
  20. 20. Hanna J, Mandelboim O. Novel APC-like properties of human NK cells directly regulate T cell activation. The Journal of Clinical Investigation. 2015;125(4):1763. DOI: 10.1172/JCI81527
  21. 21. Huntington ND, Vosshenrich CA, Di Santo JP. Developmental pathways that generate natural-killer-cell diversity in mice and humans. Nature Reviews Immunology. 2007;7(9):703-714. DOI: 10.1038/nri2154
  22. 22. Shi FD, Ljunggren HG, La Cava A, Van Kaer L. Organ-specific features of natural killer cells. Nature Reviews Immunology. 2011;11(10):658
  23. 23. Poli A, Michel T, Thérésine M, Andrès E, Hentges F, Zimmer J. CD56bright natural killer (NK) cells: An important NK cell subset. Immunology. 2009;126(4):458-465. DOI: 10.1111/j.1365-2567.2008.03027.x
  24. 24. Fauriat C, Long EO, Ljunggren HG, Bryceson YT. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood. 2010;115(11):2167-2176. DOI: 10.1182/blood-2009-08-238469
  25. 25. Carson WE, Giri JG, Lindemann M, Linett ML, Ahdieh M, Paxton R, et al. Interleukin (IL) 15 is a novel cytokine that activates human natural killer cells via components of the IL-2 receptor. The Journal of Experimental Medicine. 1994;180(4):1395-1403
  26. 26. Béziat V, Duffy D, Quoc SN, Le Garff-Tavernier M, Decocq J, Combadière B, et al. CD56brightCD16+ NK cells: A functional intermediate stage of NK cell differentiation. Journal of Immunology. 2011;186(12):6753-6761. DOI: 10.4049/jimmunol.1100330
  27. 27. Carson WE, Fehniger TA, Caligiuri MA. CD56 bright natural killer cell subsets: Characterization of distinct functional responses to interleukin-2 and the c-kit ligand. European Journal of Immunology. 1997;27(2):354-360. DOI: 10.1002/eji.1830270203
  28. 28. Takahashi E, Kuranaga N, Satoh K, Habu Y, Shinomiya N, Asano T, et al. Induction of CD16+ CD56bright NK cells with antitumour cytotoxicity not only from CD16− CD56bright NK cells but also from CD16− CD56dim NK cells. Scandinavian Journal of Immunology. 2007;65(2):126-138. DOI: 10.1111/j.1365-3083.2006.01883.x
  29. 29. Gottschalk LR, Bray RA, Kaizer H, Gebel HM. Two populations of CD56 (Leu-19)+/CD16+ cells in bone marrow transplant recipients. Bone Marrow Transplantation. 1990;5(4):259-264
  30. 30. Jacobs R, Stoll M, Stratmann G, Leo R, Link H, Schmidt RE. CD16-CD56+ natural killer cells after bone marrow transplantation. Blood. 1992;79(12):3239-3244
  31. 31. Fehniger TA, Cooper MA, Nuovo GJ, Cella M, Facchetti F, Colonna M, et al. CD56 bright natural killer cells are present in human lymph nodes and are activated by T cell–derived IL-2: A potential new link between adaptive and innate immunity. Blood. 2003;101(8):3052-3057. DOI: 10.1182/blood-2002-09-2876
  32. 32. Ferlazzo G, Thomas D, Lin SL, Goodman K, Morandi B, Muller WA, et al. The abundant NK cells in human secondary lymphoid tissues require activation to express killer cell Ig-like receptors and become cytolytic. Journal of Immunology. 2004;172(3):1455-1462. DOI: 10.4049/jimmunol.172.3.1455
  33. 33. Lanier LL, Ruitenberg JJ, Phillips JH. Functional and biochemical analysis of CD16 antigen on natural killer cells and granulocytes. Journal of Immunology. 1988;141(10):3478-3485
  34. 34. Yokoyama WM, Kim S, French AR. The dynamic life of natural killer cells. Annual Review of Immunology. 2004;22:405-429. DOI: 10.1146/annurev.immunol.22.012703.104711
  35. 35. Jacobs R, Hintzen G, Kemper A, Beul K, Kempf S, Behrens G, et al. CD56bright cells differ in their KIR repertoire and cytotoxic features from CD56dim NK cells. European Journal of Immunology. 2001;31(10):3121-3126. DOI: 10.1002/1521-4141(2001010)31:10<3121:AID-IMMU3121gt;3.0.CO;2-4
  36. 36. Kärre K. Natural killer cell recognition of missing self. Nature Immunology. 2008;9(5):477-480. DOI: 10.1038/ni0508-477
  37. 37. Podack ER. Functional significance of two cytolytic pathways of cytotoxic T lymphocytes. Journal of Leukocyte Biology. 1995;57(4):548-552
  38. 38. Konjevic G, Schlesinger B, Cheng L, Olsen KJ, Podack ER, Spuzic I. Analysis of perforin expression in human peripheral blood lymphocytes, CD56+ natural killer cell subsets and its induction by interleukin-2. Immunological Investigations. 1995;24(3):499-507
  39. 39. Simon MM, Hausmann M, Tran T, Ebnet K, Tschopp J, ThaHla R, et al. In vitro- and ex vivo-derived cytolytic leukocytes from granzyme A × B double knockout mice are defective in granule-mediated apoptosis but not lysis of target cells. The Journal of Experimental Medicine. 1997;186(10):1781-1786
  40. 40. Beresford PJ, Xia Z, Greenberg AH, Lieberman J. Granzyme A loading induces rapid cytolysis and a novel form of DNA damage independently of caspase activation. Immunity. 1999, 1999;10(5):585-595
  41. 41. Lieberman J, Fan Z. Nuclear war: The granzyme A-bomb. Current Opinion in Immunology. 2003;15(5):553-559
  42. 42. King KL, Cidlowski JA. Cell cycle regulation and apoptosis. Annual Review of Physiology. 1998;60(1):601-617. DOI: 10.1146/annurev.physiol.60.1.601
  43. 43. Bratke K, Kuepper M, Bade B, Virchow JC, Luttmann W. Differential expression of human granzymes A, B, and K in natural killer cells and during CD8+ T cell differentiation in peripheral blood. European Journal of Immunology. 2005;35(9):2608-2616
  44. 44. Kelly JM, Waterhouse NJ, Cretney E, Browne KA, Ellis S, Trapani JA, et al. Granzyme M mediates a novel form of perforin-dependent cell death. The Journal of Biological Chemistry. 2004;279(21):22236-22242. DOI: 10.1074/jbc.M401670200
  45. 45. Pardo J, Balkow S, Anel A, Simon MM. Granzymes are essential for natural killer cell-mediated and perf-facilitated tumor control. European Journal of Immunology. 2002;32(10):2881-2886. DOI: 10.1002/1521-4141(2002010)32:10<2881
  46. 46. Zamai L, Ahmad M, Bennett IM, Azzoni L, Alnemri ES, Perussia B. Natural killer (NK) cell-mediated cytotoxicity: Differential use of TRAIL and Fas ligand by immature and mature primary human NK cells. The Journal of Experimental Medicine. 1998;188(12):2375-2380
  47. 47. Srivastava S, Lundqvist A, Childs RW. Natural killer cell immunotherapy for cancer: A new hope. Cytotherapy. 2008;10(8):775-783. DOI: 10.1080/14653240802648181
  48. 48. Morel PA, Ernst LK, Metes D. Functional CD32 molecules on human NK cells. Leukemia & Lymphoma. 1999;35(1-2):47-56. DOI: 10.3109/10428199909145704
  49. 49. Leibson PJ. Signal transduction during natural killer cell activation: Inside the mind of a killer. Immunity. 1997;6(6):655-661. DOI: org/10.1016/S1074-7613(00)80441-0
  50. 50. Smyth MJ, Cretney E, Kelly JM, Westwood JA, Street SE, Yagita H, et al. Activation of NK cell cytotoxicity. Molecular Immunology. 2005;42(4):501-510. DOI: 10.1016/j.molimm.2004.07.034
  51. 51. Nagler A, Lanier LL, Cwirla S, Phillips JH. Comparative studies of human FcRIII-positive and negative natural killer cells. Journal of Immunology. 1989;143(10):3183-3191
  52. 52. Penack O, Gentilini C, Fischer L, Asemissen AM, Scheibenbogen C, Thiel E, et al. CD56 dim CD16 neg cells are responsible for natural cytotoxicity against tumor targets. Leukemia. 2005;19(5):835-840. DOI: 10.1038/sj.leu.2403704
  53. 53. Romee R, Foley B, Lenvik T, Wang Y, Zhang B, Ankarlo D. NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood. 2013;121(18):3599-3608. DOI: 10.1182/blood-2012-04-425397
  54. 54. Tarazona R, Casado JG, Delarosa O, Torre-Cisneros J, Villanueva JL, Sanchez B, et al. Selective depletion of CD56 dim NK cell subsets and maintenance of CD56 bright NK cells in treatment-naive HIV-1-seropositive individuals. Journal of Clinical Immunology. 2002;22(3):176-183
  55. 55. Mavilio D, Lombardo G, Benjamin J, Kim D, Follman D, Marcenaro E, et al. Characterization of CD56–/CD16+ natural killer (NK) cells: A highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(8):2886-2891. DOI: 10.1073/pnas.0409872102
  56. 56. Björkström NK, Ljunggren HG, Sandberg JK. CD56 negative NK cells: Origin, function, and role in chronic viral disease. Trends in Immunology. 2010;31(11):401-406. DOI: 10.1016/j.it.2010.08.003
  57. 57. Zulu MZ, Naidoo KK, Mncube Z, Jaggernath M, Goulder PJ, Ndung'u T, et al. Reduced expression of Siglec-7, NKG2A, and CD57 on terminally differentiated CD56 CD16+ natural killer cell subset is associated with natural killer cell dysfunction in chronic HIV-1 clade C infection. AIDS Research and Human Retroviruses. 2017;33(12):1205-1213. DOI: 10.1089/AID.2017.0095
  58. 58. Sivori S, Carlomagno S, Pesce S, Moretta A, Vitale M, Marcenaro E. TLR/NCR/KIR: Which one to use and when? Frontiers in Immunology. 2014;5:105. DOI: 10.3389/fimmu.2014.00105
  59. 59. Mogensen TH. Pathogen recognition and inflammatory signaling in innate immune defenses. Clinical Microbiology Reviews. 2009;22(2):240-273. DOI: 10.1128/CMR.00046-08
  60. 60. Qiu F, Maniar A, Diaz MQ , Chapoval AI, Medvedev AE. Activation of cytokine-producing and antitumor activities of natural killer cells and macrophages by engagement of toll-like and NOD-like receptors. Innate Immunity. 2011;17(4):375-387. DOI: 10.1177/1753425910372000
  61. 61. Wooldridge JE, Ballas Z, Krieg AM, Weiner GJ. Immunostimulatory oligodeoxynucleotides containing CpG motifs enhance the efficacy of monoclonal antibody therapy of lymphoma. Blood. 1997;89(8):2994-2998
  62. 62. Janeway CA Jr, Medzhitov R. Innate immune recognition. Annual Review of Immunology. 2002;20(1):197-216. DOI: 10.1146/annurev.immunol.20.083001.084359
  63. 63. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nature Immunology. 2004;5(10):987-995. DOI: 10.1038/ni1112
  64. 64. De Nardo D. Toll-like receptors: Activation, signalling and transcriptional modulation. Cytokine. 2015;74(2):181-189. DOI: 10.1016/j.cyto.2015.02.025
  65. 65. Zhang Z, Schluesener HJ. Mammalian toll-like receptors: From endogenous ligands to tissue regeneration. Cellular and Molecular Life Sciences. 2006;63(24):2901-2907. DOI: 10.1007/s00018-006-6189-1
  66. 66. Demaria S, Pikarsky E, Karin M, Coussens LM, Chen YC, El-Omar EM, et al. Cancer and inflammation: Promise for biological therapy. J Immunother (Hagerstown, Md.: 1997). 2010;33(4):335-351. DOI: 10.1097/CJI.0b013e3181d32e74
  67. 67. Łagiedo M, Sikora J, Kaczmarek M. Damage-associated molecular patterns in the course of lung cancer—A review. Scandinavian Journal of Immunology. 2015;82(2):95-101. DOI: 10.1111/sji.12308
  68. 68. Rifkin IR, Leadbetter EA, Busconi L, Viglianti G, Marshak-Rothstein A. Toll-like receptors, endogenous ligands, and systemic autoimmune disease. Immunological Reviews. 2005;20(1):27-42. DOI: 10.1111/j.0105-2896.2005.00239.x
  69. 69. Marshak-Rothstein A. Toll-like receptors in systemic autoimmune disease. Nature Reviews Immunology. 2006;6(11):823-835. DOI: 10.1038/nri1957
  70. 70. Goh FG, Midwood KS. Intrinsic danger: Activation of toll-like receptors in rheumatoid arthritis. Rheumatology. 2012;51(1):7-23. DOI: 10.1093/rheumatology/ker257
  71. 71. Drexler SK, Foxwell BM. The role of toll-like receptors in chronic inflammation. The International Journal of Biochemistry & Cell Biology. 2010;42(4):506-518. DOI: 10.1016/j.biocel.2009.10.009
  72. 72. Joosten LA, Abdollahi-Roodsaz S, Dinarello CA, O'Neill L, Netea MG. Toll-like receptors and chronic inflammation in rheumatic diseases: New developments. Nature Reviews Rheumatology. 2016;12(6):344-357. DOI: 10.1038/nrrheum.2016.61
  73. 73. Liu Q , Ding JL. The molecular mechanisms of TLR-signaling cooperation in cytokine regulation. Immunology and Cell Biology. 2016;94(6):538-542. DOI: 10.1038/icb.2016.18
  74. 74. Roach JC, Glusman G, Rowen L, Kaur A, Purcell MK, Smith KD, et al. The evolution of vertebrate Toll-like receptors. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(27):9577-9582. DOI: 10.1073/pnas.0502272102
  75. 75. Uematsu S, Akira S. Toll-like receptors and type I interferons. The Journal of Biological Chemistry. 2007;282(21):15319-15323. DOI: 10.1074/jbc.R700009200
  76. 76. Gaudreault E, Fiola S, Olivier M, Gosselin J. Epstein-Barr virus induces MCP-1 secretion by human monocytes via TLR2. Journal of Virology. 2007;81(15):8016-8024. DOI: 10.1128/JVI.00403-07
  77. 77. Hertz CJ, Wu Q , Porter EM, Zhang YJ, Weismüller KH, Godowski PJ, et al. Activation of toll-like receptor 2 on human tracheobronchial epithelial cells induces the antimicrobial peptide human β defensin-2. Journal of Immunology. 2003;171(12):6820-6826. DOI: 10.4049/jimmunol.171.12.6820
  78. 78. Guillot L, Le Goffic R, Bloch S, Escriou N, Akira S, Chignard M, et al. Involvement of toll-like receptor 3 in the immune response of lung epithelial cells to double-stranded RNA and influenza a virus. The Journal of Biological Chemistry. 2005;280(7):5571-5580. DOI: 10.1074/jbc.M410592200
  79. 79. Vivier E, Biron CA. A pathogen receptor on natural killer cells. Science. 2002;296(5571):1248-1249. DOI: 10.1126/science.1072447
  80. 80. Kadowaki N, Antonenko S, Ho S, Rissoan MC, Soumelis V, Porcelli SA, et al. Distinct cytokine profiles of neonatal natural killer T cells after expansion with subsets of dendritic cells. The Journal of Experimental Medicine. 2001;193(10):1221-1226
  81. 81. Uehori J, Matsumoto M, Tsuji S, Akazawa T, Takeuchi O, Akira S, et al. Simultaneous blocking of human Toll-like receptors 2 and 4 suppresses myeloid dendritic cell activation induced by Mycobacterium bovis bacillus Calmette-Guerin peptidoglycan. Infection and Immunity. 2003;71(8):4238-4249. DOI: 10.1128/IAI.71.8.4238-4249.2003
  82. 82. Cella M, Salio M, Sakakibara Y, Langen H, Julkunen I, Lanzavecchia A. Maturation, activation, and protection of dendritic cells induced by double-stranded RNA. The Journal of Experimental Medicine. 1999;189(5):821-829
  83. 83. Adib-Conquy M, Scott-Algara D, Cavaillon JM, Souza-Fonseca-Guimaraes F. TLR-mediated activation of NK cells and their role in bacterial/viral immune responses in mammals. Immunology and Cell Biology. 2014;92(3):256-262. DOI: 10.1038/icb.2013.99
  84. 84. Hayden MS, Ghosh S. NF-κB in immunobiology. Cell Research. 2011;21(2):223-244. DOI: 10.1038/cr.2011.13
  85. 85. Hornung V, Rothenfusser S, Britsch S, Krug A, Jahrshdörfer B, Giese T, et al. Quantitative expression of toll-like receptor 1-10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. Journal of Immunology. 2002;168(9):4531-4537. DOI: 10.4049/jimmunol.168.9.4531
  86. 86. He S, Chu J, Wu LC, Mao H, Peng Y, Alvarez-Breckenridge CA, et al. MicroRNAs activate natural killer cells through toll-like receptor signaling. Blood. 2013;121(23):4663-4671. DOI: 10.1182/blood-2012-07-441360
  87. 87. Saikh KU, Lee JS, Kissner TL, Dyas B, Ulrich RG. Toll-like receptor and cytokine expression patterns of CD56+ T cells are similar to natural killer cells in response to infection with Venezuelan equine encephalitis virus replicons. The Journal of Infectious Diseases. 2003;188(10):1562-1570. DOI: 10.1086/379196
  88. 88. Chalifour A, Jeannin P, Gauchat JF, Blaecke A, Malissard M, N’Guyen T, et al. Direct bacterial protein PAMP recognition by human NK cells involves TLRs and triggers α-defensin production. Blood. 2004;104(6):1778-1783. DOI: 10.1182/blood-2003-08-2820
  89. 89. Gorski KS, Waller EL, Bjornton-Severson J, Hanten JA, Riter CL, Kieper WC, et al. Distinct indirect pathways govern human NK-cell activation by TLR-7 and TLR-8 agonists. International Immunology. 2006;18(7):1115-1126. DOI: 10.1093/intimm/dxl046
  90. 90. Alter G, Suscovich TJ, Teigen N, Meier A, Streeck H, Brander C, et al. Single-stranded RNA derived from HIV-1 serves as a potent activator of NK cells. Journal of Immunology. 2007;178(12):7658-7666. DOI: 10.4049/jimmunol.178.12.7658
  91. 91. Lauzon NM, Mian F, MacKenzie R, Ashkar AA. The direct effects of toll-like receptor ligands on human NK cell cytokine production and cytotoxicity. Cellular Immunology. 2006;241(2):102-112. DOI: 10.1016/j.cellimm.2006.08.004
  92. 92. Siednienko J, Miggin SM. Expression analysis of the toll-like receptors in human peripheral blood mononuclear cells. Methods in Molecular Biology. 2009;517:3-14. DOI: 10.1007/978-1-59745-541-1_1
  93. 93. Girart MV, Fuertes MB, Domaica CI, Rossi LE, Zwirner NW. Engagement of TLR3, TLR7, and NKG2D regulate IFN-γ secretion but not NKG2D-mediated cytotoxicity by human NK cells stimulated with suboptimal doses of IL-12. Journal of Immunology. 2007;179(6):3472-3479. DOI: 10.4049/jimmunol.179.6.3472
  94. 94. Hart OM, Athie-Morales V, O’Connor GM, Gardiner CM. TLR7/8-mediated activation of human NK cells results in accessory cell-dependent IFN-γ production. Journal of Immunology. 2005;175(3):1636-1642. DOI: 10.4049/jimmunol.175.3.1636
  95. 95. Souza-Fonseca-Guimaraes F, Parlato M, Philippart F, Misset B, Cavaillon JM, Adib-Conquy M. Toll-like receptors expression and interferon-γ production by NK cells in human sepsis. Critical Care. 2012;16(5):R206. DOI: 10.1186/cc11838
  96. 96. Martinez J, Huang X, Yang Y. Direct TLR2 signaling is critical for NK cell activation and function in response to vaccinia viral infection. PLoS Pathogens. 2010;6(3):e1000811. DOI: 10.1371/journal.ppat.1000811
  97. 97. Cañeda-Guzmán IC, Salaiza-Suazo N, Fernández-Figueroa EA, Carrada-Figueroa G, Aguirre-García M, Becker I. NK cell activity differs between patients with localized and diffuse cutaneous leishmaniasis infected with leishmania mexicana: A comparative study of TLRs and cytokines. PLoS ONE. 2014;9(11):e112410. DOI: 10.1371/journal.pone.0112410. eCollection 2014
  98. 98. Lindgren Å, Pavlovic V, Flach CF, Sjöling Å, Lundin S. Interferon-gamma secretion is induced in IL-12 stimulated human NK cells by recognition of Helicobacter pylori or TLR2 ligands. Innate Immunity. 2011;17(2):191-203. DOI: 10.1177/1753425909357970
  99. 99. Kanevskiy L, Telford WG, Sapozhnikov AM, Kovalenko EI. Lipopolysaccharide induces IFN-γ production in human NK cells. Frontiers in Immunology. 2013;4:1-10. DOI: 10.3389/fimmu.2013.00011
  100. 100. Roda JM, Parihar R, Carson WE. CpG-containing oligodeoxynucleotides act through TLR9 to enhance the NK cell cytokine response to antibody-coated tumor cells. Journal of Immunology. 2005;175(3):1619-1627. DOI: 10.4049/jimmunol.175.3.1619
  101. 101. Merlo A, Calcaterra C, Mènard S, Balsari A. Cross-talk between Toll-like receptors 5 and 9 on activation of human immune responses. Journal of Leukocyte Biology. 2007;82(3):509-518. DOI: 10.1189/jlb.0207100
  102. 102. Tsujimoto H, Uchida T, Efron PA, Scumpia PO, Verma A, Matsumoto T, et al. Flagellin enhances NK cell proliferation and activation directly and through dendritic cell-NK cell interactions. Journal of Leukocyte Biology. 2005;78(4):888-897. DOI: 10.1189/jlb.0105051
  103. 103. Vacchelli E, Prada N, Kepp O, Galluzzi L. Current trends of anticancer immunochemotherapy. OncoImmunology. 2013;2(6):e25396. DOI: 10.4161/onci.25396
  104. 104. Lu H. TLR agonists for cancer immunotherapy: Tipping the balance between the immune stimulatory and inhibitory effects. Frontiers in Immunology. 2014;5:83. DOI: 10.3389/fimmu.2014.00083
  105. 105. Li K, Qu S, Chen X, Wu Q , Shi M. Promising targets for cancer immunotherapy: TLRs, RLRs, and STING-mediated innate immune pathways. International Journal of Molecular Sciences. 2017;18(2):E404. DOI: 10.3390/ijms18020404

Written By

Carmen Maldonado-Bernal and David Sánchez-Herrera

Submitted: 20 September 2018 Reviewed: 17 April 2019 Published: 31 May 2019