Open access peer-reviewed chapter

IL-17 Biological Effects and Signaling Mechanisms in Human Leukemia U937 Cells

Written By

Samuel Evans Adunyah, Richard Akomeah, Fareed K.N. Arthur, Roland S. Cooper and Joshua C.M. Williams

Submitted: 28 October 2020 Reviewed: 04 February 2021 Published: 30 March 2021

DOI: 10.5772/intechopen.96422

From the Edited Volume

Interleukins - The Immune and Non-Immune Systems’ Related Cytokines

Edited by Payam Behzadi

Chapter metrics overview

428 Chapter Downloads

View Full Metrics

Abstract

Human Interlekin-17 is produced by memory activated CD4+ T cells and other cells. It was initially considered unique in that its specific receptor is distinct from other cytokine receptors. IL-17 receptor is ubiquitously expressed by different cells including T cells. IL-17 plays a role in regulating growth, immune response and pro-inflammatory responses. It regulates differentiation of a subset of Th0 cells into Th-17 cells, which produce IL-17-induced cytokines. The IL-17R belongs to type 1 cytokine receptors. IL-17 belongs to a superfamily of its own, which includes IL-17A, IL-17B, IL-17C, IL-17E and IL-17F. These members of IL-17 superfamily have some sequence homology but bind to different receptors. Prior to this investigation, limited information existed on the effects of IL-17A in human leukemia cell lines. Our results show that IL-17A promotes growth, anti-apoptotic effects, chemotaxis, cytokine expression and transcriptional factor activation in leukemia cells. IL-17A activates multiple signaling pathways including PI-3 K, Jak–STAT, Raf-ERK1/2 and SRC kinase pathways, which mediate different biological effects of IL-17A in leukemia cells. Our findings implicate IL-17A in leukemia cell growth and survival, supporting potential leukemia therapy via development of anti-IL-17A drugs. This chapter focuses on IL-17A, herein referred to as IL-17.

Keywords

  • IL-17A
  • leukemia
  • cytokines
  • Jak/STAT
  • PI-3 K/Akt
  • ERK1/2 transcriptional factors

1. Introduction

IL-17 is a unique cytokine which was initially discovered through differential and subtractive screening of clones from DNA library from murine lymphoid cells and initially called T-lymphocyte associated antigen 8 (CTLA-8) [1]. IL-17 was found to have 50% sequence homology to the open reading frame 13 (ORF-13) in herpes virus Saimiri [1]. Subsequently, the human homolog of CTLA-8 was identified [1] and its incubation with human fibroblast resulted in induction of both IL-6 and IL-8. This led to renaming of the CTLA-8 human equivalent as IL-17 [2]. Human IL-17 production was also found to be limited to particular cellular elements of the immune system and that activated CD4 + T cells of the Th1/Th0 subset and stimulated memory T cells, synthesize IL-17 [3, 4]. IL-17 is a glycosylated homodimeric protein of 30 to 35 kDa also produced by nickel-specific T lymphocytes and it regulates I-CAM-1 expression and chemokine production [5]. Unlike other cytokines, IL-17 was noted to bind to a unique receptor distinct from other cytokine receptors [1, 2, 3]. IL-17 is ubiquitously expressed in thymocytes activated by CD3 mAb, CD45R0+ population of T cells, CD8+ splenic cells in mouse cells and synovial fluid of patients with rheumatoid arthritis [6, 7, 8]. IL-17 is abundantly produced in CD4+ T cells, now known as Th-17 cells [9, 10, 11]. It is also expressed in human peripheral blood mononuclear cells (PBMC) in response to ocular lysate in patients with birdshot chroioretinopathy [12]. IL-17 has biological effects in many cells and tissues [13, 14, 15]. IL-17 induces expression and secretion of IL-1-beta, IL-6, IL-8. TNF, GM-CSF, G-CSF, ICAM-1, and PGE2 [16, 17, 18, 19, 20]. The molecular characterization of IL-17 receptor (IL-17R) was reported in 1997 [21]. IL-17R is a type 1 transmembrane receptor and it is a single chain, which shares some properties with IL-2R-beta chain, and GM-CSFR, all of which are type 1 membrane receptors [21, 22]. IL-17R is also expressed in synovial endothelial cells and Chondrocytes from arthritis patients [23, 24].

Five different IL-17 ligands are now characterized as members of IL-17 superfamily of cytokines and differ from other cytokines but share some sequence homology with each other [25]. Among the IL-17 super family, IL-17A is most commonly expressed in many tissues as well as in cells of hematopoietic origin including monocytes and macrophages. In addition to IL-17A, there are IL-17B, IL-17C, IL-17E and IL-17F [25, 26]. IL-17B, IL-17C and IL-17E expression are widespread in many tissues including testis, brain and kidney [25, 26]. Each IL-17 family members have their individual specific receptor as these IL-17 family members do not bind to the same receptor type [26, 27, 28]. The different members of IL-17 superfamily have different expression patterns but with similar abilities to stimulate cytokine effectors illustrating the potential for the members of the IL-17 superfamily to differentially regulate cellular responses in a wide variety of cells [28, 29, 30, 31].

IL-17 regulates hematopoietic cell proliferation, immune response, pro-inflammatory responses [32, 33, 34, 35] and activate specific types of T cells now known as Th-17 cells [9, 10]. These Th-17 cells play a role in host defense against extracellular pathogens by mediating recruitment of neutrophils and macrophages to infected tissues [9, 10, 11]. Th-17 cells secrete IL-17 cytokines, which in turn induce expression of IL-17-depedemt cytokines. Hence, aberrant regulation of Th-17 cells may play a role in the pathogenesis of multiple inflammatory and autoimmune disorders [9, 10, 11]. IL-17 promotes chemotaxis in human monocytes and regulates angiogenesis and cytokine production in endothelial cells [12, 36, 37, 38]. Although the target cells of IL-17-mediated signaling include immune cells such as neutrophils and macrophages [32, 33, 34, 35, 36, 37, 38], majority of IL-17’s biological effects were seen in cells of either epithelial or mesenchymal origin [39, 40, 41, 42, 43]. The role of IL-17 in immunological function was initially examined in vivo in mice by overexpressing IL-17 in the liver of mice where an enhanced granulopoiesis and leukopoiesis led to an 80% increase in splenic mass [38, 39]. IL-17-induced accumulation of neutrophils in the airways requires involvement of GM-CSF [44]. Also, regulation of endogenous stem cell by IL-17 requires both GM-CSF and Stem Cell Factor (SCF) [45]. IL-17 and G-CSF are synergistically involved in the maintenance of normal granulopoiesis [45]. The IL-17R is ubiquitously expressed [12, 13, 14, 15] and may explain the ability of IL-17 to stimulate peripheral blood stem cells in in mice [44, 45]. Also, There IL-17 plays active in vivo role in chemoattraction of cells of immune system [46, 47]. In addition, IL-17 exhibits paracrine effects in different cell types [48] whereby secreted IL-17 from T cells binds to its putative receptor on neighboring cells such as fibroblasts and trigger signaling that leads to NF-kB- mediated induction of expression and secretion of ICAM-I, IL-2, IL-6, IL-8 [16, 17, 18, 19, 20] and other cytokines, which produce different biological effects [45, 46, 47, 48, 49, 50]. Also some of the T-cell secreted IL-17 become sequestered and neutralized by a soluble IL-17R (sIL-17R) [51].

Clinically, IL-17 is implicated in numerous diseases including arthritis [52, 53, 54], classical Hodgkin lymphomas [55, 56, 57], multiple myeloma [58, 59, 60], airway diseases including asthma [61, 62], musculoskeletal diseases [63, 64], inflammatory bowel diseases (IBDs) [65] autoimmune diseases [66], and different types of cancer [67, 68, 69]. Significantly elevated level of IL-17 and IL-17R are found in these diseases and IL-17 and IL-17R are known to promote anti-apoptotic effects and survival mechanisms in some types of cancer [55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66]. In most cases, IL-17 itself and/or IL-17-dependent cytokines produced downstream of the IL-17R, contribute to various pathological conditions associated with these diseases [55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68]. Furthermore, Hox3/IL-17R expression ratio has been implicated in poor prognosis in some breast cancer patients undergoing tamoxifen chemotherapy as IL-17 promotes resistance to chemotherapy in breast cancer [67, 68, 69, 70, 71]. IL-17 is also implicated in cervical and ovarian cancer [72, 73, 74]. Similarly, expression of IL-17 R-like protein has been detected in androgen independent prostate cancer cell lines and it has been implicated in conferring resistance to apoptosis and promoting prostate cancer via MM7-induced epithelial-to-mesenchymal transition [75, 76, 77]. IL-17 is implicated in CNS and other neurological diseases [78, 79, 80], and psoriasis [81, 82, 83]. Recent reports suggest potential role for IL-17 in the “cytokine storm event” seen in advance Coronavirus Disease 2019 (COVID-19) infection with inflammation, and pro-thrombic events in severe COVID-19 patients [84, 85, 86, 87]. Hence, there is strong interests in understanding IL-17’s biology and it roles in COVID-19 patients [85, 86, 87]. It is not surprising that IL-17’s role in these diseases [60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87], have prompted experts in pharmaceutical industries to develop anti-IL-17 type therapies for diseases in which IL-17 is implicated [88, 89, 90].

Earliest report on IL-17 induced activation of MAP kinases and NF-kB pathways was made in chondrocytes [91]. Subsequently, IL-17 was shown to activate Raf-MAPK and Jak/STAT signaling pathways in leukemia cells [92, 93]. These reports show that IL-17 stimulates rapid phosphorylation of RAF, Erk-1/2, Jak1, Jak2, Jak3 and Stat1, Stat2 and Stat3 in human leukemia cells [92, 93]. Currently, IL-17 is known to activate and utilize multiple signaling pathways including the aforementioned as well as JNK, p38 and PI-3 K/Akt pathways to produce diverse biological effects [91, 92, 93, 94, 95, 96]. Many reports have confirmed IL-17-induced activation of PI-3 K/Akt signaling mechanisms in both normal and transformed cells [96, 97, 98, 99]. IL-17 signaling pathways are implicated in human diseases including inflammation and cancer [100]. Furthermore, TRAF and TGF-beta-1/smad2/3 signaling pathways are activated by IL-17 [99, 100, 101, 102]. Most of the biological effects of IL-17 were initially observed in different variety of cells but to lesser extent in leukemia cells. Therefore, our rationale for initiating this study was to determine the biological effects of IL-17 in leukemia cells and elucidate the various signaling pathways utilized by IL-17 in leukemia cells. Furthermore, we wanted to determine which transcriptional factors are activated by IL-17. Finally, we wanted to determine whether IL-17 protects leukemia cells from undergoing apoptosis since previous report [75] indicated IL-17−mediates cancer cell resistance to apoptosis.

Advertisement

2. Experiment reagents and protocols

We purchased human U937 and THP-1 leukemia cell lines from American Tissue Culture Collection (ATCC) in Manassas, VA, USA. The cells were cultured in Roswell Park Memorial Institute-1640 media, which contained L-Glut (2 mM). Charcoal-filtered and frozen fetal bovine serum (FBS) was purchased from Atlanta Biological, Georgia, USA. Following careful thawing under sterile conditions we heated the serum at 55 °C for 45 minutes for inactivation. After cooling, we vacuum filtered the FBS under the hood and stored 50 ml aliquots at minus 20 °C. Both streptomycin and penicillin were purchased from Invitrogen, Carlsbad, CA. Prior to using the culture media, we added FBS to a final concentration of 10% (v/v). To prevent bacterial growth in the culture media, we added penicillin (50 U/ml) and streptomycin (50 U/ml). We cultured the cells in either 25 ml or 50 ml of complete media in tissue culture flasks in a CO2 incubator set to 5% CO2, 37 °C and 100% humidity. Typically, we passaged the cells 8x before starting a fresh culture. U937 leukemia cells were used in most of the experiments described here.

2.1 Monocyte isolation

We purchased de-identified human blood samples from New York Blood Center, Long Island, NY and Percoll gradients from GR Health Care, Piscataway, NJ [103]. To isolate monocytes for chemotaxis assays, we isolated peripheral blood mononuclear cells (PBMC) as previously described [103]. After centrifugation to remove all red blood cells, the white blood cells were carefully retrieved and suspended in 10 ml of complete media and spread across the surface of plastic dishes. The plastic dishes were incubated in the incubator for 1.5 hours to allow monocytes to attach to the surface of the dishes. Subsequently, all non-adherent cells were carefully aspirated off and discarded. The attached monocytes were carefully scraped from the dishes and suspended in culture media. The monocytes were about 95% pure based on positive staining for CD14 marker.

2.2 Detection of cytokine expression by cytokine antibody array

To determine the effect of IL-17 on cytokine expression in leukemia cells, we performed cytokine antibody array using tissue media from untreated and IL-17 treated leukemia cells [103]. Specifically, 20million cells were either untreated or treated with IL-17 (100 ng/ml) alone or with IL-17 (100 ng/ml) plus the PI-3 K inhibitor LY20094 (20 uM) or with LY20094 (20 uM) alone for 24 hours. The tissue culture media were filtered to remove debris and their protein concentration determined by Coomassie Blue Protein Assay Kit (Pierce, IL). The culture media containing 50 ug protein from untreated and treated cells were spotted onto each of the cytokine/antibody array membranes containing antibodies for over 42 cytokines (RayBiotech, Corners, GA, USA), and incubated with gentle shaking for 2 hours at 30 °C. This allowed hybridization of each cytokine in the media to its respective cytokine antibody on the array. Next, the media was carefully removed and each membrane was washed 5x with wash buffer (provided by kit) to remove all non-specific binding. Each membrane was incubated for a specified time in the color development solution provided with kit and air dried. The dark spots representing various cytokines were visualized and quantitated by digital image scanning. The spot intensities were converted to fold change relative to the corresponding spots on the membrane of the untreated cells, which was set as 1-fold.

2.3 Western blotting detection of proteins and phosphoproteins

One million leukemia cells per ml media were either untreated (control) or treated with [92, 93] IL-17 (1 ng/ml) for 2, 5, 15, 30, 60 min or with IL-17 (100 ng/ml) at 30 °C treatments up to 48 hours. Next, the cells were rapidly pelleted by micro-centrifugation at 1,800 x g for 3 minutes. The pelleted cells were washed 3x with PBS. The final pellets were collected by centrifugation at 1800 rpm for 3 min and each pellet was lysed in 500 ul of cell lysis buffer A (containing protease inhibitors, 0.5% Triton X-100, 50 mM NaF and 2 mM Vanadate) [7]. Total cell lysate protein concentration was determined by Coomassie Blue Protein Assay Kit (Pierce, IL) and 240 ug per sample was solubilized in 50 ul SDS gel sample buffer and resolved by 12% polyacrylamide gel electrophoresis. The protein bands were transferred to membrane and the membrane background blocked in a blocking buffer containing 5% milk. The membranes were incubated with specific antibodies to either total PI-3 K, p-PI-3 K, p-AktSer473, p-AktThre308, total Akt, p-STAT3, total STAT3, p-BAD, p-caspase3 or pGSK3-beta or total actin (for loading control) and protein bands detected [103]. The band intensities were scanned by digital image analyzer for quantitation and the band intensity from IL-17 treated samples compared to the intensity in the untreated sample.

2.4 Co-immunoprecipitation (co-IP) assay/Western blot

Total and phosphorylated Akt can associate with effector proteins [104, 105] via protein–protein interaction [106] and contribute to their regulation. We rationalized that if IL-17 promotes association between p-Akt and any of its downstream effectors, those proteins will be contained in the pulled down p-Akt-antibody complex and can be detected by co-IP/Western blot. To determine whether Akt/p-Akt binds to p-BAD or p-Caspase3 or p-GSK-3 (p-Akt’s downstream effectors), we carried out co-IP. Specifically, 240 mg/ml protein from untreated or IL-17 treated cells were suspended in PBS (500 ul) in Eppendorf tubes and incubated with specific antibody that recognizes both total Akt and p-AktSer473 overnight with gentle shaking at 4 °C. Next, protein A agarose slurry (500 ul) was added to the complex in each tube and incubated for 2 hours at 4 °C with gentle shaking to allow protein A agarose to capture all the phosphoproteins bound to Akt/p-Akt-antibody complex. The tubes were centrifuged at 10,000 x g for 10 minutes at 4 °C, the supernatants carefully removed and the pellets were washed 5x with lysis buffer (see above). After the final wash, the phosphoprotein complexes in each tube was solubilized in SDS-gel sample buffer (50 ul) and boiled for 3 minutes to dissociate the phosphoproteins in each complex. The dissociated phosphoproteins were separated on 12% SDS-polyacrylamide gel as described above. The phosphoprotein bands were transferred to membrane and Western blotted for either p-BAD or p-Caspase3 p-GSK-3 or total Akt using specific antibodies. The band intensity for p-BAD, p-Caspase3 and p-GSK-3 were scanned with digital image analyzer. The old levels in the IL-17 treated cells calculated relative to the bands in the untreated cells. Representative results are presented.

2.5 Detection of IL-17RA in U937 leukemia cells and THP-1 leukemia cells

Specific antibody to IL-17AR was purchased from Santa Cruz, CA and goat anti-rabbit IgG-HRP antibody was from Amersham, CA. In order to detect IL-17AR, 240 ug of total cell lysate protein from 40 million cells was separated on 15% polyacrylamide gel [106]. The protein bands were transferred to nitrocellulose membrane followed by incubation in a blocking buffer containing 5% filtered non-fat milk to block non-specific sites on the membrane. Both the IL-17AR antibody and the goat-anti-rabbit antibody were used at dilutions of 1:1000. The rest of the Western blot protocols were performed as described [103, 106]. The band intensity was quantitated using digital image analyzer.

2.6 Transcription factor array

To detect transcriptional factors regulated by IL-17 stimulation in leukemia cells, we examined the profile of 54 transcriptional factors (TFs) using Panomics Transcriptional Factor Array (1) Kit. Panomics TranSignal™ Protein/DNA Arrays simplifies the functional analysis of eukaryotic TFs and can be used to study TF activation in a variety of biological processes, including cell proliferation, differentiation, transformation, apoptosis and drug treatment [107]. The array membranes were spotted with 54 different consensus-binding sequences (oligos) and enable one to detect over 54 TFs at once in one treatment. Twenty million leukemia cells/ml were pretreated with vanadate (5 mM) for 30 minutes to inhibit endogenous phosphatases. Next, 5 million of the cells we either untreated or stimulated with IL-17 (100 ng/ml) for 4 hours in the incubator. The cells were packed by centrifugation at 1,800 x g for 3 minutes, washed 3x with PBS and gently lysed in a lysing buffer (see Western blot protocols above) without detergents by repeated aspiration through a 22-gauge needle to prevent rupturing of the nuclei. Intact nuclei were isolated by layering the cell lysate over 50% glycerol solution in Eppendorf tubes followed by centrifugation at 1,000 x g for 5 minutes. The supernatants were carefully aspirated, the nuclei pellet harvested and washed 2x with PBS. Next, the nuclei were disrupted in a nuclei lysing buffer (provided by the kit) and protein concentration determined as described above. In a slightly modified version, 12 ug of nuclei proteins in 200 ul of incubation buffer were incubated with each array membrane containing the oligos for hybridization of each oligo to its specific transcriptional factor in the nuclei extract. The membranes were washed several times and the oligo/transcriptional factor complexes (DNA/protein complexes) were detected by detection per the kit. The spots representing the various transcription factors were identified based on the charts provided by the kit. The intensities of the spots were scanned by digital image analyzer and the fold stimulation by IL-17 compared to the intensities in the untreated cells.

2.7 NF-kB/DNA and STAT3/DNA binding assays for detection of NF-kB and STAT3 activation by IL-17

To study the effect of IL-17 on NF-kB and STAT3 DNA binding functions [108, 109], 4 million leukemia cells/ml were untreated or IL-17 in time course experiments. The cells were used for specific NF-kB/DNA binding or STAT3/DNA binding assays using the NF-kB and STAT transcription factor assay kits (Active Motif, Chemicon). The kit enabled us to monitor the activation or repression of NF-kB or STAT3 proteins. The experiments were performed in triplicate.

2.8 Cell proliferation assays

To determine whether IL-17 stimulates cell proliferation in leukemia cells, 6 x 105 cells were either untreated or stimulated with IL-17 (100 ng/ml) for 48 hours [110]. The cells were harvested and aliquots were diluted into 0.4% trypan blue/PBS solution at a ratio of 1:10. The cells were counted in triplicate and the average viable cell count was recorded from each sample. We also performed MTT proliferation assay using 4 x 105 cells untreated or cells treated with IL-17 (100 ug/ml) for 48 hours in 96 well plates in triplicate. The rest of the details of the MTT assay protocols were as previously described [110].

2.9 Caspase3 activity assays as evidence for apoptosis

We purchased caspase3 assay kit from MBL International, Woburn, MA, US [111]. Sodium butyrate (NaB) is a strong inducer of apoptosis in cancer cells [112]. To determine the effects of IL-17 on NaB-induced apoptosis in leukemia cells we performed caspases3 enzymatic (colorimetric) assays in lysates from 15 x 106 untreated or NaB treated cells. Specifically, the cells were either untreated or treated with NaB (5 mM) alone, or treated with NaB (5 mM) plus IL-17 (100 ng/ml) or with IL-17 (100 ng/ml) alone in tissue culture media for up to 48 hours. At the end of the incubation, aliquot of cells were counted prior to lysing in RIPA buffer (provided in the caspase3 assay kit) and total lysate protein concentration was determined as indicated above. To measure caspase3 activity in the cell lysates, 30 ug total lysate protein from each sample was added to each experimental assay well. The enzymatic activity was measured in triplicate in microtiter plate reader according to instructions provided by the kit.

2.10 Assessment of chemotactic effects of media from IL-17 treated cells

In order to ascertain if media from IL-17 treated cells has chemotactic effects towards monocytes [103, 113], we employed the Boden Chamber chemotaxis assay method as previously described [103]. Specifically, 2000 monocytes/ml in fresh complete media were placed in the upper portion of the Boden Chamber. Equal volume of media from either untreated or IL-17 stimulated cells was put in the lower chamber to serve as a source of chemotaxis. The setup was incubated for 2-hours. Next, estimation of number of monocytes which crossed the membrane barrier to the lower chamber was performed according to the kit. Experiments were conducted in triplicate.

2.11 Assessing IL-17-induced cytokine expression by cytokine ELISA

In order to validate IL-17-induced cytokine expression observed in the cytokine antibody array, we performed cytokine ELISA assay as specified in the cytokine ELISA Kits (Ray Biotech, Norcross, GA) using equal amount of tissue culture proteins from untreated or IL-17 treated cells [103]. The ELISA monitored expression of IL-2, IL-3 and IL-8. In addition, to determine whether IL-17-induced IL-2 expression was mediated by either PI-3 K/Akt or by Jak2, in some experiments, we pre-incubated some of the cells with PI-3 K inhibitor LY20094 (20 nM) or Jak2 inhibitor AG490 (15 nM) for 30 minutes prior to stimulating the cells with IL-17 (100 ng/ml) for 24 hours.

Advertisement

3. Results

3.1 Effects of IL-17 on cytokine expression in human leukemia cells

To determine whether IL-17 stimulates cytokine expression in leukemia cells, we performed cytokine antibody array using tissue culture media from untreated and treated cell. As seen in Figure 1, within 24 hours IL-17 stimulated several fold differential expression of various cytokines in the ranking order of IL-2 > IL-3 > GRO > IL-10 > RANTES>IL-15 > IL-1. However, IL-17 failed to simulate IL-8 expression. Stimulation of cytokine expression by IL-17 was significantly inhibited by the PI-3 K inhibitor LY20094 (Figure 1), suggesting a role for PI-3 K in the mechanism by which IL-17 stimulates cytokine expression. Similar results were observed in THP-1 human leukemia cell line (data not shown). Also, a neutralizing antibody against of IL-17 blocked IL-17 from stimulating cytokine expression (data not shown), confirming that the observed stimulation of cytokine expression is attributed to L-17. Using ELISA assay, we confirmed stimulation of IL-2 and IL-3 expression by IL-17 without effect on IL-8 expression (Figure 2). The lack of effect of IL-17 on IL-8 expression seen in leukemia cells are in contrast to IL-17-induced IL-8 expression reported in different cell types [16, 17, 18, 19, 20]. As a follow up to our results in Figure 1, and our previous report that IL-17 activates Jak/STAT pathway [93], we determined whether both the PI-3 K and Jak2 mediate stimulation of specific cytokine expression by IL-17. To do so, we examined the effects of PI-3 K inhibitors LY20094 (LY) and wortmannin (WM) and Jak2 inhibitor AG490 (AG)) on IL-17-induced IL-2 expression. The ELISA array results in Figure 3 indicate that individually LY20094 and wortmannin partially inhibited IL-17 stimulated IL-2 expression. The Jak2 inhibitor AG490 also exhibited similar inhibitory effect on ability of IL-17 to stimulate IL-2 expression. A combination of both LY20094 and AG490 completely blocked stimulation of IL-2 expression by IL-17 (not shown). These results confirmed roles for both PI-3 K and Jak2 in the mechanisms by which IL-17 stimulates IL-2 expression.

Figure 1.

IL-17 Stimulation of differential expression of cytokines: Inhibition by PI-3K Inhibitor (LY20094). Tissue culture media from untreated and treated cells were assayed for cytokine expression by cytokine-antibody array. Data is an average of two experiments.

Figure 2.

Effects of IL-17 on IL-2, IL-3 and IL-8 expression. Asterisk (*) indicates significant differences between IL-17 treated and untreated cells.

Figure 3.

Inhibition of IL-17-induced IL-2 expression by PI-3K inhibitors (LY and WM) and Jak2 inhibitor (AG). Asterisk (*) indicates significant differences between IL-17 alone and IL-17 plus PI-3K Inhibitors (LY and WM) or Jak2 inhibitor (AG490).

3.2 Media from IL-17 treated leukemia cells produce chemotaxis

Given that IL-17 stimulated significant expression of two chemokines (GRO and RANTES), we examined whether the culture media from IL-17 treated leukemia cells could serve as chemoattractant to human monocytes from PBMC. As seen in Figure 4, as compared to culture media from untreated cells, culture media from IL-17 treated cells exhibited significant time-dependent chemotaxis towards monocytes, confirming that culture media from IL-17 treated cells contains secreted chemotactic chemokines that induced chemotaxis [46].

Figure 4.

IL-17 induced Chemotaxis. Tissue culture media from untreated IL-17 cells were assayed for chemotaxis in a Boyden Chamber with monocytes on the upper chamber.

3.3 IL-17 stimulates leukemia cell growth and protection from apoptosis

Next, we investigated the effect of IL-17 on leukemia cell proliferation. Untreated or IL-17 stimulated cells were assessed for cell growth using trypan blue exclusion and MTT assays. As shown in Table 1, IL-17 exhibited a time-dependent stimulation of leukemia cell growth by 3.3-fold within 48 hours. Similar results were seen in MMT assays (data not shown). Next, we examined whether IL-17 promotes leukemia cell survival and anti-apoptotic effects in leukemia cells by protecting the leukemia cells from apoptosis. The results in Table 2 indicates that NaB alone causes significant reduction in leukemia cell survival from 100% to 52% in 24 hours. However, in the presence of IL-17, Na-induced decline in cell survival is markedly inhibited cell survival improved from 52% to 83%. As shown in Table 3, NaB alone stimulated activation of caspase3 activity from 1-fold in untreated cells to 3.2 fold in 24 hours and 4.3-fold in 48 hours, indicating NaB-induced apoptosis in cells in the absence of IL-17. However, in the presence of IL-17, NaB-induced caspase3 activation is markedly reduced from 3.2-fold to 1.7-fold in 24 hours and from 4.3-fold to 2.0-fold in 48 hours. IL-17 also upgrades Bcl2 in the presence of NaB (not shown). Thus, IL-17 protects leukemia cells from undergoing apoptosis and enhances their survival. The results suggest that IL-17 may be inducing inactivation of pro-apoptotic signals while partially restoring the anti-apoptotic protein Bcl-2 expression.

Treatment24 hours Cell Growth (Fold)48 hours Cell Growth (Fold)
Cells1.01.0
Cells + IL-172.3.03.3 ± 0.2

Table 1.

Effects of IL-17 on cell growth, cell survival and caspase3 activity—IL-17 promotes cell growth.

Treatment24 hours Cell Survival (%)
Cells alone100
Cells + NaB52 ± 2.2
Cells + IL-17 + NaB83 ± 1.3

Table 2.

Effects of IL-17 on cell growth, cell survival and caspase3 activity—IL-17 protects cells from butyrate-induced apoptosis.

Treatment24 hour; Relative Caspase 3 Activity (Fold)48 hours Relative Caspase 3 Activity (Fold)
Cells alone1.01.0
Cells + NaB3.2 ± 0.44.3 ± 0.1
Cells + IL-17 + NaB1.7 ± 0.22.0 ± 0.3

Table 3.

Effects of IL-17 on cell growth, cell survival and caspase3 activity—IL-17 inhibits butyrate-induced caspase3 activation. Data represent mean plus/minus SD.

3.4 IL-17 stimulates differential activation of transcription factors in leukemia cells

Within 4 hours IL-17 stimulated significant and differential activation of several transcription factors in the order of c-Myb (5.5-fold) > EGR-1 (5.0-fold) > STAT3 (4.0-fold)> > Smad3/4 (3.4-fold) > SRE (3.0 fold>CDP (2.5-.fold). IL-17 failed to activate NF-kB. Using individual transcription factor/DNA binding assays, we confirmed that STAT3/DNA binding activity is significantly enhanced by IL-17 (Figure 5). In contrast, IL-17 did not stimulate NF-kB/DNA binding activity in these leukemia cells (Figure 6). Together these results show that IL-17 differentially activates several transcriptional factors associated with regulation of cell growth, cell differentiation and apoptosis but failed to stimulate NF-kB in these cells even though IL-17 is known to stimulate NF-kB in many cell types [50]. Of note, NF-kB is already highly constitutively expressed in active from in these leukemia cells. This could explain why IL-17 failed to stimulate NF-kB activation further in these cells.

Figure 5.

Activation of STAT3 Transcriptional Activity by IL-17.

Figure 6.

Lack of IL-17 stimulation of NF-kB DNA Binding Activity.

3.5 Direct evidence that IL-17 activates PI-3 K/Akt signaling pathway in leukemia cells

Because the PI-3 K inhibitor Ly20094 inhibited IL-17-induced cytokine expression, we examined the direct effects of IL-17 on PI-3 K and Akt phosphorylation and activation. As shown in Figure 7a and 7b, in as early as 0.5 minutes, IL-17 stimulated PI-3 K tyrosine phosphorylation by 4.5-fold. PI-3 K phosphorylation and activation usually lead to downstream Akt (PKB) activation [113]. Therefore, we next examined the effects of IL-17 on Akt (PKB) phosphorylation and activation. Akt can be phosphorylated on Serine 473 (Ser473) and/or Threonine 308 (Thr308), which is in the activation domain. The western blot results in Figure 8a show that IL-17 stimulated Akt phosphorylation on Serine473 to 5-fold within 10 min in these cells. The results in Figure 8b show that IL-17 stimulates rapid phosphorylation of Akt on Thr308 with maximum effect noted at 5 minutes. Stimulation of Akt phosphorylation on Serine473 by IL-17 was inhibited by the PI-3 K inhibitor wortmannin (WM) (no shown). These results imply that stimulation of Akt phosphorylation by IL-17 is mediated by PI-3 K. Once Akt is activated, it phosphorylates a host of downstream effectors including BAD, Caspase3, forkhead transcription factor (FKHR), glycogen synthase kinase-3 (GSK3-beta), AFX, eNOS,TSC2, MDM2, P21/CIP1 and other downstream effectors as shown in Figure 9a Dephosphorylated BAD, caspase3 and GSK3-beta play vital roles in induction of apoptosis [114]. However, upon their phosphorylation, these pro-apoptotic proteins lose their pro-apoptotic activities [114] as phosphorylation of both BAD, caspase3 and GSK-3 leads to their inactivation. The results in Figure 9b and c show that IL-17 stimulates Akt-mediated BAD, Caspase3 and GSK-3-beta phosphorylation as p-BAD, p-Caspase3 and p-GSK3-beta were contained in Akt pulled down complex from IL-17 treated cells. Phosphorylation of caspse3, BAD, GSK3-beta and STAT3 are associated with enhanced cell survival [115, 116] and could explain in part how IL-17 promotes cell survival. Also, IL-17 stimulated Akt-dependent phosphorylation of mammalian target of rapamycin (mTor) on serine 2448 (motor Ser 2448) [117], which was inhibited by the Akt inhibitor SH5 (not shown).

Figure 7.

Time course of IL-17-induced PI-3K phosphorylation detected by Western blot using either specific antibody for tyrosine phosphorylated PI-3K or total PI-3K as loading control. Scanned values represent ptyr-PI-3K/PI-3K ratios from 3 experiments (b). Asterisk (*) indicates significant differences between IL-17 treated and untreated cells. Results are representation form several experiments.

Figure 8.

Time course of IL-17 stimulation of AktSer473 phosphorylation (a) and (b) AktThr308 Specific antibody to either AktSer473 or AktThr308 was used to monitor Akt phosphorylation by Western blot. Blots were stripped and reprobed for total Akt for loading control. The blots from 3 experiments were scanned and results are presented. Asterisk (*) indicates significant differences between IL-17 treated and untreated cells.

Figure 9.

Model showing activated Akt phosphorylation of its downstream targets (a). Effect of IL-17 on Caspase3 and BAD phosphorylation (b), GSK-3-beta phosphorylation (c). In (b) and (c) cells were untreated or stimulated with IL-17 and total cell lysates were monitored for Caspase3, BAD and GSK-3-beta phosphorylation by Western blot using specific phosphoprotein antibody to each protein. Total Akt (b) or total GSK-3-beta (c) was probed for loading control. Results are representation of several experiments.

Advertisement

4. Discussions

We have provided strong evidence that IL-17 stimulates significance and differential expression of IL-2, IL-3, IL-10, IL-15, GRO, and RANTES in human leukemia cells. The stimulatory effect of IL-17 on cytokine expression in these cells is similar to previous reports in non-hematopoietic cells by IL-17 [16, 17, 18, 19, 20]. However, IL-17 does not stimulate IL-8 expression in these cells, which contradicts early reports that IL-17-induces IL-8 expression in different cell types [17, 18, 19, 20]. Induction of cytokine expression by IL-17 in these leukemia cells could have strong biological relevance in vivo because increases in IL-17 level in a tumor microenvironment can trigger induction of other cytokines including chemokines that could generate combination of proinflammatory, anti-inflammatory and chemotactic responses [16, 17, 18, 19, 20, 54]; [79, 80, 81]. IL-2 is a proinflammatory cytokine [118], which also regulates helper T cell differentiation [119]. IL-3 stimulates regulation of multipotent hematopoietic stem cell function and differentiation of all lineages as well as promote proliferation of myeloid progenitor cells [120, 121]. IL-10 is a master regulator of immunity to infection and an anti-inflammatory cytokine that can counteract the pro-inflammatory effects of IL-2 [122]. Secondly, IL-10 is known to synergize with IL-2 to promote CD8 + T cell cytotoxicity [123]. IL-15 is known to suppress apoptosis in T-lymphocytes by inducing Bcl2 and/or Bcl-xl in humans [124]. Perhaps, IL-15 contributes to the anti-apoptotic effect of IL-17 in these cells. Both IL-2 and IL-15 have structural and functional similarities, share the common gamma chain of their receptors and promote immune response [125]. Both GRO and RANTES are chemokines and are associated with induction of chemotaxis and recruitment of neutrophils and macrophages to sites of infection [37]. Thus, IL-17- induced GRO and RANTES expression and secretion from leukemia cells into the culture media, could account for the chemotactic effect IL-17 seen in our studies.

These leukemia cells express receptors for some of the cytokines secreted to the culture media in response to IL-17. Therefore, some of the cytokines secreted into culture media can promote both autocrine and paracrine effects on the leukemia cells. We have provided evidence that IL-17 stimulates phosphorylation of the pro-apoptotic proteins BAD, caspase3 and GSK3-beta, thus negating their functions. In addition, IL-17 promotes inhibition of Caspase3 activity in these leukemia cells. Furthermore, IL-17 enhances Akt phosphorylation and activation, which are associated with cell survival [126]. The ability of IL-17 to enhance protection of the leukemia cells from apoptosis implies that elevated IL-17 levels in a tumor microenvironment could lead to promotion of leukemia cell proliferation and survival, both of which could potentially produce poor prognosis in leukemia patients. Another interesting outcome of this study is that IL-17 stimulates activation of several transcriptional factors including cMyb, EGR-1, STAT3, Smad3/4, SRE, CDP, which are known to regulate proliferation, differentiation and survival [115, 127, 128]. This effect of IL-17 could in part contribute to the mechanism growth promotion and survival in these leukemia cells. Stimulation of smad3/4 transcriptional factors of the TGF-beta signaling pathway [102, 115] by IL-17 may point to potential cross talk between IL-17 and TGF-beta-induced signaling pathways to synergize their biological effects [127, 128]. The lack of activation of NF-kB by IL-17 in these leukemia cells is not surprising since typically these leukemia cells constitutively express high levels of active NF-kB, which could explain the apparent lack of NF-kB response to IL-17. Lack of NF-kB activation by IL-17 in these cells is in contrast to IL-17-induced NF-kB activation reported in many cells [50].

We have provided ample evidence that IL-17 activates and utilizes the Jak/STAT signaling pathway in these leukemia cells. In this pathway, IL-17 stimulates phosphorylation of Jak1, Jak2 and Jak3, STAT1, STAT2, and STAT3 [55, 93, 97]. We have also shown that Jak2 partially mediates IL-17-induced IL-2 expression. Furthermore, IL-17 strongly stimulates phosphorylation and activation of PI-3 K/ Akt pathway and promoting Akt-mediated phosphorylation of its downstream effectors. Another interesting observation is that Akt-partially mediates stimulation of IL-2 expression and secretion by IL-17. Also, IL-17 promotes phospho-Akt’s association with BAD, caspase3 and GSK3-beta, supporting Akt-mediated phosphorylation of these proteins in IL-17 treated cells. These observations could in part explain how IL-17 promotes anti-apoptosis and survival in these leukemia cells [75, 76, 77, 126].

IL-17 stimulates activation of Raf–MEK–ERK1/2 pathway [92, 93, 94, 95, 101], which could partially account for the growth promoting effects of IL-17 in leukemia cells. Previous thesis research in our laboratory revealed that IL-17 stimulates activation of LCK [129] and PKC [130]. Also, IL-17 promotes association between LCK and the p85 subunit of the PI-3 K, thus providing another mechanism for PI-3 K activation by IL-17 via LCK, a member of the Src kinases family [129]. Activation of PKC by IL-17 is associated with enhanced PKC ability to regulate cell cycle progression in leukemia cells [130]. As indicated earlier, IL-17 is profoundly implicated in many human diseases [55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74], thus supporting the suggestion that design and production of anti-IL-17 drugs could lead to better strategies for development of new therapies for those diseases [88, 89, 90]. Although the recent reports implicating IL-17 in the mechanism of the “cytokine storm” event in COVID-19 infection is far from conclusion, there are calls for development of anti-IL-17 drugs as adjunct therapy for diseases in which IL-17 plays an active role [87, 131]. IL-17-enhanced leukemia cell growth, survival and anti-apoptosis strengthens the argument in favor of inclusion of leukemia in the list of human diseases for which anti-IL-17 adjunct therapy should be considered. Our model in Figure 10 explains the paracrine role of T-cell secreted IL-17 in leukemia cells. Elucidation of the multiple signaling mechanisms of IL-17 in leukemia cells in our study and illustrated in Figure 11 further enrich our knowledge on the biological effects and mechanisms of IL-17.

Figure 10.

Model showing activated memory T cell secreted IL-17: Paracrine mechanism of how secreted IL-17 activates cytokine expression and secretion in leukemia cells.

Figure 11.

Model showing multiple signaling mechanisms used by IL-17 in Leukemia Cells.

Advertisement

5. Conclusion

Our studies on effects and mechanisms of IL-17 in human U937 leukemia cells revealed that these cells express IL-17A receptor and IL-17 stimulates cell growth, survival, chemotaxis and differential expression of cytokines. These results suggest that IL-17 could trigger expression and secretion of various cytokines including chemotactic chemokines in leukemia patients. Also, IL-17 promotes anti-apoptotic effects in these cells. If these biological effects of IL-17 described here, were to occur in leukemia patients, IL-17 could promote poor prognosis in the patients. Furthermore, IL-17 stimulates differential activation of several transcriptional factors including c-Myb, EGR-1, STAT3, smad3/4 CDP and SRE but not NF-kB in these cells. Lastly, multiple signaling pathways including PI-3 K/Akt, Jak/STAT, Raf–MEK-ERK-1/2 and Lck signaling pathways differentially mediate the biological effects of IL-17 in the U937 leukemia cells. Any of these pathways could serve as a target for anti-IL-17 drugs.

Advertisement

Acknowledgments

This work was partially supported by NIAMS/NIH R03 grant, U54 cancer partnership NCI grant U54CA091408 and NIGMS/NIH SCORE grant to Professor Adunyah, who was also supported by cancer partnership grant U54CA163069/NCI during preparation of this chapter. Professor Arthur was partially supported by Biochemistry Department, KNUST, Kumasi, Ghana during his sabbatical. We thank Dr. S. V. Subramaniam and W. Williams for their contribution in the initial stages of this work.

References

  1. 1. Rouvier E, Luciani MF, Mattéi MG, Denizot F, Golstein P. CLTA cloned from activated T cell, bearing au-rich messenger RNA instability sequences, and homologous to a herpes saimiri gene. The Journal of Immunology. 1993;150(12):5445–5456
  2. 2. Yao Z, Painter SL, Fanslow WC, Ulrich D, Macduff BM, Springs MK, Armitage RJ. Human IL-17: a novel cytokine derived from T cells. J. Immunology.1995b;155:5483–5486
  3. 3. Avarvak T, Chabbbaud M, Natvig JB. IL-17 produced by some pro-inflammatory Th1/Th0 cells but not Th2 cells. J. Immunol.1999;162:1246–1251
  4. 4. Tartour E, Fossiez F, Joyeux I, Galinha A, Gey A, Claret E, et al. Interleukin-17, a T-cell-derived cytokine, promotes tumorigenicity of human cervical tumors in nude mice. Cancer Research. 1999;59:3698–3704
  5. 5. Albanesi C, Caavni A, Giromori G. IL-17 is produced by nickel-specific T lymphocytes and regulates ICAM-I expression and chemokine production in human keratinocytes: synergistic or antagonist effects with IFN-gamma and TNF-alpha. J. Immunol. 1999;162:494–502
  6. 6. Shin HCK, Benbemou N, Esnault S, Guenounou M. Expression of IL-17 in human memory CD45RO+T lymphocytes and its regulation by protein kinase A. 1999;11(4):257–266
  7. 7. Kotake S, Udagawa N, Takahashi N, et al. IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. J. Clin. Invest. 1999;103(9):1345–1352
  8. 8. Nakae S, Saijo S, Horai E, Sudo K, Morai S, Iwakura Y. IL-17 production from activated T cells is required for the spontaneous development of destructive arthritis in mice with deficient in IL-1 receptor antagonist. Proc. Nat. Acad. Sci. 2003;100(10):5986–5990
  9. 9. Betelli E, Korn T, Kuchroo VK. Th17: The third member of the effector T cell Trilogy. Curr. Opin. Immunol. 2007;19(6):652–657
  10. 10. Chen Z, Osahea JJ. Th17 Cells: a new fate for differentiating helper T cells. Immunol. Res. 2008;41:87–102
  11. 11. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Ann. Rev. Immunol. 2009;27:495–517
  12. 12. Lenarczyk A, Helsloot J, Farmer K, Peters L, Sturgess A, Kirkham B. Antigen-induced IL-17 response in peripheral blood mononuclear cells PBMC) of healthy controls. Clin. Exp. Immunol. 2000;122(1):41–48
  13. 13. Kuiper, JJW, Emmelot ME, Rothova A, Mutis T. Interleukekin-17 production and T helper cells in peripheral blood mononuclear cells in response to ocular lysate in patients with birdshot chorioretinopathy. Molecular Vision. 2013;19:2606–2614
  14. 14. Lourda M, Olsson-Åkefeldt S, Gavhed D, et al. Detection of IL-17A-producing peripheral blood monocytes in Langerhans cells histiocytosis patients. Clin. Immunol. 2014;153(1)112–122
  15. 15. Szabo PA, Goswami A, Mazzuca DM, Kim K, et al. Rapid and Rigorous IL-17A production in distinct subpopulation of effector memory T lymphocytes constitute a novel mechanism of toxic shock syndrome immunopathology. Journal Immunol.2017;doi:10.4049/jimmunol.1601366:1-19
  16. 16. Hirata T, Osuga Y, Hamasaki K, Yoshino O, Ito M, Hasegawa A. et al. Interleukin-17 stimulates IL-8 secretion, cyclo0xygenease-2 expression, and cell proliferation of endometriotic stromal cells. Endocrinology. 2008;149(3):1260–1267
  17. 17. Kuwabara T, Ishikawa F, Kondo M, Kakiuchi T. The role of IL-17 and related cytokines in Inflammatory autoimmune diseases. Mediators of Inflammation. 2017;2017:doi.org/10.1155/2017/390861, 11 pages
  18. 18. Numasaki M, Tomioka Y, Takahashi H, Sasaki H. IL-17 and IL-17F modulate GM-CSF production by lung microvascular endothelial cells stimulated with IL-1beta and/or TNF-alpha. Immunology Letters, 2004;95(2):175–184
  19. 19. Fossiez F, Djossou O, Chomarat P, Flores-Romo L, Ait-Yahia S, Maat C, et al cell Interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines. J. Exp. Med. 1996;183(6):2593–2603
  20. 20. Honda K, Wada H, Nakamura M, Nakamoto K, et al. IL-17A synergistically stimulates TNF-alpha-induced IL-8 production in human airway epithelial cells: A potential role in amplifying airway inflammation. Exp. Lung Res. 2016;42(4):2052016
  21. 21. Yao Z, Springs MK, Deny JMJ, Stockbine L, Park LS, VendenBos T, Zappone J, Painter SL, Armitage RJ. Molecular Characterization of the human IL-17 receptor. Cytokine. 1997;9(11):794–800
  22. 22. Gaffen S. Structure and signaling of the IL-17 receptor superfamily. Nat Rev. Immunol. 2009;9(8):556]
  23. 23. Honrati MC, Meliconi R, Pulsatelli S, Cané S, Frizziero L, Facchini. A High in vivo expression of Interleukin-17 receptor in synovial endothelial cells and chondrocytes from arthritis patients. Rheumatology. 2001;40(5):522–527
  24. 24. Haudenchild D, Moseley T, Rose L, Redd AH. Soluble and transmembrane isoforms of novel Interleukin-17 receptor-like protein by RNA splicing and expression in prostate cancer. J.Biol.Chem. 2002;277(6):4309–4316
  25. 25. Kawaguchi M, Adachi M, et al. IL-17 cytokine Family. J. Allergy and Clin, Immunol. 2004;114(6):1265–1273
  26. 26. Gaffen SL, Kramer JM, Yu JJ, Shen F. The IL-17 Cytokine Family. Vitamins and Hormones. 2006;74:255–282
  27. 27. Li H, Chen J, Huang A, Stinson J, Heldens S, Foster J, Dowd P, Gurney A, Wood WL. Cloning and characterization of IL-17B And IL-7C, two new members of the IL-17 family. Proc. Nat. Acad. Sci. 2000;97(2):773–778
  28. 28. Lee J, Maruoka M, Corpuz T, Baldin DT, Foster JS, Goddard AD, Yamsura DG, Vandlen RL, Gurney AL. IL-17E, a novel proinflammatory ligand for the IL-17 receptor homolog IL-17Rh1.J. Biol. Chem. 2001;276(2):1660–1664
  29. 29. Moseley A, Haudenchild DR, Rose L, Reddi AH. Interleukin-17 family and IL-17 receptors. Cytokine Growth Factor Rev. 2003;14(2):155–174
  30. 30. McAllister F, Henry A, Kreinder JJ, et al. Role of IL-17A, IL-17F, and the IL-17 receptor in regulating stimulating factors in bronchial epithelial: implications for airway inflammation in Cystic Fibrosis. J. Immunol. 2005;175(1):404–412
  31. 31. Kuestner RE, Taft DW, Haran A, Brandt CS, Brender T, Lum K, et al. Identification of the IL-17 receptor related molecule IL-17C as the receptor for IL-17F. J. Immunol. 2007;179(8):5462–5373
  32. 32. Han Q, Das S, Hirano M, Holland SJ, McCurley MD, Guo P, Rosenberg CS, Boehm T, Cooper MD. Characterization of Lamprey IL-17 Family members and their receptors. J. Immunol. 2015;195:5440–5451
  33. 33. Broxmeyer HE, Starnes T, Ramey H, Cooper S, Dahl R, Williamson E, Hromas R. The IL-17 cytokine family members are inhibitors of human hematopoietic progenitor proliferation. Blood. 2006,108(2):770–771
  34. 34. Krstic A, Mojsiović S, Bugarski D, The potential of Interleukin-17 to mediate hematopoietic response. Immunologic Res. 2012;52:34–41
  35. 35. Mojsilović S, Jauković A, Santibaňez JF, Bugarski, D. Interleu-17 and its implication in the regulation of differentiation and function of hematopoietic and mesenchymal stem cells. Mediators of Inflammation. 2015; doi.org/10.1155/2015/470458
  36. 36. Schwarzenberger P, La Russa V, Miller A, Ye P, Huang W, Zieske A, Nelson S, et al. IL-17 stimulates granulopoiesis in mice: use of an alternative, novel gene therapy-derived method for in vivo evaluation of cytokine. J Immunol. 1998;161:6383–6389
  37. 37. Witowski J, Pawlaczyk K, Breborowicz A, Scheuren A, Kuzlan-Pawlaczk, Wiseniewska J, Polubinska A, et al. IL-17 stimulates intraperitoneal neutrophils infiltration through GRO-alpha chemokine from mesothelial cells. J. Immunol. 2000;165:5814–5821
  38. 38. Schwarzenberger P, Huang W, Oliver P, Byme P, La Russa V, Zhang Z, Kolls JK. IL-17 mobilizes peripheral blood stem cells with short-and long-term responding ability in mice. J. Immunol. 2001;167:2081–2086
  39. 39. Bradley Fellow S, Schurr JR, Kolls JK, Bagby GJ, Schwarzenberger P, Ley K. Increased granulopoiesis through IL-17 and granulocyte colony-stimulating factor in leukocyte adhesion molecule-deficient mice. Blood. 2001;98:3309–3314
  40. 40. Huang H, Kim HJ, Chang EJ, Lee ZH, Huang SJ. IL-17 stimulates the proliferation and differentiation of human mesenchymal stem cells. Cell Differ. 2009;16(10):1332–1343
  41. 41. Han X, Yang Q, Shi Y. Interleukin-17 enhances immunosuppression by mesenchymal stem cells. Cell Death and Differ. 2014;21:1758–1768
  42. 42. Ma T, Wang X, Jiao Y, Halto W, Qi Y, Gong H, Zhang L, Jiang D. Interleukin-17-induced mesenchymal stem cells prolong the survival of allogeneic skin. Ann Transplant. 2018;23:615–621
  43. 43. Liao S, Zhang C, Jin L, Yang Y. IL-17 alters the mesenchymal stem cell niche towards osteogenesis in corporation with osteocytes. J. Cell. Phys. 2020;235(5):4466–4480
  44. 44. Laan M, Prause O, Miyamoto M, Sjostrand M, et al. A role of GM-CSF in the accumulation of neutrophils in the airways caused by IL-17 and TNF. Euro. Res. J. 2003;21(3):387–393
  45. 45. Schwarzenberger P, Huang W, Ye P, Oliver P. Requirement of endogenous stem cell factor and Granulocyte-Colony Stimulating Factor for IL-17-mediated granulopoiesis. The J. Immunol. 2000;164(9):4783–4789
  46. 46. Shahara S, Pickins SR, Mandelin II AM, Karpus, WJ, Huang Q, Kolls JK, Pope RM. IL-17-mediate monocyte migration occurs partially through CCL2/MCP-1 induction. J. Immunol. 2010;184(8):4479–4487
  47. 47. Chin C-C, Chen C-N, Kuo H-C, Shi C-S, Hsieh MC, et a. Interleukin-17 induces CC chemokine receptor 6 expression and cell migration in colorectal cancer cells. J. Cell Physiol. 2015;230(7):1403–1437
  48. 48. Chung AS, Wu X, Zhuang G, et.al. An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nature Med.2013;19:1114–1123 Res. Ther. 2004;6(2).doi.org/10.1186/ar1038
  49. 49. Wang L, Yi T, Kortylewski M, Pandoll DM, Zang D, Yu H. IL-17 can promote tumor growth through and IL-6-STAT3 signaling pathway. J. Exp. Med. 2009;206(7):1457–1464
  50. 50. Hwang S-Y, Kim J-Y, Kim K-W, ParkM-K, Moon Y, Kim W-U, Kim H-Y. IL-17 induces production of IL-6 and IL-8 in rheumatoid arthritis synovial fibroblasts via NF-kB- and PK-3Kinase/Akt-dependent pathways. Arthritis Res. & Therapy. 2004;6(2):1–9
  51. 51. Zaretsky M, Etzyoni R, Kaye J, Sklair-Tarvon L, Aharoni A. Directed for evolution of a soluble human IL-17A receptor for the inhibition of psoriasis plaque formation in a mouse model. J. Chem. Biol. 2012. Doi.org/10.1016/J. ChemBiol.2012.11.012
  52. 52. Gaffen SL. Role of IL-17 in the pathogenesis of Rheumatoid arthritis. Curr. Rheumatol. Rep. 2009;11(5):385–370
  53. 53. Robert M, Miossec P. IL-17 in rheumatoid arthritis and precision medicine: from synovitis expression to circulating bioactive levels. Frontiers of Med. 2018;doi.org/10.3389/fmed.2018.00364
  54. 54. Beringer A, Miossec P. Systemic effects of IL-17 in inflammatory arthritis. Nat. Rev. Rheumatol. 2019:15:491–501
  55. 55. Krejsgaard T, Ralfkiaer U, Clasen-Linde E, Eriksen KW, at al. Malignant cutaneous T-cell lymphoma cells express IL-17 utilizing the Jak3/Stat3 signaling pathway. J. Invest. Dermatol. 2001;131(6):1331–1338
  56. 56. Ferrarini I, Rigo A, Zamb A, Vinante F. Classical Hodgkin lymphoma cells may promote an IL-17-enriched microenvironment. Leukemia and Lymphoma. 2019;60(14):1–11
  57. 57. Zhong W, Li Q. Rituximab or irradiation promotes IL-17 secretion thereby induces resistance to rituximab or irradiation. Cell. Mol. Immunol. 2017;14:1020–1022
  58. 58. Prabhala RH, Pelluru D, Fulciniti M, Song W, Pai C, et. Al. Elevated IL-17 level produced by Th-17 cells promote myeloma cell growth and inhibits immune function in multiple myeloma. Blood. 2010;115(26):5385–5392
  59. 59. Prabhala RH, Fulciniti M, Pelluru D, Rashid N, Nigroiu A, Nanpappa P, et al. Targeting IL-17A in multiple myeloma: a potential novel therapeutic approach in myeloma.Leukemia. 2016;30(2):379–389
  60. 60. Calcinotto A, Brevi A. Chesi M, et al. Microbiota-driven IL-17-producing cells and eosinophils synergize accelerate multiple myeloma progression. Nature Commun. 2018;9:4832.doi.org/10.1038/s41467-018-07305-8
  61. 61. Yanagisawa H, Hashimoto H, Minagawa S, Takasaka N, Moermans MR, et al. Role of IL-17 in murine model of COPD airway disease. Am J. Physiol. Lung Cell Mol.Physiol. 2017;312(1):L122-L130.doi.10.1152/ajplumg.00301.2016
  62. 62. Hynes GM, Timothy SC, Hinks ER.J. The role of Interleukin-17 in asthma: a protective response?. Open Research. 2020;6:00364–2019;doi.10.1183/23120541.00384-2019
  63. 63. Lee Y. The role of interleukin-17 in bone metabolism and inflammatory skeletal diseases. BMB Rep.2013;46(10):479–483
  64. 64. Yago, T, Nanke Y, Ichikawa N, Kobashigawa T, Mogi M, Kamatani N, Kotake S. IL-17 induces osteoclastogenesis from human monocytes alone in the absence of osteoblasts, which is potentially inhibited by anti-TNF-alpha antibody: a novel mechanism of osteoclastogenesis by IL-17. J. Cell Biochem. 2009;108:947–955
  65. 65. Fujino, S, Andoh A, Bamba S, Ogawa A, Hata K, Anaki Y, Bamba T, Fujiyama Y. Increased expression of interleukin-17 in inflammatory bowel disease. Gut. 2003;52(1):65–70
  66. 66. Kuwabara T, Ishikawa T, Kondo M, Kakiuchi T. The role of IL-17 and related cytokines in inflammatory autoimmune diseases. Mediators of Inflammation. 2017;2017.Doi.org/10.1155/2017/3908061
  67. 67. Yang B, Kang H, Fung A, Zhao H, Wang T, Ma D. The role of interleukin-17 in tumor proliferation, angiogenesis and metastasis. Mediators of Inflam. 2014.doi.org/10.1155/2014/623739
  68. 68. Zhao J, Chen X, Herjan T, Li X. The role of interleukin-17 in tumor development and progression. J. Exp. Med. 2019;217(1)1–13
  69. 69. Zhu L, Mulcahy A, Mohammed AAA, et al. IL-17 expression by breast -cancer-associated macrophages: IL-17 promotes invasiveness of breast cancer cell lines. Breast Cancer Res. 2008;10(6):article R95
  70. 70. Bian G. Zhao W-Y. “IL-17, an important prognostic factor and potential therapeutic target for breast cancer”? Euro J. Immunol. 2014;33(2):604–605
  71. 71. Cochaud S, Giustiniani J, Thomas C, et al. IL-17A is produced by breast cancer TILs and promotes chemoresistance and proliferation through ERK1/2. Sci. Rep 3, article #3456 (2013).doi.org/10.1038/srep03456
  72. 72. Tartour E, Fossiez F, Joyeux I, Galinha A, Gey A, et al. Interleukin-17, a T-cell-derived cytokine, promotes tumorigenicity of human cervical tumors in nude mice. Can. Res. 1999;59:3698–3704
  73. 73. Bonin CM, Almeida-Lugo LZ, dos Santo, AR, Junqueira CT, et al. Interleukin-17 expression in the serum and exfoliated cervical cells of patients with high-risk oncogenic human papillomavirus. Cytokine. 2019;120:97–98
  74. 74. Xiang T, Long H, He L, Han X, Lin K, Liang Z, Zhuo W, Xie R, Zhu B. Intterleukin-17 produced by tumor microenvironment promotes self-renewal of CD133+ cancer stem-like cells in ovarian cancer. Oncogene. 2015;34(2):165–176
  75. 75. You Z, Shi X-B, DuRaine, G, Haudenschild D, Clifford G, et al. Interleukin-17 receptor-like gene is a novel anti-apoptotic gene highly expressed in androgen-independent prostate cancer. Can Res. 2006;66(1):175–183
  76. 76. Zhang Q, Liu S, Ge D, Zhang Q, Xue Y, Xiong Z, et al. Interleukin-17 promotes formation and growth of prostate cancer adenocarcinoma in mouse models. Can Res. 2012;72(10);2589–2599
  77. 77. Zhang Q, Liu S, Parakuli KR, Zhang W, Zhang K, Mao Z, et al. Interleukin-17 promotes prostate cancer via MMP-induced epithelial-to-mesenchymal transition. Oncogene. 2017;36(5):687–699
  78. 78. Waisman A, Hauptmann J, Regen T. The role of IL-17 in CNS diseases. Acta Neuropathilogica. 2015;129:625–637
  79. 79. Milovanovic J, Arsenijevic A, Stojanovic B, et al. Interleukin-17 in chronic inflammatory neurological diseases. Frontier in Immunol. 2020;11(947):doi.10.3389/fimmu.2020.00947
  80. 80. Parajuli P, Mittal S. The role of IL-17 Giloma. J Spine and Neursurgey. 2013;suppl.1:S1–004.doi.10.4172/2325-9701.s1-004
  81. 81. Fietcher JM, Moran B, Petrasca A, Smith CM. IL-17 in inflammatory skin diseases psoriasis and hidradenitis suppurativa. Clin. Exp. Immunol. 2020;201(2):121–134
  82. 82. Kaur R, Rawat KA, Kumar S, Aadi W, Akhtar T, Narang T, Dimple C. Association of genetic polymorphism of IL-17A and IL-17F with susceptibility of psoriasis. India J. Med. Res. 2018;148(4):422–426
  83. 83. Blauvelt A, Chiricozzi A. The immunologic role of IL-17 in psoriasis and psoriatic arthritis pathogenesis. Clin. Rev. Allergy & Immunol. 2018;55(3):379–390
  84. 84. Wu D, Yang XO. Th-17 responses in cytokine storm of COVID-19: an emerging target of Jak2 inhibitor Fedratinib. J. Microbiol. Infect. 2020;53(3)368–370
  85. 85. Raucci F, Mansour AA, Casillo MG, Saviano A, et al. Interleukin-17 (IL-17A), a key molecule of innate and adaptive immunity and its potential involvement in COVID-19-related thrombotic and vascular mechanisms. Autoimun. 2020;19(7):102572
  86. 86. Shibabaw T. Inflammatory cytokine: IL-17A signaling pathway in patients with COVID-19 and current treatment strategy. J. Inflamm. Res. 2020;13:673–680
  87. 87. Bulat V, Situm M, Azadajic MD, Likic R. Potential role of IL-17 blocking agents in treatment of COVID-19. Br. J. Clin. Pharmacol. 2020;Jul 5.doi.org/10.1111/bcp.14437
  88. 88. Canavan TN, Elmets CA, Cantrell WL, Evans JM, Elewski BE. Anti-IL-17 medications used in the treatment of plaque psoriasis and psoriatic arthritis: A comprehensive review. Am. J. Clin. Dermatol. 2016;17(1):33–47
  89. 89. Yin Y, Wang M, Wu J. Efficacy and safety of IL-17 inhibitors for the treatment of ankylosing spondylitis: a systemic review and meta-analysis. Arthritis Res. & Therapy. 2020;22(111).Doi..org/10.1186/s13075-020-02208-w
  90. 90. Pacha O, Sallman MA, Evans SE. COVID-19: a case for inhibiting IL-17? Nature Reviews Immunol. 2020;20:345–346
  91. 91. Shalom-Barak T, Quach J, Lotz M. Interleukin-17-induced gene expression in articular chondrocytes is associated with activation of mitogen-associated protein kinases and NF-kB. J. Biol. Chem. 1998;42(16):27457–27473
  92. 92. Subramaniam SV, Pearson LT, Adunyah SE. Interleukin-17 induces rapid tyrosine phosphorylation and activation of raf-kinase in human monocytic progenitor cell line U937.Biochem. Biophys. Res. Commun. 1999;259(1):172–177
  93. 93. Subramaniam SV, Cooper RS, Adunyah SE. Evidence for the involvement of JAK/STAT pathway in the signaling mechanism of interleukin-17. Biochem. Biophys. Res. Commun. 1999;262:14–19
  94. 94. Lann M, Lötvall J, Fan Chun K, Lindén A. IL-17-induces release in human bronchial epithelial cells in vitro: role of mitogen-activated protein (MAP) kinases. British J. Pharmacol. 2001;133:200–206
  95. 95. Chen Y, Kijstra A, Chen Y, Peizeng Y. IL-17A stimulates the production of inflammatory mediators via Erk1/2, p38, PI-3K/Akt and NF-kB pathways in ARPE-19 cells. Molecular Vision. 2011;17:3072–3077
  96. 96. Zheng Q, Diao S, Wang Q, Zhu C, et al. IL-17 promotes cell migration and invasion of glioblastoma cells via activation of PI-3K/Akt signaling pathway. A J. Cell Mol. Med. 2019;23(1):357–369
  97. 97. You T, Yihui Bi, Li J, Zhang M, Chen X, Zhang K. IL-17 induces reactive astrocytes and up-regulation of vascular endothelial growth factor (VEGF) through JAK/STAT signaling. Sci. Rep.7.2017:41779 doi.org/10.1038/srep41779
  98. 98. Amatya N, Garg AV, Gaffen S. IL-17 Signaling: The Yin and the Yang. Trends in Immunol. 2017;39(5)310–322
  99. 99. Swaidani S, Liu C, Zhao J, Bulek K, Li X. TRAF Regulation of IL-17 signaling. Front Immunol. 2019;doi.org/10.3389/fimmu.2019.0129
  100. 100. Li X, Bechara R, Zhao J, McGeachy, MJ, Gaffen S. IL-17 receptor-based signaling and implications for diseases. Nature Immunol. 2019;20:1594–1602
  101. 101. Wang T, Liu Y, Zou, J-F, Chen Z-S. Interleukin-17 induces human alveolar epithelial to mesenchymal cell transition via TGF-beta-1 mediated Smad2/3 and ERK-1/2 activation. PLOS ONE2017;12(9): eO183972.doi.10.137/journal.pone.0183972
  102. 102. Weng C-H, Li Yi-J, Wu H-H, Liu S-H, Hsu H-H, Chen Y-C, et al.Interleikin-17A induces renal fibrosis through ERK and Smad signaling pathways. Biomed. Pharmacother. 2020;123:109741.doi.10.1016/j.biopha.2019.109741
  103. 103. Fuqua CF, Akomeah R, Price JO, Adunyah, SE. Evidence for the involvement of Erk1/2 in IL-21-induced cytokine production in leukemia cells and human monocytes. Cytokine. 2008;44:101–107
  104. 104. Phizicky EM. Protein-protein interactions: methods of detection and analysis. Microbil.Rev.1995;59:94–123
  105. 105. Golemis E. Protein-protein interactions: a molecular cloning manual. Cold Spring Harbor (NY). 2002;Cold Spring Harbor Laboratory Press. p ix,682
  106. 106. de Totero TD. Meazza R, Simona Zupo S, Cutrona G, Matis S, Colombo M, et al. Imterleukin-17 21 receptor (IL-21R) is up-regulated by CD40 triggering and mediates proapoptotic signals in chronic lymphocytic leukemia B cells. Blood. 2006;107:3708–3715
  107. 107. Dignam JD, Lebovitz RM, Roeder RG. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 1983;11:1475–1489
  108. 108. Lafarge S, Hamzeh-Cognasse H, Charvarin P, Genin C, Garraaud O, Cognasse F. Flow cytometry technique to study intracellular signals NF-kappaB and STAT3 in peripheral blood mononuclear cells. BMC Molecular Biology. 2007;8(64)1–9
  109. 109. Jiang H, Yu j, Guo H, Song H, Chen S. Upregulation of survivin by leptin/STAT3 signaling in MCF-7 cells. Biochem. Biophys. Res. Commun. 2008;368(1):1–5
  110. 110. Berridge MV, Herst PM, Tan AS. Tetrazolium dyes as tools in cell biology: New insights into their cellular reduction. Biotechnology Annual Rev. 2005:127–150
  111. 111. Namura S, Zhu J, Fink K, Endres M, Srinivasan A, Tomaselli KJ, et al. Activation and cleavage of Caspase-3 in apoptosis induced by experimental cerebral ischemia. Journal. Neuroscience. 1998;18(10):3659–3668
  112. 112. Hague A, Manning AM, Hanlon KA, Huschtscha LI, Dart D, Paraskeva C. Sodium butyrate induces apoptosis in human colonic tumor cell lines in a P53-independent pathway: implications for the possible role in dietary fiber in the prevention of large-bowel cancer. Int. J. Cancer. 1993;55(3):498–505
  113. 113. McCubrey WA, May WS, Duronio V, Mufson A. Serine/Threonine phosphorylation in cytokine signal transduction. Leukemia. 2000;14(1):9–21
  114. 114. Condorelli F, Salomoni P, Cotteret S, Cesi, V, Srinivasa M, et al. Caspase enhances the apoptosis-inducing effects of BAD. Mol. Cell Biol. 2001;21(9):3025–3036
  115. 115. Kanda N, Seno H, Konda Y, Murusawa H, Kanai M, et al. STAT3 is constitutively activated and supports cell survival in association with surviving expression in gastric cancer cells. Oncogene. 2004;23:4921–4929
  116. 116. La Fortezza M, Schenk M, Cosolo A, Kolybaba A, Grass I, Classen A-K. JAK’STAT signaling pathway mediates cell survival in response to tissue stress. Development. 2016;143:2907–2919
  117. 117. Nave BT, Ouvens M, Withers DJ, Alessi DR, Shepherd PR. Mammalian target of rapamycin is a direct target for protein kinase B: Identification of a convergence point for opposing effects of insulin and amino-acid deficiency on protein translation. Biochem J. 1999;344(Pt.2):427–431
  118. 118. de Rham C, Ferrari-Lacraz S, Jendy S, et al. The proinflammatory cytokines IL-2. IL-15 and IL-21 modulate the repertoire of mature human natural killer cell receptors. Arthritis Res. Ther. 2007;9:R125.doi.org/10.1186/ar2336
  119. 119. Liao W, Lin J-X, Leonard W. IL-2 Family of Cytokines: New insights into the complex roles of IL-2 as a broad regulator of helper T cell differentiation. Curr. Opin. Immunol. 2011;23(5):598–604
  120. 120. Ihle JN. Immunological regulation of hematopoietic stem cell function by IL-3 and its role inkeukemogenesis.IL-3 The Panspecific Hematopoietin. 1988;pages 127–161
  121. 121. Sieff CA, Niemeyer CM, Nathan DG, Ekern SC, Yang YC, Wong G, Clark SC. Stimulation of human hematopoietic colony formation by recombinant gibbon multi-stimulating factor or Interleukin-3.J. Clin. Invest. 1987;80(3):818–829
  122. 122. Couper KN, Blount DG, Riley EM.IL-10: The master regulator of immunity to infection.J.Immunol. 2008;180(9):5771–5777
  123. 123. Li X, Lu P, Li B, Zhang W, Yang R, Chu Y, Luo K. Interleukin-2 and Interleukin-10 function synergistically to promote CD+T cell cytotoxicity, which is suppressed by regulatory T cells in breast cancer. Int. J. of Biochem. & Cell Biol. 2017;87:1–7
  124. 124. Inoue S, Unsinger J, Davis, CG, Muenzer JT, et al. IL-15 prevents apoptosis, reverses innate and adaptive immune dysfunction, and improves survival in sepsis. J. Immunol. 2010;18(3):1401–1409
  125. 125. Waldmann TA. The shared and contrasting roles of Interleukin-2 (IL-2) and Inteleukin-15 (IL-15) in the life and death of normal and neoplastic lymphocytes: implications for cancer therapy. Cancer Immunol. Res. 2015;3(3):219–227
  126. 126. Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead Transcription Factor. Cell. 1999;96:857–868
  127. 127. Trizzino M, Zucco A, Deliard S, Wang F, et al. EGR-1 is a gatekeeper of inflammatory enhancers in human macrophages. Science Advances. 2021;7(3):eaaz8836 doi.10.1126/sciadv,aaz8836
  128. 128. Yang H, Wang L, Zhao J, et al. TGF-beta-activated smad3/4 complex transcriptionally upregulates N-cadherin expression in non-small cell lung cancer. Lung Cancer. 2015;87(3):249–257
  129. 129. Williams JCM. Evidence of the involvement of specific Src kinases and phospholipase D in IL-17A signaling mechanisms in U937 leukemia cells [thesis]. Biochemistry and Cancer Biology Department: Mehary Medical College, Nashville, TN; 2004
  130. 130. Davis WL, Jr. An in vitro examination of the effects of Interleukin-17 in proliferation and differentiation of U937 leukemia cells [thesis]. Biochemistry and Cancer Biology Department: Mehary Medical College, Nashville, TN; 2004
  131. 131. Wu D, Yang XO. TH17 responses in cytokine storm of COVID-19: An emerging target of Jak2 inhibitor Fedratinib. J. MicroBiol. Immunol. & Infect. 2020;53:368–370

Written By

Samuel Evans Adunyah, Richard Akomeah, Fareed K.N. Arthur, Roland S. Cooper and Joshua C.M. Williams

Submitted: 28 October 2020 Reviewed: 04 February 2021 Published: 30 March 2021