Endometriosis is a chronic inflammatory disease in which endometrial-like tissue grows ectopically, resulting in pelvic pain and infertility. IL-23 is a key contributor in the development and differentiation of TH17 cells, driving TH17 cells toward a pathogenic profile. In a variety of inflammatory and autoimmune disorders, TH17 cells secrete proinflammatory cytokines, including IL-17, contributing to disease pathophysiology. Our studies and others have implicated IL-17 and TH17 cell dysregulation in endometriosis, which is associated with disease severity. In this article, we address whether IL-23–driven TH17 cells contribute to cardinal features of lesion proliferation, vascularization, and inflammation in endometriosis using patient samples, representative cell lines, and our established mouse model of endometriosis. The results indicated dysregulated expression of key genes in the IL-23/TH17 axis in patient ectopic and eutopic endometrial samples and increased IL-23 protein in patient plasma compared with controls. In vitro studies using primary human TH cells determined that rIL-23 mixture treatment increased pathogenic TH17 cell frequency. Similarly, rIL-23 treatment of cell lines (12Z cells, EECCs, HUVECs, and hESCs) representative of the endometriotic lesion microenvironment increased cytokines and growth factors, which play a role in lesion establishment and maintenance. In a syngeneic mouse model of endometriosis, rIL-23 treatment altered numbers of myeloid and T cell subsets in peritoneal fluid and increased giant cells within the lesion. Lesions from rIL-23–treated mice did not reveal significant alterations in proliferation/vascularization, although trends of increased proliferation and vascularization were observed. Collectively, these findings provide insights into the impact of the IL-23/TH17 axis on local immune dysfunction and broadly on endometriosis pathophysiology.

Endometriosis is characterized by the growth of endometrial-like tissue in ectopic sites and is commonly associated with dysmenorrhea, dyspareunia, chronic pelvic pain, and infertility (1, 2). Although endometriosis incidence and prevalence are difficult to establish due to the invasive nature of disease diagnosis, it is estimated that endometriosis afflicts 10% (190 million) of reproductive-aged women and an unknown percentage of gender-diverse individuals (3–5). Although the origin and pathogenesis of endometriosis remain largely unknown, we and others postulate that immune dysfunction promotes ectopic lesion growth and survival by impeding lesion clearance (1, 6). Specifically, proinflammatory IL-17 is well -established to contribute to the pathogenesis of several inflammatory diseases, including endometriosis (7–10). Indeed, we have demonstrated that endometriotic lesions produce IL-17 and that IL-17 is significantly increased in patient plasma compared with controls (10). Additionally, surgical removal of lesions significantly reduces IL-17 plasma concentration, suggesting that endometriotic lesions are likely a predominant source of circulating IL-17 (10). IL-17 is largely produced by TH17 cells, which also coproduce IL-21 and IL-22, contributing to tissue inflammation and angiogenesis (10, 11). TH17 cells act pathogenically in various inflammatory and autoimmune disorders, including psoriasis and rheumatoid arthritis, by secreting proinflammatory cytokines, which promote immune cell accumulation at inflammatory sites (11, 12). However, TH17 cells are highly plastic, adopting differing fates in response to environmental cues, such as cytokines/chemokines (8, 13, 14). In fact, TH17 cells may also play a beneficial (nonpathogenic) role against opportunistic pathogens and promote and/or build intestinal barrier integrity (11, 14–16). Specifically, IL-17A+IL-10 TH17 cells are termed “pathogenic” (pTH17), whereas IL-17A+IL-10+ are termed “nonpathogenic” (npTH17), promoting an anti-inflammatory/immunosuppressive phenotype (12, 17).

Studies show TH17 differentiation is initiated from naive CD4+ T cells by a combination of cytokines TGF-β1, IL-1β, IL-6, IL-21, and IL-23 (11, 18, 19). IL-17 and IL-21 secreted by developing TH17 cells further drive TH17 cell fate and early proliferation, whereas IL-23 is fundamental for TH17 cell expansion and phenotypic stabilization (11, 18–21). When exposed to IL-23, developing TH17 cells produce significantly reduced anti-inflammatory IL-10, further evoking a proinflammatory, pathogenic phenotype (17, 18). In fact, TH17 cells are incapable of inducing autoimmune disease (18) and unable to upregulate proinflammatory cytokines without exposure to IL-23 (17). Hence, complete acquisition of pathogenic potential in TH17 cells is likely mediated by IL-23. This is supported by reports that IL-23–specific p19 subunit neutralization ameliorates/protects from autoimmune disease, including experimental autoimmune encephalomyelitis and autoimmune colitis, whereas loss of downstream IL-17 or IL-17 receptor A (IL-17RA) failed to reduce disease emergence/severity (11, 22). Thus, if IL-23 is a key contributor in driving TH17 cells to a pathogenic profile, IL-23 may govern the balance between TH17 and T-regulatory cells (TREGs), which is critical to avoid immune dysfunction and maintain immune homeostasis (23–25).

Some theorize that in endometriosis, a tolerant, anti-inflammatory peritoneal microenvironment initially allows for early lesion development/establishment, preventing lesion clearance (26), although later lesion growth and maintenance are supported by a proinflammatory microenvironment, promoting inflammation, angiogenesis, and fibrosis (26). This scenario suggests a skewed TH17/TREG axis toward a tolerant (TREG-dominant) microenvironment as lesions initially establish, followed by a proinflammatory (TH17-dominant) microenvironment allowing for lesion survival, because downstream TH17 cytokines, such as IL-17, are well known to promote lesion angiogenesis, proliferation, and inflammation (10, 27, 28). Results from our laboratory support this theory, finding decreased TH17 cells in patient menstrual effluent compared with controls (29). Based on Sampson’s theory of retrograde menstruation, these data provide insights into early events in endometriosis lesion establishment, because decreased TH17 cells in patient menstrual effluent may influence the local immune microenvironment and facilitate initial lesion establishment following retrograde menstruation. Although further studies are warranted, this theory is also supported by findings that TH17 cells were significantly increased in the peritoneal fluid (PF) of patients with severe (III/IV stage) endometriosis compared with early (I/II stage) endometriosis (13).

IL-23, a heterodimeric cytokine largely secreted by activated dendritic cells and macrophages (30), has been established to dysregulate the TH17/IL-17 axis in a variety of inflammatory and autoimmune disorders, including psoriasis and rheumatoid arthritis (19, 31, 32). Because evidence suggests dysregulated levels of TH17 cells (13, 24, 33) and IL-17 (10, 28) in endometriosis, as well as an association of TH17 cells with increasing disease severity (13), IL-23 is likely regulating the TH17/IL-17 axis in endometriosis to exacerbate disease. Although downstream IL-17 has been previously evaluated for its therapeutic potential and role in various autoimmune and chronic inflammatory diseases (32), the role of upstream IL-23 has yet to be fully evaluated in endometriosis pathophysiology. Unlike targeting IL-17, targeting IL-23 reduces proinflammatory IL-17 production by pTH17 cells, while preserving overall IL-17 production from other IL-17–producing cells, ameliorating safety concerns around eliminating protective effects of IL-17 (32). This study depicts a dysregulated IL-23/TH17 axis in endometriosis patient samples, which was replicated in vitro. We also provide insights using an endometriosis mouse model, revealing IL-23 to significantly alter the local immune microenvironment and TH17/TREG axis, indicating a plausible role of IL-23 in endometriosis pathophysiology and highlighting IL-23 as a potential nonhormonal therapeutic target for endometriosis patients.

The ethics for the use of human samples in this study was approved by the Health Sciences Research Ethics Board at Kingston Health Sciences Center, Queen’s University (Kingston, ON, Canada), the Greenville Health System (Greenville, SC, USA), the University of North Carolina at Chapel Hill (Chapel Hill, NC, USA), and Wake Forest Baptist Health (Winston-Salem, NC, USA). Written, informed consent was attained from endometriosis patients and healthy controls prior to the collection and use of clinical of samples. Animal studies were approved by the Queen’s University Animal Care Committee at Queen’s University (Kingston, ON, Canada).

Endometriosis patients undergoing excision surgery due to endometriosis-associated infertility and/or pelvic pain were included in this study (Table I). Healthy, fertile controls without endometriosis were recruited for endometrial biopsy and plasma collection prior to elective tubal ligations. All patients and controls recruited for this study were free of hormonal therapy for a minimum of 3 mo prior to sample collection.

Table I.
Endometriosis patient and control study parameters
ParametersEndometriosisControls
Number of patients/controls n = 42 n = 29 
Age, y 30.8 ± 4.1 26.1 ± 4.1 
Race 
 African American n = 1 n = 4 
 Asian n = 2 n = 2 
 Caucasian n = 39 n = 23 
Pathology report 
 Menses n = 2 n = 0 
 Proliferative n = 13 n = 10 
 Early Secretory n = 5 n = 3 
 Mid Secretory n = 7 n = 15 
 Late Secretory n = 15 n = 1 
Stage of endometriosis  
 I to II n = 28 
 III to IV n = 9 
 Not indicated n = 5 
ParametersEndometriosisControls
Number of patients/controls n = 42 n = 29 
Age, y 30.8 ± 4.1 26.1 ± 4.1 
Race 
 African American n = 1 n = 4 
 Asian n = 2 n = 2 
 Caucasian n = 39 n = 23 
Pathology report 
 Menses n = 2 n = 0 
 Proliferative n = 13 n = 10 
 Early Secretory n = 5 n = 3 
 Mid Secretory n = 7 n = 15 
 Late Secretory n = 15 n = 1 
Stage of endometriosis  
 I to II n = 28 
 III to IV n = 9 
 Not indicated n = 5 

Patients and controls were free of hormonal therapy for a minimum of 3 mo prior to sample collection. The data are represented as means ± SD, unless indicated otherwise.

Total RNA was extracted from ectopic (n = 9) and eutopic (n = 9) endometrium of patients, and normal healthy endometrium (n = 9) using a total RNA purification kit (17200, Norgen Biotek Corp., Canada) as per the manufacturer’s protocol. RNA quality was assessed and cDNA synthesized as previously detailed (34). RT-qPCR was conducted with custom array plates (330171, Qiagen, Canada) using the LightCycler 480 real-time PCR system (Roche Molecular Systems Inc., Switzerland) with RT2 SYBR Green qPCR Mastermix (330501, Qiagen, Canada) and 200 ng cDNA, as per the manufacturer’s protocol. Data analysis was conducted using the ΔΔCt method, and relative quantification was performed by geometric averaging of Gapdh and Actb reference genes. The custom RT-qPCR array was designed to target 11 genes of interest in the IL-23/TH17/IL-17 axis: IL23A, IL12B, IL12A, IL23R, IL21, IL22, IL17A, IL17F, IL17RA, IL6, and TGFB1.

IL-23 concentration was measured in endometriosis patient tissue protein extracts (ectopic [n = 10] and eutopic [n = 16] endometrium) and patient plasma (n = 13) as compared with healthy controls (n = 19), as well as between stratified patient plasma samples (n = 7) over three time points using an IL-23 human ELISA kit (ab64708, Abcam, UK), as per the manufacturer’s protocol. A total of 20 mg of tissues were homogenized in PowerBead tubes (13113-50, Qiagen, Canada) containing protease inhibitor mixture (535140, Sigma-Aldrich, Canada) and tissue protein extraction reagent (78510, ThermoFisher, Canada). Protein concentration was determined using a Pierce BCA protein assay kit (23225, ThermoFisher) as per kit protocol. The samples were normalized to the lowest concentration and stored at −80°C prior to analysis. ELISA results were read in a SpectraMax iD5 microplate reader (Molecular Devices, USA) at an absorbance of 450 nm and a reference wavelength of 620 nm.

An endometrioma tissue microarray was constructed using patient tissue samples from Kingston General Hospital (Kingston, ON, Canada), as previously described (35). Matched patient ectopic and eutopic tissues (n = 17), as well as normal endometrium controls (n = 10), were subjected to immunohistochemistry (IHC) staining using a polyclonal IL-23 Ab (1:200; ab45420, Abcam, UK), and analysis using HALO image analysis software (Indica Labs, USA) was conducted as previously outlined (10, 34). Briefly, Ag retrieval was conducted using citrate buffer for 30 min prior to staining with primary anti–IL-23 Ab (30 min). Following this, samples were incubated with a biotinylated secondary polyclonal goat anti-rabbit IgG Ab (1:500; DakoCytomation, Denmark), stained with diaminobenzidine chromogen (DakoCytomation), and counterstained with Harris hematoxylin (Fisher Scientific, Canada) (10).

A total of 30 mL of whole blood was collected from healthy volunteers into BD Vacutainer tubes (CABDL366643L, VWR, USA). PBMCs were isolated using Lymphoprep (07811, StemCell Technologies, Canada) and SepMate-50 PBMC isolation tubes (85460, StemCell Technologies, Canada). Primary CD4+ T cells were isolated from PBMCs using an EasySep Human CD4+ T cell isolation kit (17952, StemCell Technologies, Canada) as per the manufacturer’s instructions. Primary CD4+ T cells were seeded at 1 × 106 cells/well in 24-well plates and maintained in X-VIVO-15 medium (CA12001-988, VWR). Four treatment groups were used: 1) mixture and activation, 2) activation alone, 3) mixture alone, and 4) control medium. Activation consisted of 10 µg/ml plate-bound anti-human CD3 mAb (300437) and 1 µg/ml soluble anti-human CD28 mAb (302902) purchased from BioLegend (USA). Based on previously published protocols (36–39), the TH17 mixture consisted of 5 ng/ml recombinant human TGF-β1 (rhTGF-β1; 240-B-002), 12.5 ng/ml rhIL-1β (201-LB-005), 25 ng/ml rhIL-6 (206-IL-010), 25 ng/ml rhIL-21 (8879-IL-010), 25 ng/ml rhIL-23 (1290-IL-010), 500 ng/ml anti–IFN-γ (506531, BioLegend), and 500 ng/ml anti-IL-4 (500837, BioLegend) purchased from R&D Systems unless stated otherwise. Primary CD4+ T cells were treated in triplicate and incubated at 37°C with 5% CO2 for 4 d, as per established protocols (36, 37). On day 4, the cells were restimulated with 50 ng/ml of PMA (74042, StemCell Technologies, Canada) and 250 ng/ml of ionomycin (73722, StemCell Technologies, Canada) in combination with 2 µl/ml of protein transport inhibitor mixture (brefeldin A and monensin; 00-4980-93, ThermoFisher) and incubated at 37°C with 5% CO2 for 5 h prior to analysis. Supernatant was analyzed using a commercially available multiplex cytokine/chemokine analysis (HD48-plex, EveTechnologies, Canada). To examine cell fate following in vitro polarization, flow cytometry was conducted on day 4.

An immortalized human endometriotic epithelial cell line derived from epithelial cells of peritoneal endometriosis (12Z cells; kindly gifted by Dr. Anna Starzinski-Powitz), endometrial epithelial carcinoma cells (EECCs; CRL-1671, ATCC), HUVECs (CRL-1730, ATCC), and human endometrial stromal cells (hESCs; T0533 ABM, Canada) were used in this study. EECCs were maintained in DMEM (D6429, Sigma-Aldrich, Canada) supplemented with 10% FBS (97068-085, VWR) and 1% penicillin and streptomycin (15140122, ThermoFisher). Cell line cultivation and 12Z, HUVEC, and hESC maintenance were conducted as previously described (34).

12Z cells, EECCs, HUVECs, and hESCs were seeded at 3 × 105 cells/well onto 6-well plates and rested for 24 h prior to stimulation with PBS or rhIL-23 (1290-IL-010, R&D Systems, USA) at concentrations of 1, 10, 50, and 100 ng/ml. The cells were treated in triplicate and incubated at 37°C with 5% CO2 for 24 h, and supernatant was analyzed via multiplex cytokine array (HD48-plex, EveTechnologies, Canada).

12Z cells, EECCs, HUVECs, and hESCs were seeded onto 96-well plates at 5 × 103 cells/well and rested for 24 h. The cells were then treated in triplicate for 24 h with varying concentrations of rhIL-23 for analysis via a WST-1 proliferation assay using WST-1 reagent (0501594400, Sigma-Aldrich, Canada) and an apoptosis assay using Caspase-Glo 3/7 reagent (G8091, Promega, Canada), as per the manufacturer’s protocol. The results of WST-1 and Caspase-Glo assays were acquired as previously outlined (34).

To determine the influence of IL-23 on angiogenesis, a tubulogenesis assay was conducted using IBIDI µ-plates (81506, IBIDI, Germany), as per the manufacturer’s protocol. Briefly, each µ-plate well was coated with 10 µl of Matrigel (353230, Corning, USA) and incubated for 1 h in a humid chamber at 37°C with 5% CO2. HUVECs were then seeded at 1 × 104 cells/well in triplicate on top of polymerized Matrigel in 50 µl of medium containing either VEGF (25 ng/ml), PBS (vehicle), or rhIL-23 (1, 10, 50, or 100 ng/ml). The cells were incubated for 4 h at 37°C with 5% CO2 prior to image acquisition. The images were captured at 4× objective using an Olympus CKX41 microscope (Olympus Life Science, USA) with a Lumenera Infinity 1-3 microscope camera. The images were analyzed for tube formation using WIMASIS software (WimTube: tube formation assay image analysis solution) (40).

Spleens from female C57BL/6 mice (n = 6) were collected and mechanically digested through 70-µm strainers before centrifugation at 400g for 5 min at 4°C, and the pellets were resuspended and counted. An EasySep isolation kit (19765, StemCell Technologies, Canada) was used to isolate naive CD4+ T cells, following the manufacturer’s protocol. CD4+ T cells were washed with 2% FACS buffer (PBS with 2% FBS) prior to resuspension in TexMACS complete medium (130-097-196, Miltenyi Biotec, USA), supplemented with 10% FBS, 1% penicillin and streptomycin, and 0.1 mM 2-ME (31350-010, ThermoFisher). The cells were counted and seeded at 2.5 × 105 cells/well in 96-well plates. Four treatment groups were used: 1) mixture and activation, 2) mixture and activation supplemented with an additional 10 ng/ml recombinant mouse (rm)IL-23, 3) 10 ng/ml rmIL-23 (no mixture) and activation, and 4) medium and activation (control). Activation consisted of 5 µg/ml plate-bound anti-mouse CD3ε mAb (100340) and 5 µg/ml soluble anti-mouse CD28 mAb (102116) purchased from BioLegend. A commercially available CytoBox TH17 mouse mixture (130-107-758, Miltenyi Biotec) was used for polarization of murine CD4+ T cells in vitro, as per the manufacturer’s instructions. To examine cell fate, flow cytometry was conducted on day 5 following restimulation for 5 h with 50 ng/ml PMA, 750 ng/ml ionomycin, and 2 µl/ml protein transport inhibitor mixture (last 4 h).

Seven-to-eight-week-old female C57BL/6 mice (n = 12; Charles River Laboratories, USA) were cohoused in conventional housing at three or four animals per cage. Endometriosis was induced (n = 12) as previously detailed (34). Briefly, uterine horns were harvested from donor mice (n = 3), and an epidermal punch tool was used to obtain 3-mm endometrium fragments. Recipient mice were anesthetized with 3.5% isoflurane, and a small incision was made in the abdominal wall to access the peritoneum, onto which two uterine fragments (3 mm3) were adhered using Vetbond tissue adhesive (1469SB, 3M, USA). Lesions were allowed to establish for 1 wk postoperative. Sham surgery was conducted (n = 12), in which mice underwent surgery, although no uterine fragments were adhered to the peritoneum. Postsurgical care was conducted as outlined (28). To examine the effects of IL-23 in vivo, both endometriosis and sham mice were treated with i.p. injections of 1 µg of rmIL-23 (n = 6/group; 589006, BioLegend) or PBS for controls (n = 6/group), three times a week for 3 weeks (Fig. 6A). The mice were euthanized 21 d postoperative by 5% isoflurane inhalation followed by cervical dislocation. PF, endometriosis-like lesions, uterine horns, and spleens were collected, processed, and stored prior to analysis as previously detailed (34). For flow cytometry, PF and splenocytes were thawed and resuspended in 10% FACS. The cells were centrifuged at 300g for 5 min at 4°C, and supernatant was decanted before resuspension in 100 µg/ml DNAse I (10104159001, Sigma-Aldrich, Canada), as per the manufacturer’s protocol. The cells were neutralized with 10% FACS, centrifuged at 300g for 5 min at 4°C, and supernatant was decanted before resuspension in 2% FACS for flow cytometry.

Paraformaldehyde-fixed, paraffin-embedded tissue blocks were sectioned at 4-µm thickness and deparaffinized using Citra Solv and rehydrated in graded concentrations of ethanol. Following Ag retrieval for 20 min using citrate buffer and blocking of endogenous peroxidase activity, sections were stained with polyclonal Abs for anti-mouse Ki67 (1:1500; ab15580, Abcam, UK) and anti-mouse CD31 (1:100; 77699S, New England Biolab, USA) to assess proliferation and vascularization, respectively, and digitally scanned for analysis as previously described (34). A computer-generated algorithm for Ki67 and CD31 was used to digitally analyze lesions. Custom trained cytonuclear and area quantification modules were used to quantify Ki67 and CD31 staining, respectively. Standard H&E staining was conducted as previously explained (41).

To assess whether TH17 cells may be primed in vivo during lesion development, endometriosis was surgically induced in recipient mice (n = 16) as described (34). Endometriosis-induced mice either received daily i.p. injections of a TH17 priming mixture (n = 8; 0.5 µg rmIL-6 [575704, BioLegend] and 1 µg rmTGF-β1 [763104, BioLegend]) or rested (n = 8) from days 0 to 3 prior to daily i.p. injections of 1 µg rmIL-23 (n = 4/group) or PBS (n = 4/group) for 10 d (Fig. 8A). The mice were euthanized 14 d postoperative, and PF, spleen, and endometriosis-like lesions were collected. Lesions were snap-frozen in liquid nitrogen prior to storage at −80°C for later analysis, and PF was processed for cryopreservation, as detailed (34), for later analysis via flow cytometry. The splenocytes were isolated as previously outlined (34) prior to subjection to ACK lysis buffer (A1049201, ThermoFisher) per the manufacturer’s instructions. The splenocytes were neutralized with RPMI 1640 (11875085, ThermoFisher) and centrifuged at 400g for 5 min at 4°C prior to resuspension and counting. The samples were seeded in 24-well plates at 1 × 106 cells/well and incubated at 37°C with 5% CO2 with 50 ng/ml PMA and 500 ng/ml ionomycin for 4 h to stimulate intracellular cytokine production. After 1 h of incubation, 2 µl/ml of protein transport inhibitor mixture was added. Flow cytometry was conducted to examine cell fate.

Cell suspensions were counted and aliquoted (5 × 105 cells/sample) before staining with human TruStain FcX (422302) or TruStain FcX (101319) to block nonspecific Ag binding, as per the manufacturer’s instructions. All products were purchased from BioLegend unless otherwise stated. The cells were stained with extracellular Abs as detailed in Table II. To assess viability, human samples were stained with eBioscience fixability viability dye eFluor 780 (65-0865-18, ThermoFisher), whereas murine samples were stained with Zombie green fixable viability dye (423111). All cells were then fixed and permeabilized using the eBioscience Foxp3/transcription factor staining buffer set (00-5523-00, ThermoFisher), as per the manufacturer’s instructions. Following permeabilization, the cells were stained with intracellular Abs (Table II). The cells were washed with 2% FACS, and the data were acquired using CytoFLEX S (Beckman Coulter, USA) and analyzed via FlowJo version 10 software (BD, USA). Fluorescent minus one controls were used appropriately to determine gating strategy. To set a viability gate, an aliquot of heat-killed cells was combined 1:1 with live cells and stained with viability dye.

Table II.
List of extracellular and intracellular Abs used for flow cytometric analyses
AbFluorophore(s)Verified ReactivityCloneCatalog NumberVendor
CD45 PB Human HI30 304029 BioLegend 
 APC/Cy7 Mouse 30-F11 103115 
CD3 Alexa Fluor 700 Mouse 17A2 100215 BioLegend 
CD4 PE/Cy7 Human RPA-T4 300512 BioLegend 
Mouse GK1.5 100421 
CD11b Alexa Fluor 700 Mouse, human, cynomolgus, rhesus monkey M1/70 101222 BioLegend 
CD25 Brilliant Violet 650 Mouse PC61 102038 BioLegend 
CD196 (CCR6) FITC Human, cynomolgus, rhesus monkey G034E3 353412 BioLegend 
PE/Dazzle 594 Mouse 29-2L17 129822 
ROR-γt APC Human, mouse, rhesus monkey AFKJS-9 17-6988-82 ThermoFisher 
Foxp3 PE Mouse MF-14 126404 BioLegend 
IL-17A PE Human BL168 512306 BioLegend 
Brilliant Violet 421 Mouse TC11-18H10.1 506926 
IL-10 Brilliant Violet 605 Mouse JES5-16E3 505031 BioLegend 
IFN-γ PE Mouse XMG1.2 505807 BioLegend 
AbFluorophore(s)Verified ReactivityCloneCatalog NumberVendor
CD45 PB Human HI30 304029 BioLegend 
 APC/Cy7 Mouse 30-F11 103115 
CD3 Alexa Fluor 700 Mouse 17A2 100215 BioLegend 
CD4 PE/Cy7 Human RPA-T4 300512 BioLegend 
Mouse GK1.5 100421 
CD11b Alexa Fluor 700 Mouse, human, cynomolgus, rhesus monkey M1/70 101222 BioLegend 
CD25 Brilliant Violet 650 Mouse PC61 102038 BioLegend 
CD196 (CCR6) FITC Human, cynomolgus, rhesus monkey G034E3 353412 BioLegend 
PE/Dazzle 594 Mouse 29-2L17 129822 
ROR-γt APC Human, mouse, rhesus monkey AFKJS-9 17-6988-82 ThermoFisher 
Foxp3 PE Mouse MF-14 126404 BioLegend 
IL-17A PE Human BL168 512306 BioLegend 
Brilliant Violet 421 Mouse TC11-18H10.1 506926 
IL-10 Brilliant Violet 605 Mouse JES5-16E3 505031 BioLegend 
IFN-γ PE Mouse XMG1.2 505807 BioLegend 

PB, Pacific blue.

GraphPad Prism9 (USA) was used for all statistical analyses. When appropriate, an unpaired, nonparametric Student t test with Mann–Whitney correction was used to analyze two groups, and a one-way ANOVA with Tukey post hoc test was used to compare three or more groups. Error bars signify SD, and statistical significance was denoted by a p value ≤ 0.05.

To assess endometriotic lesions as a source of IL-23 and gene expression variations between matched patient ectopic and eutopic tissues, a RT-qPCR array was designed to target 11 key genes known to play a role in the IL-23/TH17 axis (Fig. 1). We discovered IL23R, IL17A, IL17F, IL21, IL22, IL12B, and TGFB1 were significantly upregulated in patient ectopic tissues compared with both matched eutopic tissues and controls (Fig. 1A, 1B). Notably, IL23R (IL-23 receptor) was also significantly upregulated in patient eutopic tissues compared with controls (Fig. 1C).

FIGURE 1.

Endometriosis patient tissue samples reveal significantly dysregulated expression of key genes in the IL-23/TH17 axis, and IL-23 protein was significantly increased in the plasma of patients as compared with healthy controls. (AC) Volcano plots depict differential mRNA expression of key genes in the IL-23/TH17 axis within patient ectopic tissues (n = 9) versus controls (n = 9; A), patient ectopic versus eutopic tissues (n = 9; B), and patient eutopic versus control endometrium (C). The horizontal gray line indicates a significance of p = 0.05, and dashed vertical lines indicate a fold change of ± 1, whereby upregulated genes are displayed to the right of the dashed vertical line at x = 1.0 (black data points). IL-23 protein was measured in patient plasma and tissues (eutopic and ectopic) compared with healthy controls via ELISA. (D) IL-23 concentration was significantly increased in endometriosis patient plasma (n = 13) as compared with controls (n = 19). (E) However, no significant differences were found when stratifying patient plasma (n = 7) by time points. (F) IL-23 protein was also detected in patient eutopic (n = 16) and ectopic tissues (n = 10), although no significant differences were found. The data are represented as means ± SD. Statistical analyses were conducted by a nonparametric Student t test with Mann–Whitney correction (D); a paired, nonparametric, one-way ANOVA with Friedman post hoc test (E); or a paired, nonparametric Student t test (Wilcoxon test; F). **p < 0.01.

FIGURE 1.

Endometriosis patient tissue samples reveal significantly dysregulated expression of key genes in the IL-23/TH17 axis, and IL-23 protein was significantly increased in the plasma of patients as compared with healthy controls. (AC) Volcano plots depict differential mRNA expression of key genes in the IL-23/TH17 axis within patient ectopic tissues (n = 9) versus controls (n = 9; A), patient ectopic versus eutopic tissues (n = 9; B), and patient eutopic versus control endometrium (C). The horizontal gray line indicates a significance of p = 0.05, and dashed vertical lines indicate a fold change of ± 1, whereby upregulated genes are displayed to the right of the dashed vertical line at x = 1.0 (black data points). IL-23 protein was measured in patient plasma and tissues (eutopic and ectopic) compared with healthy controls via ELISA. (D) IL-23 concentration was significantly increased in endometriosis patient plasma (n = 13) as compared with controls (n = 19). (E) However, no significant differences were found when stratifying patient plasma (n = 7) by time points. (F) IL-23 protein was also detected in patient eutopic (n = 16) and ectopic tissues (n = 10), although no significant differences were found. The data are represented as means ± SD. Statistical analyses were conducted by a nonparametric Student t test with Mann–Whitney correction (D); a paired, nonparametric, one-way ANOVA with Friedman post hoc test (E); or a paired, nonparametric Student t test (Wilcoxon test; F). **p < 0.01.

Close modal

Using ELISA, we found IL-23 protein was significantly increased in patient plasma compared with controls (Fig. 1D). Patient plasma was then stratified into preoperative, postoperative, and 3-mo post endometriosis lesion excision surgery time points to determine whether lesion presence alters circulating IL-23 levels. However, no significant differences were found between time points (Fig. 1E). IL-23 protein was also detected in patient ectopic lesion and eutopic samples, although no significant differences were revealed when comparing eutopic and ectopic tissues (Fig. 1F).

We aimed to depict spatial localization of IL-23 within a human tissue microarray (Fig. 2) containing matched patient ectopic (endometrioma) and eutopic tissue samples (n = 17; Fig. 2A, 2B) compared with control endometrium (n = 10; Fig. 2C). IL-23 protein expression was quantified using area quantification of percentage of positive stain of anti–IL-23 stain across the total core area (Fig. 2D) and independently within epithelial (luminal and glandular) and stromal tissue (Fig. 2E). Although there were no significant differences in IL-23 protein expression between matched patient (ectopic/eutopic) or control endometrium, IHC staining illustrates that IL-23 is predominately expressed in epithelial tissues compared with stroma of matched patient (ectopic/eutopic) and control endometrium.

FIGURE 2.

IL-23 protein is expressed in endometriosis lesions and is significantly increased in luminal and glandular epithelium as compared with stroma. (AC) Anti–IL-23–stained endometrioma tissue microarray of matched endometriosis (eutopic and ectopic, n = 17; A and B) and control endometrium (n = 10; C). (D and E) Area quantification of percentage of positive stain was calculated for total core area (D) and luminal and glandular epithelium as compared with stromal tissue (E). Statistical significance was assessed using a one-way ANOVA with Tukey post hoc test. ****p < 0.0001. Scanned IHC images were digitally analyzed using HALO imaging software (Indica Labs) and provided at 5× and 20× magnification. Scale bars, 100 µm and 50 µm, respectively.

FIGURE 2.

IL-23 protein is expressed in endometriosis lesions and is significantly increased in luminal and glandular epithelium as compared with stroma. (AC) Anti–IL-23–stained endometrioma tissue microarray of matched endometriosis (eutopic and ectopic, n = 17; A and B) and control endometrium (n = 10; C). (D and E) Area quantification of percentage of positive stain was calculated for total core area (D) and luminal and glandular epithelium as compared with stromal tissue (E). Statistical significance was assessed using a one-way ANOVA with Tukey post hoc test. ****p < 0.0001. Scanned IHC images were digitally analyzed using HALO imaging software (Indica Labs) and provided at 5× and 20× magnification. Scale bars, 100 µm and 50 µm, respectively.

Close modal

In accordance with literature, we sought to investigate IL-23 involvement in human pTH17 cell derivation using flow cytometry (Fig. 3A) and multiplex cytokine array. We found primary human CD4+ T cells treated in vitro with IL-23 mixture (rhIL-23, rhIL-21, rhTGF-β1, rhIL-1β, rhIL-6, anti–IFN-γ, and anti–IL-4) produced significantly increased the frequency and number of TH17 cells (Fig. 3B, 3C). We then aimed to determine how TH17 cells driven in the presence of IL-23 may influence the endometriotic microenvironment. Multiplex cytokine array results reveal that activated TH17 cells driven in the presence of IL-23 produce significantly increased IL-17A/IL-17F and decreased IL-10, suggesting a pathogenic phenotype (Fig. 3D, 3E). Activated TH17 cells driven in the presence of IL-23 also secreted significantly increased MCP-1 and CCL5/RANTES (Fig. 3F, 3G), as well as CXCL9 and macrophage-derived chemokine (MDC; data not shown).

FIGURE 3.

IL-23 mixture treatment significantly promotes the production of pathogenic TH17 cells from primary human naive CD4+ T cells and alters the production of factors involved in endometriosis lesion establishment and maintenance. Primary naive CD4+ T cells isolated from human PBMCs were treated in vitro in triplicate for 4 d with an IL-23 mixture and CD3/CD28 activation, CD3/CD28 activation alone, IL-23 mixture alone, or media. (A) Cells were restimulated with PMA/ionomycin and protein transport inhibitor mixture prior to supernatant collection and flow cytometric analysis for TH17 cells (CD45+CD4+CCR6+RORγt+IL17A+). (B and C) Primary CD4+ T cells treated with mixture and activation produced a significantly higher frequency of TH17 cells within the CD45+CD4+ cell population (B) and a number of TH17 cells per 100,000 events (C), as compared with all other treatment groups. Primary CD4+ T cells treated with mixture and activation were confirmed to be acting as pTH17 cells via multiplex cytokine array. (DG) All 48 analytes measured in supernatant showed significant differences, including IL-17A, IL-10, MCP-1, and CCL5 (RANTES). The data are represented as means ± SD. A one-way ANOVA with Tukey post hoc test was used to assess significance. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. FSC, forward light scatter.

FIGURE 3.

IL-23 mixture treatment significantly promotes the production of pathogenic TH17 cells from primary human naive CD4+ T cells and alters the production of factors involved in endometriosis lesion establishment and maintenance. Primary naive CD4+ T cells isolated from human PBMCs were treated in vitro in triplicate for 4 d with an IL-23 mixture and CD3/CD28 activation, CD3/CD28 activation alone, IL-23 mixture alone, or media. (A) Cells were restimulated with PMA/ionomycin and protein transport inhibitor mixture prior to supernatant collection and flow cytometric analysis for TH17 cells (CD45+CD4+CCR6+RORγt+IL17A+). (B and C) Primary CD4+ T cells treated with mixture and activation produced a significantly higher frequency of TH17 cells within the CD45+CD4+ cell population (B) and a number of TH17 cells per 100,000 events (C), as compared with all other treatment groups. Primary CD4+ T cells treated with mixture and activation were confirmed to be acting as pTH17 cells via multiplex cytokine array. (DG) All 48 analytes measured in supernatant showed significant differences, including IL-17A, IL-10, MCP-1, and CCL5 (RANTES). The data are represented as means ± SD. A one-way ANOVA with Tukey post hoc test was used to assess significance. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. FSC, forward light scatter.

Close modal

We sought to determine the independent effects of IL-23 on cell lines representative of the endometriotic lesion microenvironment (Fig. 4). Specifically, selected cell lines were representative of endometriotic epithelial (12Z) cells and hESCs, as well as vasculature/endothelial cells (HUVECs) and EECCs. Multiplex cytokine array revealed that IL-23 treatment significantly alters production of platelet-derived growth factor (PDGF)-AA and PDGF-AB/BB by 12Z cells, EECCs, and HUVECs (Fig. 4A–C), as well as IFN-γ–inducible protein 10 (IP-10) and fibroblast growth factor-2 (FGF-2) production by EECCs and HUVECs, respectively (Fig. 4E, 4F). Although hESCs also produced notable PDGF levels in response to IL-23 treatment, there were no significant differences between treatment groups (data not shown). Additionally, rhIL-23 treatment had dose-dependent effects on hESCs, with significant increases in both IL-1β (Fig. 4D) and Fms-related tyrosine kinase 3 ligand (FLT-3L; Fig. 4G).

FIGURE 4.

rhIL-23 stimulation of cells representative of the endometriotic microenvironment significantly alter the production of cytokines/chemokines known to play a role in lesion establishment and maintenance. 12Z cells, EECCs, HUVECs, and hESCs were stimulated in triplicate for 24 h with 0.01% BSA in PBS (vehicle) or varying concentrations of rhIL-23 (1, 10, 50, and 100 ng/ml) prior to supernatant collection and analysis via multiplex cytokine array. A total of 48 analytes were measured with focus on inflammatory and angiogenic factors, as well as mediators in the TH17 pathway. (AC) Most notably, IL-23 treatment significantly altered the production of PDGF by 12Z cells, EECCs, and HUVECs. (DF) In a dose-dependent fashion, IL-23 stimulation significantly increased IL-1β and FLT-3L production by hESCs (D, G) and IP-10 production by EECCs (E). IL-23 treatment also significantly increased FGF-2 levels in HUVECs (F). The data are represented as means ± SD. A one-way ANOVA with Tukey post hoc test was used to assess significance. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 4.

rhIL-23 stimulation of cells representative of the endometriotic microenvironment significantly alter the production of cytokines/chemokines known to play a role in lesion establishment and maintenance. 12Z cells, EECCs, HUVECs, and hESCs were stimulated in triplicate for 24 h with 0.01% BSA in PBS (vehicle) or varying concentrations of rhIL-23 (1, 10, 50, and 100 ng/ml) prior to supernatant collection and analysis via multiplex cytokine array. A total of 48 analytes were measured with focus on inflammatory and angiogenic factors, as well as mediators in the TH17 pathway. (AC) Most notably, IL-23 treatment significantly altered the production of PDGF by 12Z cells, EECCs, and HUVECs. (DF) In a dose-dependent fashion, IL-23 stimulation significantly increased IL-1β and FLT-3L production by hESCs (D, G) and IP-10 production by EECCs (E). IL-23 treatment also significantly increased FGF-2 levels in HUVECs (F). The data are represented as means ± SD. A one-way ANOVA with Tukey post hoc test was used to assess significance. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

To assess IL-23 influence on cell proliferation and apoptosis, we conducted WST-1 and caspase 3/7 glo assays, respectively (Supplemental Fig. 1). rhIL-23 treatment significantly increased hESC proliferation in 1 and 100 ng/ml treatment groups, although there were no significant differences in 12Z cell, EECC, or HUVEC proliferation. There were also no differences in cell death of rhIL-23–treated 12Z cells, EECCs, HUVECs, or hESCs. Moreover, we sought to determine IL-23 influence on angiogenesis using a tubulogenesis assay with HUVECs. However, no significant differences were found in tube length or number of total branching points when comparing rhIL-23–treated groups to the PBS control (Supplemental Fig. 2).

The literature shows that IL-23 signaling is necessary to drive, maintain, and expand pTH17 cell fate (18, 20, 42). Thus, we sought to model this in vitro, examining whether IL-23 alters production of pTH17 cells from naive CD4+ T cells isolated from murine splenocytes using flow cytometry (Fig. 5A). The results depicted treatment with an IL-23 containing TH17 derivation mixture significantly increased frequency and number of pTH17 cells from murine naive CD4+ T cells compared with all other treatment groups (Fig. 5B, 5C).

FIGURE 5.

TH17 derivation mixture significantly promotes the production of pathogenic TH17 cells from murine naive CD4+ T cells. Naive CD4+ T cells isolated from murine splenocytes were treated in vitro in triplicate for 5 d with the mixture and CD3/CD28 activation, mixture and 10 ng/ml rmIL-23 with activation; 10 ng/ml rmIL-23 and activation; or media with activation. (A) Cells were restimulated with PMA/ionomycin and protein transport inhibitor mixture prior to flow cytometric analysis for pTH17 cells (CD45+CD3+CD4+CCR6+RORγt+IL-17A+IFN-γIL-10). (B and C) Naive CD4+ T cells treated with the TH17 derivation mixture and activation produced a significantly higher frequency of pTH17 cells within the CD45+CD3+CD4+ population (B), as well as the number of TH17 cells per 71,388 events (C), as compared with all other treatment groups. The number of TH17 cells was normalized to the lowest acquired events among all samples (71,388 events) using FlowJo software. The data are represented as means ± SD. A one-way ANOVA with Tukey post hoc test was used to assess significance. ***p < 0.001, ****p < 0.0001.

FIGURE 5.

TH17 derivation mixture significantly promotes the production of pathogenic TH17 cells from murine naive CD4+ T cells. Naive CD4+ T cells isolated from murine splenocytes were treated in vitro in triplicate for 5 d with the mixture and CD3/CD28 activation, mixture and 10 ng/ml rmIL-23 with activation; 10 ng/ml rmIL-23 and activation; or media with activation. (A) Cells were restimulated with PMA/ionomycin and protein transport inhibitor mixture prior to flow cytometric analysis for pTH17 cells (CD45+CD3+CD4+CCR6+RORγt+IL-17A+IFN-γIL-10). (B and C) Naive CD4+ T cells treated with the TH17 derivation mixture and activation produced a significantly higher frequency of pTH17 cells within the CD45+CD3+CD4+ population (B), as well as the number of TH17 cells per 71,388 events (C), as compared with all other treatment groups. The number of TH17 cells was normalized to the lowest acquired events among all samples (71,388 events) using FlowJo software. The data are represented as means ± SD. A one-way ANOVA with Tukey post hoc test was used to assess significance. ***p < 0.001, ****p < 0.0001.

Close modal

To assess IL-23 influence on immune cell recruitment/immune dysfunction in endometriosis, C57BL/6 mice (n = 12) underwent endometriosis induction surgery, and mice were treated with rmIL-23 (n = 6) or PBS (controls, n = 6) via i.p. injections three times a week for 3 weeks. Murine splenocytes and PF were collected to assess systemic and local immune alterations, respectively, via flow cytometry (Fig. 6). The results depicted significant alterations to CD4+ T cells, myeloid cells, TREGs, and pTH17 cells in PF, although there were no significant differences in immune cell subsets within murine splenocytes. As speculated, IL-23 significantly altered the TH17/TREG axis in murine PF, increasing TREGs and decreasing pTH17 cells (Fig. 6J, 6K), although these differences were not statistically significant in murine splenocytes (Fig. 6D, 6E). Significant differences seen in immune cell subsets within PF were not detected in our sham mouse model (Supplemental Fig. 3), highlighting the influence of IL-23 specifically in the context of endometriosis. IL-23 also significantly increased immune cell recruitment to the peritoneal cavity, irrespective of disease context (endometriosis/sham; data not shown).

FIGURE 6.

IL-23 treatment results in significant immune dysfunction localized to the murine endometriotic microenvironment, confirmed via flow cytometry. (A) At 1 wk following surgery, endometriosis (EM)–induced mice were treated (i.p.) with 1 µg of rmIL-23 (n = 6) or PBS (n = 6) three times a week for 3 wk before splenocytes, and PF was isolated for flow cytometric analysis. (B) Markers were used to detect CD4+ T cells (CD45+CD4+), myeloid cells (CD45+CD11b+), TREGs (CD45+CD4+CD25+Foxp3+), pTH17 cells (CD45+CD4+CCR6+ROR-γt+IL-17A+IL-10), npTH17 cells (CD45+CD4+CCR6+RORγt+IL-17A+IL-10+), and total TH17 cells (CD45+CD4+CCR6+ROR-γt+IL-17A+). (C–N) The results depict respective immune cell populations within the spleen (C–H), as well as the PF (I–N). The data are represented as means ± SD. A nonparametric Student t test with Mann–Whitney correction was used to assess significance. *p < 0.05, **p < 0.01. Graphic adapted from “Mouse High Fat Diet Experimental Timeline,” by BioRender.com (2023). Retrieved from https://app.biorender.com/biorender-templates.

FIGURE 6.

IL-23 treatment results in significant immune dysfunction localized to the murine endometriotic microenvironment, confirmed via flow cytometry. (A) At 1 wk following surgery, endometriosis (EM)–induced mice were treated (i.p.) with 1 µg of rmIL-23 (n = 6) or PBS (n = 6) three times a week for 3 wk before splenocytes, and PF was isolated for flow cytometric analysis. (B) Markers were used to detect CD4+ T cells (CD45+CD4+), myeloid cells (CD45+CD11b+), TREGs (CD45+CD4+CD25+Foxp3+), pTH17 cells (CD45+CD4+CCR6+ROR-γt+IL-17A+IL-10), npTH17 cells (CD45+CD4+CCR6+RORγt+IL-17A+IL-10+), and total TH17 cells (CD45+CD4+CCR6+ROR-γt+IL-17A+). (C–N) The results depict respective immune cell populations within the spleen (C–H), as well as the PF (I–N). The data are represented as means ± SD. A nonparametric Student t test with Mann–Whitney correction was used to assess significance. *p < 0.05, **p < 0.01. Graphic adapted from “Mouse High Fat Diet Experimental Timeline,” by BioRender.com (2023). Retrieved from https://app.biorender.com/biorender-templates.

Close modal

In addition to inflammation, lesion proliferation and vascularization are known hallmark features of endometriosis pathophysiology (6, 43, 44). Thus, we aimed to examine the role of IL-23 on lesion vascularization and proliferation using an established endometriosis mouse model (Fig. 7). H&E staining confirmed that murine endometriotic lesions were representative of endometriosis lesion histology (Fig. 7A, 7B). Additionally, lesions from IL-23–treated mice had large, multinucleated cells consistent with giant cell histology (Fig. 7C). To our knowledge, this is the first report identifying giant cells within murine endometriosis-like lesions. Although present in some PBS-treated mice, IL-23–treated mice lesions had significantly more giant cells (Fig. 7D). Lesions were also subjected to IHC for markers of proliferation (Ki67) and vascularization (CD31; Fig. 7E, 7F, 7H, 7I). IL-23–treated mice had trends of increased lesion proliferation and vascularization, although the differences were not statistically significant (Fig. 7G, 7J).

FIGURE 7.

IL-23 treatment results in significantly increased numbers of giant cells within lesions as well as trends of increased lesion proliferation and vascularization. At 1 wk after endometriosis induction surgery, the mice were administered (i.p.) 1 µg rmIL-23 (n = 6) or PBS (n = 6) three times a week for 3 wk. (A and B) The lesions of both PBS (A) and rmIL-23 (B) treated mice well recapitulate endometriosis lesion histology, as shown by H&E staining. (C and D) rmIL-23–treated mice had significantly increased numbers of large, multinucleated cells within their lesions consistent with giant cell histology, as indicated by black arrows. (E, F, H, and I) Murine lesions were subjected to IHC for markers of proliferation (Ki67; E, F) and vascularization (CD31; H, I). (G and J) A cytonuclear algorithm was used to detect percentage of cells expressing Ki67 compared with the total cell population (G), whereas the percentage area quantification was used to analyze vascularization (J). rmIL-23–treated mice had trends of increased proliferation and vascularization, although these differences were not statistically significant. Statistical significance was assessed using a nonparametric Student t test with Mann–Whitney correction. **p < 0.01. Scanned H&E images were digitally analyzed using HALO imaging software (Indica Labs) and provided at 3× and 20× magnification. Scale bars, 500 µm and 50 µm, respectively. Similarly, scanned IHC images are provided at 15× magnification. Scale bar, 100 µm.

FIGURE 7.

IL-23 treatment results in significantly increased numbers of giant cells within lesions as well as trends of increased lesion proliferation and vascularization. At 1 wk after endometriosis induction surgery, the mice were administered (i.p.) 1 µg rmIL-23 (n = 6) or PBS (n = 6) three times a week for 3 wk. (A and B) The lesions of both PBS (A) and rmIL-23 (B) treated mice well recapitulate endometriosis lesion histology, as shown by H&E staining. (C and D) rmIL-23–treated mice had significantly increased numbers of large, multinucleated cells within their lesions consistent with giant cell histology, as indicated by black arrows. (E, F, H, and I) Murine lesions were subjected to IHC for markers of proliferation (Ki67; E, F) and vascularization (CD31; H, I). (G and J) A cytonuclear algorithm was used to detect percentage of cells expressing Ki67 compared with the total cell population (G), whereas the percentage area quantification was used to analyze vascularization (J). rmIL-23–treated mice had trends of increased proliferation and vascularization, although these differences were not statistically significant. Statistical significance was assessed using a nonparametric Student t test with Mann–Whitney correction. **p < 0.01. Scanned H&E images were digitally analyzed using HALO imaging software (Indica Labs) and provided at 3× and 20× magnification. Scale bars, 500 µm and 50 µm, respectively. Similarly, scanned IHC images are provided at 15× magnification. Scale bar, 100 µm.

Close modal

IL-23 is known to promote inflammation and pTH17 cell fate in combination with other inflammatory cytokines (18, 45). Thus, we sought to determine whether treatment with TH17 priming mixture and/or rmIL-23 influenced production of pTH17 cells, and in turn the inflammatory milieu, within an endometriosis mouse model (Fig. 8). Splenocytes and PF were collected and analyzed via flow cytometry; however, no statistically significant differences in immune cell subsets were detected in treatment groups compared with controls.

FIGURE 8.

IL-23 and/or TH17 priming mixture do not significantly alter local or systemic immune response during a short time-course study in a mouse model of endometriosis. Following endometriosis (EM) induction surgery in C57BL/6 mice (n = 16), the mice were rested for 4 d (n = 8) or treated daily (i.p.) with a TH17 priming mixture (0.5 µg of rmIL-6, 1 µg of rmTGF-β1; n = 8). (A) Following this, half of the mice in each cohort were treated daily (i.p.) for a further 10 d with either 1 µg of rmIL-23 (n = 4) or PBS (controls; n = 4) prior to euthanasia. (B) Markers were used to detect CD4+ T cells (CD45+CD4+), myeloid cells (CD45+CD11b+), TREGs (CD45+CD4+CD25+Foxp3+), pTH17 cells (CD45+CD4+CCR6+RORγt+IL17A+IL10), npTH17 cells (CD45+CD4+CCR6+RORγt+IL17A+IL10+), and total TH17 cells (CD45+CD4+CCR6+RORγt+IL17A+). (C–N) Results depict respective immune cell populations within the spleen (C–H), as well as the PF (I–N). The data are represented as means ± SD. No statistically significant differences were found, as assessed by a one-way ANOVA with Tukey post hoc test. Graphic adapted from “Mouse High Fat Diet Experimental Timeline,” by BioRender.com (2023). Retrieved from https://app.biorender.com/biorender-templates.

FIGURE 8.

IL-23 and/or TH17 priming mixture do not significantly alter local or systemic immune response during a short time-course study in a mouse model of endometriosis. Following endometriosis (EM) induction surgery in C57BL/6 mice (n = 16), the mice were rested for 4 d (n = 8) or treated daily (i.p.) with a TH17 priming mixture (0.5 µg of rmIL-6, 1 µg of rmTGF-β1; n = 8). (A) Following this, half of the mice in each cohort were treated daily (i.p.) for a further 10 d with either 1 µg of rmIL-23 (n = 4) or PBS (controls; n = 4) prior to euthanasia. (B) Markers were used to detect CD4+ T cells (CD45+CD4+), myeloid cells (CD45+CD11b+), TREGs (CD45+CD4+CD25+Foxp3+), pTH17 cells (CD45+CD4+CCR6+RORγt+IL17A+IL10), npTH17 cells (CD45+CD4+CCR6+RORγt+IL17A+IL10+), and total TH17 cells (CD45+CD4+CCR6+RORγt+IL17A+). (C–N) Results depict respective immune cell populations within the spleen (C–H), as well as the PF (I–N). The data are represented as means ± SD. No statistically significant differences were found, as assessed by a one-way ANOVA with Tukey post hoc test. Graphic adapted from “Mouse High Fat Diet Experimental Timeline,” by BioRender.com (2023). Retrieved from https://app.biorender.com/biorender-templates.

Close modal

IL-17, a proinflammatory and proangiogenic cytokine largely secreted by TH17 cells, has been linked in endometriosis pathogenesis (10). Although the exact mechanisms are unknown, IL-17 and other downstream TH17 cell cytokines may exacerbate endometriosis development by recruiting immune cells to the lesion site, such as neutrophils, macrophages, and lymphocytes, collectively enhancing lesion establishment (12, 28). Specifically, IL-17 is increased in patient plasma and PF, and this is correlated with disease severity (46). Additionally, IL-17 is produced by lesions, and postexcision surgery systemic IL-17 levels are significantly reduced (10), highlighting the potential link between IL-17 and endometriosis. TH17 cells are also dysregulated in endometriosis (24, 29, 33) and associated with disease severity (13). Thus, whereas downstream IL-17 has been examined in endometriosis, a knowledge gap exists regarding the factor(s) driving TH17/IL-17 axis dysregulation in endometriosis. IL-23 is known to govern TH17 cell fate (20, 47), driving pTH17 cells producing IL-17 and downregulating anti-inflammatory IL-10 (17, 18). Hence, as emerging evidence has underscored the role of IL-23 in driving this axis to produce IL-17 in various other diseases (30, 48), we sought to examine whether IL-23 may be driving TH17/IL-17 axis dysregulation in endometriosis to exacerbate disease.

Our studies using patient samples reveal significant dysregulation in the IL-23/TH17 axis in patient tissues. To our knowledge, this is the first report quantifying RNA and protein expression of IL-23 (p19 [IL23A] and p40 [IL12B] subunits) and key mediators in the IL-23/TH17 axis in endometriosis patient ectopic (lesion) and matched eutopic endometrial tissues compared with healthy controls. RT-qPCR results show that IL17A, IL17F, IL21, IL22, IL12B (IL-23 p40 subunit), and TGFB1 are significantly upregulated in ectopic tissues compared with matched eutopic tissue and control samples. This is substantial because IL-17A, IL-17F, IL-21, and IL-22 are secreted by TH17 cells, whereas IL-23 and TGF-β1 are important in driving and maintaining pTH17 cell fate (20, 47). RT-qPCR results also that suggest patients may have increased sensitivity to IL-23 signaling, because IL23R was significantly upregulated in both ectopic and eutopic patient tissues compared with controls. Moreover, IL12B, encoding the p40 subunit shared between IL-23 and IL-12, was significantly increased in accordance with literature (49). This increase is unlikely to indicate IL-12 involvement because the p35 subunit (IL12A), specific to IL-12, was not significantly increased. Instead, this may indicate involvement of p40 homodimers or p40 monomers, which are increased in endometriosis PF (50).

Complimentary to RT-qPCR results, IL-23 protein was detected in endometriotic lesions and was significantly increased in patient plasma compared with controls. Although IL-23 protein expression was not significantly different across patient ectopic (ovarian endometrioma) or eutopic tissues compared with controls, IL-23 was significantly increased in luminal and glandular epithelium compared with stroma. Because endometrioma lesions are more cystic in nature, they are comprised of less luminal and glandular epithelium compared with noncystic lesions. Thus, because we illustrate that IL-23 is predominately localized to epithelial tissue, IL-23 protein is likely reduced in endometrioma lesions compared with other lesion subtypes. However, further research is required to quantify differential IL-23 protein expression across lesion subtypes. Others do report significantly increased IL-23 protein in endometriosis patient PF, ectopic lesions, and serum, which is correlated with increased IL-17 levels (51–54). Moreover, IL-23 is significantly increased in both patient follicular fluid and serum in later stages of disease (III and IV) compared with earlier stages (I and II) (52), highlighting that IL-23 may be associated with disease severity. However, some report no significant differences in IL-23 in patient serum when not stratified by disease stage (52, 53). Although we did not capture significant differences in IL23A (p19 subunit) in patient tissues at the mRNA level, dysregulation of other key members within IL-23/TH17 axis combined with significantly increased IL-23 protein levels in patient plasma indicate involvement in endometriosis pathophysiology.

In our in vitro studies, IL-23 mixture treatment in combination with CD3/CD28 activation significantly promoted TH17 cell differentiation from human and murine naive CD4+ T cells. This mixture was selected because evidence shows the endometriosis milieu contains and is influenced by TGF-β1, IL-1β, IL-6, IL-21, and IL-23 cytokines (52, 55–58), which are also vital in driving pTH17 cell fate (17, 59). Thus, we aimed to partially recapitulate this environment in vitro. The results depict that naive human CD4+ T cells driven in the presence of this combination treatment produce significantly increased IL-17A/F and decreased IL-10, characteristic of pTH17 cells, confirming that TH17 cells exposed to IL-23 mixture acquire a pathogenic phenotype consistent with literature (12, 20, 47). Additionally, human naive CD4+ T cells driven with this combination treatment produce significantly increased MCP-1, CCL5, CXCL9, and MDC. Because these factors play important chemotactic roles, IL-23–driven and activated TH17 cells may promote immune cell recruitment and produce proinflammatory factors, such as IL-17, to further exacerbate lesion inflammation, proliferation, and angiogenesis. However, although there are reports of pTH17 cells producing CCL5 (12), it is unclear to what abundance TH17 cells may produce MCP-1, CXCL9, and MDC. Further studies are required to examine coproduction of these factors by TH17 cells.

Broadly, IL-23 is produced in response to damage-associated molecular patterns (DAMPs) (60). Because the literature suggests that DAMPs/alarmins are released during endometriosis lesion establishment (56), IL-23 may not only act in endometriosis to promote and support pTH17 cell fate but also act directly on the lesion to perpetuate inflammatory signals in response to DAMPs. Thus, we sought to determine the direct effects of IL-23 treatment on cells representative of the endometriotic microenvironment, specifically 12Z cells, EECCs, HUVECs, and hESCs. The results depicted that in vitro rhIL-23 stimulation of cell lines significantly altered production of PDGF-AA from 12Z cells, PDGF-AB/BB levels from both EECCs and HUVECs, and FGF-2 production from HUVECs. Additionally, in vitro rhIL-23 treatment produced significant dose-dependent increases of IP-10 by EECCs and IL-1β and FLT-3L by hESCs. rhIL-23 treatment also significantly increased proliferation of hESCs, a major component of the endometriotic lesion. Collectively, our in vitro data suggest that IL-23 stimulation significantly alters production of factors known to play a role in lesion establishment and maintenance and may promote lesion growth/survival.

Our mouse model of endometriosis has been extensively used to gain mechanistic insights into the complex pathophysiology of endometriosis (34, 61–63). Indeed, we identify significantly increased numbers of large, multinucleated cells in lesions obtained from IL-23–treated mice compared with controls. Histologically, these cells appear to be giant cells of monocyte origin; however, further characterization is necessary. Although we captured trends of increased lesion proliferation and vascularization in IL-23–treated mice, these differences were not statistically significant. This may be reflective of our mouse model, time course, and dose/route of IL-23 administration, because IL-23 has known effects on cell proliferation and vascularization (60, 64–66). We also used this mouse model to determine whether IL-23 treatment alters systemic and/or local immune cell subsets and in turn may promote immune dysfunction associated with endometriosis. Indeed, IL-23 treatment significantly reduced CD4+ T cells and myeloid cells in murine PF compared with controls, possibly due to homing of CD4+ T cells and myeloid cells to the lesion. Additionally, IL-23 treatment dysregulated the local TH17/TREG axis, where pTH17 cells were significantly reduced and TREGs were significantly increased in PF of IL-23–treated mice. This TH17/TREG axis dysregulation is noteworthy because balance between TH17 and TREGs is necessary to maintain immune homeostasis and avoid dysfunction (23, 67–69). These significant alterations in immune cell subsets within murine PF were not observed in our sham mouse model, highlighting that these differences were specific to an endometriosis-like microenvironment. Finally, because the literature suggests that TGF-β1 and IL-6 are required for initial derivation of TH17 cells, whereas IL-23 is necessary for full induction and maintenance of pTH17 cell fate (70, 71), we used a mouse model to assess the influence of IL-23 alone and in combination with a TH17 priming mixture (TGF-β1 and IL-6) during lesion establishment. Our short time-course study revealed no significant differences in murine immune cell subsets between groups. Thus, further research is required to establish the specific combination of cytokines and timeframe necessary to capture murine TH17 differentiation in vivo. Our in vivo data reveal that IL-23 treatment significantly influences local immune dysfunction in endometriosis and promotes immune cell recruitment to the peritoneal cavity, which may exacerbate local immune dysfunction to promote lesion survival and the well known inflammatory milieu in endometriosis.

We acknowledge the limitations of this study and others investigating endometriosis due to limited access acquiring well categorized patient samples stratified by disease stage, subtype, and menstrual cycle. Invasive diagnostic measures, heterogenous symptom presentation, and stigmatization of reproductive disorders also complicate this, commonly resulting in long diagnostic delays and misdiagnosis of endometriosis patients. Due to limitations in obtaining fresh patient samples, we were unable to isolate primary cells from endometriosis lesions, and thus we acknowledge known limitations of immortalized cell lines. However, we supplement these data with data from primary human CD4+ T cells. Moreover, although EECCs are widely used in endometriosis literature to examine the molecular mechanisms of endometriosis pathophysiology (72–74), we acknowledge the limitations of this cell line. Specifically, because EECCs originate from the endometrium of a carcinoma patient, EECCs may not be exactly representative of “normal” eutopic endometrium (75). Finally, while we use our syngeneic immunocompetent endometriosis mouse model to provide insights into the role of IL-23 in endometriosis pathophysiology, this model is not able to completely recapitulate the chronic nature and heterogenous presentation of human disease, because rodents do not spontaneously develop endometriosis.

In conclusion, while our data suggest IL-23/TH17 axis dysregulation in endometriosis, it is important to note that no existing evidence in literature has mechanistically linked this axis to endometriosis pathology. In this article, we report significant IL-23/TH17 axis dysregulation in endometriosis patients, and this was captured in in vitro studies and our in vivo mouse model. Because there is currently no effective long-term, noninvasive, or nonhormonal therapy option for endometriosis patients who aim to conceive, there is an urgent need to explore novel therapeutics. Future directions should investigate the correlation between IL-23 and IL-17 levels in circulation and locally in patient PF, because others report a significant positive correlation of IL-23 and IL-17 levels in endometriosis patient plasma and PF (51). Similarly, examining the correlation between IL-23 and IL-10 levels in circulation and PF may provide insights into local and systemic immunomodulatory effects of IL-23. Ultimately, IL-23 may provide a promising therapeutic avenue to reduce the large burden of endometriosis, because IL-23–targeting therapeutics have been approved for human use and/or are currently in development for various chronic inflammatory diseases, including rheumatoid arthritis and psoriasis (32, 76–80).

The authors have no financial conflicts of interest.

We thank Oliver Jones at Queen’s CardioPulmonary Unit (Queen’s University) for all histology processing. We also thank Wei Wang at Kingston Health Sciences Centre (Queen’s University) and Yuexin Yang in the Department of Pathology and Molecular Medicine (Queen’s University) for immunohistochemistry processing. Finally, we thank Xiao Zhang at Queen’s Laboratory for Molecular Pathology (Queen’s University) for aid with slide scanning.

This work was supported by Canadian Institutes of Health Research Grant 394570 (to D.J.S., K.B.Z., J.E.M., H.L., S.H.A., A.M., and C.T.). D.J.S. is a recipient of a Canadian Institutes of Health Research Canadian Graduate Scholarships Doctoral Research Award.

The online version of this article contains supplemental material.

D.J.S. conceived and conducted experiments, analyzed data, and wrote the manuscript; K.B.Z., J.E.M., H.L., S.H.A., and A.M. conducted experiments; O.B. and B.A.L. contributed human patient samples; C.T. contributed reagents, conceived experiments, and provided financial support; all authors read, edited, and approved the manuscript.

DAMP

damage-associated molecular pattern

EECC

endometrial epithelial carcinoma cell

hESC

human endometrial stromal cell

IHC

immunohistochemistry

MDC

macrophage-derived chemokine

PDGF

platelet-derived growth factor

PF

peritoneal fluid

qPCR

quantitative PCR

RA

receptor A

rh

recombinant human

rm

recombinant mouse

TREG

T-regulatory cell

1
Giudice
,
L. C.
2010
.
Endometriosis
.
N. Engl. J. Med.
362
:
2389
2398
.
2
Alimi
,
Y.
,
J.
Iwanaga
,
M.
Loukas
,
R. S.
Tubbs
.
2018
.
The clinical anatomy of endometriosis: a review
.
Cureus
10
:
e3361
.
3
Allaire
,
C.
,
M. A.
Bedaiwy
,
P. J.
Yong
.
2023
.
Diagnosis and management of endometriosis
.
CMAJ
195
:
E363
E371
.
4
Shafrir
,
A. L.
,
L. V.
Farland
,
D. K.
Shah
,
H. R.
Harris
,
M.
Kvaskoff
,
K.
Zondervan
,
S. A.
Missmer
.
2018
.
Risk for and consequences of endometriosis: a critical epidemiologic review
.
Best Pract. Res. Clin. Obstet. Gynaecol.
51
:
1
15
.
5
Horne
,
A. W.
,
S. A.
Missmer
.
2022
.
Pathophysiology, diagnosis, and management of endometriosis
.
BMJ
379
:
e070750
.
6
Zondervan
,
K. T.
,
C. M.
Becker
,
K.
Koga
,
S. A.
Missmer
,
R. N.
Taylor
,
P.
Viganò
.
2018
.
Endometriosis
.
Nat. Rev. Dis. Primers
4
:
9
.
7
Osuga
,
Y.
,
Y.
Hirota
,
T.
Hirata
,
M.
Takamura
,
Y.
Urata
,
M.
Harada
,
G.
Izumi
,
T.
Fujii
,
K.
Koga
.
2016
.
Th2 cells and Th17 cells in the development of endometriosis—possible roles of interleukin-4 and interleukin-17A
.
J. Endometr.
8
:
136
140
.
8
Miossec
,
P.
,
J. K.
Kolls
.
2012
.
Targeting IL-17 and TH17 cells in chronic inflammation
.
Nat. Rev. Drug. Discov.
11
:
763
776
.
9
Bartlett
,
H. S.
,
R. P.
Million
.
2015
.
Targeting the IL-17–TH17 pathway
.
Nat. Rev. Drug. Discov.
14
:
11
12
.
10
Ahn
,
S. H.
,
A. K.
Edwards
,
S. S.
Singh
,
S. L.
Young
,
B. A.
Lessey
,
C.
Tayade
.
2015
.
IL-17A contributes to the pathogenesis of endometriosis by triggering proinflammatory cytokines and angiogenic growth factors
.
J. Immunol.
195
:
2591
2600
.
11
Patel
,
D. D.
,
V. K.
Kuchroo
.
2015
.
Th17 cell pathway in human immunity: lessons from genetics and therapeutic interventions
.
Immunity
43
:
1040
1051
.
12
Wu
,
X.
,
J.
Tian
,
S.
Wang
.
2018
.
Insight into non-pathogenic Th17 cells in autoimmune diseases
.
Front. Immunol.
9
:
1112
.
13
Gogacz
,
M.
,
I.
Winkler
,
A.
Bojarska-Junak
,
J.
Tabarkiewicz
,
A.
Semczuk
,
T.
Rechberger
,
A.
Adamiak
.
2016
.
Increased percentage of Th17 cells in peritoneal fluid is associated with severity of endometriosis
.
J. Reprod. Immunol.
117
:
39
44
.
14
Stockinger
,
B.
,
S.
Omenetti
.
2017
.
The dichotomous nature of T helper 17 cells
.
Nat. Rev. Immunol.
17
:
535
544
.
15
Hirota
,
K.
,
J.-E.
Turner
,
M.
Villa
,
J. H.
Duarte
,
J.
Demengeot
,
O. M.
Steinmetz
,
B.
Stockinger
.
2013
.
Plasticity of TH17 cells in Peyer’s patches is responsible for the induction of T cell–dependent IgA responses
.
Nat. Immunol.
14
:
372
379
.
16
Conti
,
H. R.
,
A. C.
Peterson
,
L.
Brane
,
A. R.
Huppler
,
N.
Hernández-Santos
,
N.
Whibley
,
A. V.
Garg
,
M. R.
Simpson-Abelson
,
G. A.
Gibson
,
A. J.
Mamo
, et al
2014
.
Oral-resident natural Th17 cells and γδ T cells control opportunistic Candida albicans infections
.
J. Exp. Med.
211
:
2075
2084
.
17
McGeachy
,
M. J.
,
K. S.
Bak-Jensen
,
Y.
Chen
,
C. M.
Tato
,
W.
Blumenschein
,
T.
McClanahan
,
D. J.
Cua
.
2007
.
TGF-β and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain TH-17 cell–mediated pathology
.
Nat. Immunol.
8
:
1390
1397
.
18
Lee
,
Y.
,
A.
Awasthi
,
N.
Yosef
,
F. J.
Quintana
,
S.
Xiao
,
A.
Peters
,
C.
Wu
,
M.
Kleinewietfeld
,
S.
Kunder
,
D. A.
Hafler
, et al
2012
.
Induction and molecular signature of pathogenic TH17 cells
.
Nat. Immunol.
13
:
991
999
.
19
Teng
,
M. W. L.
,
E. P.
Bowman
,
J. J.
McElwee
,
M. J.
Smyth
,
J.-L.
Casanova
,
A. M.
Cooper
,
D. J.
Cua
.
2015
.
IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases
.
Nat. Med.
21
:
719
729
.
20
Langrish
,
C. L.
,
Y.
Chen
,
W. M.
Blumenschein
,
J.
Mattson
,
B.
Basham
,
J. D.
Sedgwick
,
T.
McClanahan
,
R. A.
Kastelein
,
D. J.
Cua
.
2005
.
IL-23 drives a pathogenic T cell population that induces autoimmune inflammation
.
J. Exp. Med.
201
:
233
240
.
21
Ma
,
R.
,
H.
Su
,
K.
Jiao
,
J.
Liu
.
2023
.
Role of Th17 cells, Treg cells, and Th17/Treg imbalance in immune homeostasis disorders in patients with chronic obstructive pulmonary disease
.
Immun. Inflamm. Dis.
11
:
e784
.
22
Wang
,
R.
,
S. Z.
Hasnain
,
H.
Tong
,
I.
Das
,
A.
C.-H. Chen
,
I.
Oancea
,
M.
Proctor
,
T. H.
Florin
,
R. D.
Eri
,
M. A.
McGuckin
.
2015
.
Neutralizing IL-23 is superior to blocking IL-17 in suppressing intestinal inflammation in a spontaneous murine colitis model
.
Inflamm. Bowel. Dis.
21
:
973
984
.
23
Lee
,
G.
2018
.
The balance of Th17 versus Treg cells in autoimmunity
.
Int. J. Mol. Sci.
19
:
730
.
24
Khan
,
K. N.
,
K.
Yamamoto
,
A.
Fujishita
,
H.
Muto
,
A.
Koshiba
,
H.
Kuroboshi
,
S.
Saito
,
S.
Teramukai
,
M.
Nakashima
,
J.
Kitawaki
.
2019
.
Differential levels of regulatory T cells and T-helper-17 cells in women with early and advanced endometriosis
.
J. Clin. Endocrinol. Metab.
104
:
4715
4729
.
25
Maloy
,
K. J.
,
M. C.
Kullberg
.
2008
.
IL-23 and Th17 cytokines in intestinal homeostasis
.
Mucosal Immunol.
1
:
339
349
.
26
Vallvé-Juanico
,
J.
,
S.
Houshdaran
,
L. C.
Giudice
.
2019
.
The endometrial immune environment of women with endometriosis
.
Hum. Reprod. Update
25
:
564
591
.
27
Hirata
,
T.
,
Y.
Osuga
,
K.
Hamasaki
,
O.
Yoshino
,
M.
Ito
,
A.
Hasegawa
,
Y.
Takemura
,
Y.
Hirota
,
E.
Nose
,
C.
Morimoto
, et al
2008
.
Interleukin (IL)-17A stimulates IL-8 secretion, cyclooxygensase-2 expression, and cell proliferation of endometriotic stromal cells
.
Endocrinology
149
:
1260
1267
.
28
Miller
,
J. E.
,
S. H.
Ahn
,
R. M.
Marks
,
S. P.
Monsanto
,
A. T.
Fazleabas
,
M.
Koti
,
C.
Tayade
.
2020
.
IL-17A modulates peritoneal macrophage recruitment and M2 polarization in endometriosis
.
Front. Immunol.
11
:
108
.
29
Miller
,
J. E.
,
H.
Lingegowda
,
D. J.
Sisnett
,
C. N.
Metz
,
P. K.
Gregersen
,
M.
Koti
,
C.
Tayade
.
2022
.
T helper 17 axis and endometrial macrophage disruption in menstrual effluent provides potential insights into the pathogenesis of endometriosis
.
F S Sci.
3
:
279
287
.
30
Schinocca
,
C.
,
C.
Rizzo
,
S.
Fasano
,
G.
Grasso
,
L.
La Barbera
,
F.
Ciccia
,
G.
Guggino
.
2021
.
Role of the IL-23/IL-17 pathway in rheumatic diseases: an overview
.
Front. Immunol.
12
:
637829
.
31
Iwakura
,
Y.
,
H.
Ishigame
.
2006
.
The IL-23/IL-17 axis in inflammation
.
J. Clin. Invest.
116
:
1218
1222
.
32
Torres
,
T.
2018
.
Selective IL-23 inhibitors: the new kids on the block in the treatment of psoriasis
.
Actas Dermosifiliogr. (Engl. Ed.)
109
:
674
676
.
33
Takamura
,
M.
,
K.
Koga
,
G.
Izumi
,
T.
Hirata
,
M.
Harada
,
Y.
Hirota
,
O.
Hiraike
,
T.
Fujii
,
Y.
Osuga
.
2015
.
Simultaneous detection and evaluation of four subsets of CD4+ T lymphocyte in lesions and peripheral blood in endometriosis
.
Am. J. Reprod. Immunol.
74
:
480
486
.
34
Zutautas
,
K. B.
,
D. J.
Sisnett
,
J. E.
Miller
,
H.
Lingegowda
,
T.
Childs
,
O.
Bougie
,
B. A.
Lessey
,
C.
Tayade
.
2023
.
The dysregulation of leukemia inhibitory factor and its implications for endometriosis pathophysiology
.
Front. Immunol.
14
:
1089098
.
35
Symons
,
L. K.
,
J. E.
Miller
,
K.
Tyryshkin
,
S. P.
Monsanto
,
R. M.
Marks
,
H.
Lingegowda
,
K.
Vanderbeck
,
T.
Childs
,
S. L.
Young
,
B. A.
Lessey
, et al
2020
.
Neutrophil recruitment and function in endometriosis patients and a syngeneic murine model
.
FASEB J.
34
:
1558
1575
.
36
Kleinewietfeld
,
M.
,
A.
Manzel
,
J.
Titze
,
H.
Kvakan
,
N.
Yosef
,
R. A.
Linker
,
D. N.
Muller
,
D. A.
Hafler
.
2013
.
Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells
.
Nature
496
:
518
522
.
37
Delens
,
L.
,
G.
Ehx
,
J.
Somja
,
L.
Vrancken
,
L.
Belle
,
L.
Seidel
,
C.
Grégoire
,
G.
Fransolet
,
C.
Ritacco
,
M.
Hannon
, et al
2019
.
In vitro Th17-polarized human CD4+ T cells exacerbate xenogeneic graft-versus-host disease
.
Biol. Blood Marrow Transplant.
25
:
204
215
.
38
Esplugues
,
E.
,
S.
Huber
,
N.
Gagliani
,
A. E.
Hauser
,
T.
Town
,
Y. Y.
Wan
,
W.
O’Connor
,
A.
Rongvaux
,
N.
Van Rooijen
,
A. M.
Haberman
, et al
2011
.
Control of TH17 cells occurs in the small intestine
.
Nature
475
:
514
518
.
39
Bettelli
,
E.
,
Y.
Carrier
,
W.
Gao
,
T.
Korn
,
T. B.
Strom
,
M.
Oukka
,
H. L.
Weiner
,
V. K.
Kuchroo
.
2006
.
Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells
.
Nature
441
:
235
238
.
40
Wimasis
.
2016
. Available at: https://www.wimasis.com. Accessed: December 1, 2023.
41
Cotechini
,
T.
,
O.
Jones
,
C. C. T.
Hindmarch
.
2023
.
Imaging mass cytometry in immuno-oncology
.
Methods Mol. Biol.
2614
:
1
15
.
42
Meyer Zu Horste
,
G.
,
C.
Wu
,
C.
Wang
,
L.
Cong
,
M.
Pawlak
,
Y.
Lee
,
W.
Elyaman
,
S.
Xiao
,
A.
Regev
,
V. K.
Kuchroo
.
2016
.
RBPJ controls development of pathogenic Th17 cells by regulating IL-23 receptor expression
.
Cell. Rep.
16
:
392
404
.
43
Laschke
,
M. W.
,
M. D.
Menger
.
2018
.
Basic mechanisms of vascularization in endometriosis and their clinical implications
.
Hum. Reprod. Update
24
:
207
224
.
44
Tayade
,
C.
2022
.
S11.2: role of endocannabinoids in endometriosis
.
Am. J. Reprod. Immunol.
87
:
55
55
.
45
Manel
,
N.
,
D.
Unutmaz
,
D. R.
Littman
.
2008
.
The differentiation of human TH-17 cells requires transforming growth factor-β and induction of the nuclear receptor RORγt
.
Nat. Immunol.
9
:
641
649
.
46
Zhang
,
X.
,
H.
Xu
,
J.
Lin
,
Y.
Qian
,
L.
Deng
.
2005
.
Peritoneal fluid concentrations of interleukin-17 correlate with the severity of endometriosis and infertility of this disorder
.
BJOG
112
:
1153
1155
.
47
Kleiner
,
J. C.
,
C. F.
Krebs
.
2022
.
Persistence, pathogenicity and plasticity: the role of IL-23 in Th17 fate
.
J. Cell. Immunol.
4
:
121
130
.
48
Morrison
,
P. J.
,
S. J.
Ballantyne
,
M. C.
Kullberg
.
2011
.
Interleukin-23 and T helper 17-type responses in intestinal inflammation: from cytokines to T-cell plasticity
.
Immunology
133
:
397
408
.
49
Zhao
,
W.
,
S.
Kang
,
J.
Zhao
,
L.
Wang
,
S.
Cao
,
Y.
Li
.
2019
.
Aberrant methylation of the IL‐12B promotor region contributes to the risk of developing ovarian endometriosis
.
Mol. Reprod. Dev.
86
:
632
638
.
50
Mazzeo
,
D.
,
P.
Viganó
,
A. M.
Di Blasio
,
F.
Sinigaglia
,
M.
Vignali
,
P.
Panina-Bordignon
.
1998
.
Interleukin-12 and its free p40 subunit regulate immune recognition of endometrial cells: potential role in endometriosis
.
J. Clin. Endocrinol. Metab.
83
:
911
916
.
51
Tarokh
,
M.
,
M. G.
Novin
,
T.
Poordast
,
Z.
Tavana
,
H.
Nazarian
,
M.
Norouzian
,
B.
Gharesi-Fard
.
2019
.
Serum and peritoneal fluid cytokine profiles in infertile women with endometriosis
.
Iran. J. Immunol.
16
:
151
162
.
52
Zhang
,
Q.-F.
,
G.-Y.
Chen
,
Y.
Liu
,
H.-J.
Huang
,
Y.-F.
Song
.
2017
.
Relationship between resistin and IL-23 levels in follicular fluid in infertile patients with endometriosis undergoing IVF-ET
.
Adv. Clin. Exp. Med.
26
:
1431
1435
.
53
Andreoli
,
C. G.
,
V. K.
Genro
,
C. A.
Souza
,
T.
Michelon
,
J. P.
Bilibio
,
C.
Scheffel
,
J. S.
Cunha-Filho
.
2011
.
T helper (Th)1, Th2, and Th17 interleukin pathways in infertile patients with minimal/mild endometriosis
.
Fertil. Steril.
95
:
2477
2480
.
54
Sugahara
,
T.
,
Y.
Tanaka
,
M.
Hamaguchi
,
M.
Fujii
,
K.
Shimura
,
K.
Ogawa
,
T.
Mori
,
I.
Kusuki
,
M.
Fukui
,
J.
Kitawaki
.
2022
.
Reduced innate lymphoid cells in the endometrium of women with endometriosis
.
Am. J. Reprod. Immunol.
87
:
e13502
.
55
Montagna
,
P.
,
S.
Capellino
,
B.
Villaggio
,
V.
Remorgida
,
N.
Ragni
,
M.
Cutolo
,
S.
Ferrero
.
2008
.
Peritoneal fluid macrophages in endometriosis: correlation between the expression of estrogen receptors and inflammation
.
Fertil. Steril.
90
:
156
164
.
56
Symons
,
L. K.
,
J. E.
Miller
,
V. R.
Kay
,
R. M.
Marks
,
K.
Liblik
,
M.
Koti
,
C.
Tayade
.
2018
.
The Immunopathophysiology of Endometriosis
.
Trends. Mol. Med.
24
:
748
762
.
57
Harada
,
T.
,
H.
Yoshioka
,
S.
Yoshida
,
T.
Iwabe
,
Y.
Onohara
,
M.
Tanikawa
,
N.
Terakawa
.
1997
.
Increased interleukin-6 levels in peritoneal fluid of infertile patients with active endometriosis
.
Am. J. Obstet. Gynecol.
176
:
593
597
.
58
Naseri
,
S.
,
Y.
Rosenberg‐Hasson
,
H. T.
Maecker
,
M. I.
Avrutsky
,
P. D.
Blumenthal
.
2023
.
A cross‐sectional study comparing the inflammatory profile of menstrual effluent vs. peripheral blood
.
Health Sci. Rep.
6
:
e1038
.
59
Yang
,
L.
,
D. E.
Anderson
,
C.
Baecher-Allan
,
W. D.
Hastings
,
E.
Bettelli
,
M.
Oukka
,
V. K.
Kuchroo
,
D. A.
Hafler
.
2008
.
IL-21 and TGF-β are required for differentiation of human TH17 cells
.
Nature
454
:
350
352
.
60
Yan
,
J.
,
M. J.
Smyth
,
M. W. L.
Teng
.
2018
.
Interleukin (IL)-12 and IL-23 and their conflicting roles in cancer
.
Cold. Spring Harb. Perspect. Biol.
10
:
a028530
.
61
McCallion
,
A.
,
Y.
Nasirzadeh
,
H.
Lingegowda
,
J. E.
Miller
,
K.
Khalaj
,
S.
Ahn
,
S. P.
Monsanto
,
M.
Bidarimath
,
D. J.
Sisnett
,
A. W.
Craig
, et al
2022
.
Estrogen mediates inflammatory role of mast cells in endometriosis pathophysiology
.
Front. Immunol.
13
:
961599
.
62
Lingegowda
,
H.
,
J. E.
Miller
,
A.
McCallion
,
T.
Childs
,
B. A.
Lessey
,
M.
Koti
,
C.
Tayade
.
2021
.
Implications of dysregulated endogenous cannabinoid family members in the pathophysiology of endometriosis
.
F S Sci.
2
:
419
430
.
63
Lingegowda
,
H.
,
R. M.
Marks
,
L. K.
Symons
,
T.
Alward
,
A. E.
Lomax
,
M.
Koti
,
C.
Tayade
.
2021
.
Synthetic cannabinoid agonist WIN 55212-2 targets proliferation, angiogenesis, and apoptosis via MAPK/AKT signaling in human endometriotic cell lines and a murine model of endometriosis
.
Front. Reprod. Health
3
:
726936
.
64
Wang
,
P.
,
Y.
Zhang
,
J.
Zhang
,
J.
Jiang
,
F.
Jin
,
Z.
Sun
.
2018
.
IL-23 concentration-dependently regulates T24 cell proliferation, migration and invasion and is associated with prognosis in patients with bladder cancer
.
Oncol. Rep.
40
:
3685
3693
.
65
Suzuki
,
H.
,
H.
Ogawa
,
K.
Miura
,
S.
Haneda
,
K.
Watanabe
,
S.
Ohnuma
,
H.
Sasaki
,
T.
Sase
,
S.
Kimura
,
T.
Kajiwara
, et al
2012
.
IL-23 directly enhances the proliferative and invasive activities of colorectal carcinoma
.
Oncol. Lett.
4
:
199
204
.
66
Subhadarshani
,
S.
,
N.
Yusuf
,
C. A.
Elmets
.
2021
.
IL-23 and the tumor microenvironment
.
Adv. Exp. Med. Biol.
1290
:
89
98
.
67
Zhang
,
W.
,
X.
Liu
,
Y.
Zhu
,
X.
Liu
,
Y.
Gu
,
X.
Dai
,
B.
Li
.
2021
.
Transcriptional and posttranslational regulation of Th17/Treg balance in health and disease
.
Eur. J. Immunol.
51
:
2137
2150
.
68
Astry
,
B.
,
S. H.
Venkatesha
,
K. D.
Moudgil
.
2015
.
Involvement of the IL-23/IL-17 axis and the Th17/Treg balance in the pathogenesis and control of autoimmune arthritis
.
Cytokine
74
:
54
61
.
69
Paradowska-Gorycka
,
A.
,
A.
Wajda
,
K.
Romanowska-Próchnicka
,
E.
Walczuk
,
E.
Kuca-Warnawin
,
T.
Kmiolek
,
B.
Stypinska
,
E.
Rzeszotarska
,
D.
Majewski
,
P. P.
Jagodzinski
, et al
2020
.
Th17/Treg-related transcriptional factor expression and cytokine profile in patients with rheumatoid arthritis
.
Front. Immunol.
11
:
572858
.
70
Morishima
,
N.
,
I.
Mizoguchi
,
K.
Takeda
,
J.
Mizuguchi
,
T.
Yoshimoto
.
2009
.
TGF-β is necessary for induction of IL-23R and Th17 differentiation by IL-6 and IL-23
.
Biochem. Biophys. Res. Commun.
386
:
105
110
.
71
Ghoreschi
,
K.
,
A.
Laurence
,
X.-P.
Yang
,
C. M.
Tato
,
M. J.
McGeachy
,
J. E.
Konkel
,
H. L.
Ramos
,
L.
Wei
,
T. S.
Davidson
,
N.
Bouladoux
, et al
2010
.
Generation of pathogenic TH17 cells in the absence of TGF-β signalling
.
Nature
467
:
967
971
.
72
Mosher
,
A. A.
,
M. W.
Tsoulis
,
J.
Lim
,
C.
Tan
,
S. K.
Agarwal
,
N. A.
Leyland
,
W. G.
Foster
.
2019
.
Melatonin activity and receptor expression in endometrial tissue and endometriosis
.
Hum. Reprod.
34
:
1215
1224
.
73
Debrock
,
S.
,
B.
Destrooper
,
S.
Vander Perre
,
J. A.
Hill
,
T. M.
D’Hooghe
.
2002
.
Tumor necrosis factor-alpha inhibits the in vitro adhesion of endometrial epithelial cells to mesothelial cells
.
Fertil. Steril.
77
:
S15
.
74
Miller
,
J. E.
,
S. P.
Monsanto
,
S. H.
Ahn
,
K.
Khalaj
,
A. T.
Fazleabas
,
S. L.
Young
,
B. A.
Lessey
,
M.
Koti
,
C.
Tayade
.
2017
.
Interleukin-33 modulates inflammation in endometriosis
.
Sci. Rep.
7
:
17903
.
75
Fan
,
H.
2019
.
In-vitro models of human endometriosis
.
Exp. Ther. Med.
19
:
1617
1625
.
76
Tang
,
C.
,
S.
Chen
,
H.
Qian
,
W.
Huang
.
2012
.
Interleukin-23: as a drug target for autoimmune inflammatory diseases
.
Immunology
135
:
112
124
.
77
Yang
,
K.
,
A. S. W.
Oak
,
B. E.
Elewski
.
2021
.
Use of IL-23 inhibitors for the treatment of plaque psoriasis and psoriatic arthritis: a comprehensive review
.
Am. J. Clin. Dermatol.
22
:
173
192
.
78
Li
,
W.
,
R.
Ghamrawi
,
W.
Haidari
,
S. R.
Feldman
.
2020
.
Risankizumab for the treatment of moderate to severe plaque psoriasis
.
Ann. Pharmacother.
54
:
380
387
.
79
Graier
,
T.
,
W.
Weger
,
C.
Jonak
,
P.
Sator
,
C.
Zikeli
,
K.
Prillinger
,
C.
Sassmann
,
B.
Gruber
,
W.
Saxinger
,
G.
Ratzinger
, et al
2022
.
Real-world effectiveness of anti-interleukin-23 antibodies in chronic plaque-type psoriasis of patients from the Austrian Psoriasis Registry (PsoRA)
.
Sci. Rep.
12
:
15078
.
80
Yuan
,
N.
,
G.
Yu
,
D.
Liu
,
X.
Wang
,
L.
Zhao
.
2019
.
An emerging role of interleukin-23 in rheumatoid arthritis
.
Immunopharmacol. Immunotoxicol.
41
:
185
191
.

Supplementary data