ArticlePDF Available

Targeting host tyrosine kinase receptor EphA2 signaling via small-molecule ALW-II-41-27 inhibits macrophage pro-inflammatory signaling responses to Pneumocystis carinii β-glucans

American Society for Microbiology
Antimicrobial Agents and Chemotherapy
Authors:

Abstract and Figures

Pneumocystis jirovecii, the fungus that causes Pneumocystis jirovecii pneumonia (PJP), is a leading cause of morbidity and mortality in immunocompromised individuals. We have previously shown that lung epithelial cells can bind Pneumocystis spp. β-glucans via the EphA2 receptor, resulting in activation and release of proinflammatory cytokines. Herein, we show that in vivo Pneumocystis spp. β-glucans activation of the inflammatory signaling cascade in macrophages can be pharmacodynamically inhibited with the EphA2 receptor small-molecule inhibitor ALW-II-41-27. In vitro, when ALW-II-41-27 is administrated via intraperitoneal to mice prior to the administration of highly proinflammatory Saccharomyces cerevisiae β-glucans in the lung, a significant reduction in TNF-alpha release was noted in the ALW-II-41-27 pre-treated group. Taken together, our data suggest that targeting host lung macrophage activation via EphA2 receptor-fungal β-glucans interactions with ALW-II-41-27 or other EphA2 receptor kinase targeting inhibitors might be an attractive and viable strategy to reduce detrimental lung inflammation associated with PJP.
This content is subject to copyright. Terms and conditions apply.
Eukaryotic Cells | Short Form
Targeting host tyrosine kinase receptor EphA2 signaling
via small-molecule ALW-II-41-27 inhibits macrophage pro-
inammatory signaling responses to Pneumocystis cariniiβ-
glucans
Theodore J. Kottom,1,2 Eva M. Carmona,1,2 Andrew H. Limper1,2
AUTHOR AFFILIATIONS See aliation list on p. 8.
ABSTRACT Pneumocystis jirovecii, the fungus that causes Pneumocystis jirovecii
pneumonia (PJP), is a leading cause of morbidity and mortality in immunocompromised
individuals. We have previously shown that lung epithelial cells can bind Pneumocystis
spp. β-glucans via the EphA2 receptor, resulting in activation and release of proinam-
matory cytokines. Herein, we show that in vivo Pneumocystis spp. β-glucans activation
of the inammatory signaling cascade in macrophages can be pharmacodynamically
inhibited with the EphA2 receptor small-molecule inhibitor ALW-II-41-27. In vitro, when
ALW-II-41-27 is administrated via intraperitoneal to mice prior to the administration of
highly proinammatory Saccharomyces cerevisiae β-glucans in the lung, a signicant
reduction in TNF-alpha release was noted in the ALW-II-41-27 pre-treated group. Taken
together, our data suggest that targeting host lung macrophage activation via EphA2
receptor-fungal β-glucans interactions with ALW-II-41-27 or other EphA2 receptor kinase
targeting inhibitors might be an attractive and viable strategy to reduce detrimental lung
inammation associated with PJP.
KEYWORDS Pneumocystis, pneumonia, inammation, mycology
EphA2 receptor is a member of the receptor tyrosine kinase family. It is a trans
membrane receptor containing an extracellular region that binds activating ligands
(ephrins) and more recently discovered to bind fungal β-glucans (1–3), leading to
the activation of the intracellular tyrosine kinase domain (4). Others have shown the
specicity of EphA2 receptor for fungal β-glucans and activation (phosphorylation) via
binding of this receptor to zymosan-coated beads as well as Candida albicans, Aspergillus
fumigatus, and Rhizopus delemar fungal organisms and the absence of binding and
phosphorylation of the receptor by Staphylococcus aureus and Escherichia coli bacteria
(3). More recently, our lab has shown that puried recombinant EphA2 protein alone can
specically and signicantly bind Pneumocystis β-glucans, verifying that the protein is
a receptor for fungal β-glucans (1). Traditionally, the EphA2 receptor kinase pathway
has important roles in carcinogenesis, pathological angiogenesis, and inammation
in atherosclerosis (5–7). Expression of EphA2 receptor is high in both epithelial and
endothelial cells (4). More recently, this receptor pathway has also emerged as an
important regulatory pathway for host defense against microbial pathogens, including
bacterial, viral, and fungal pathogens (2, 8–11). For example, it has been demonstrated
previously that the EphA2 receptor is active in the binding and trapping of the hook
worm Nippostrongylus brasiliensis by bone marrow-derived macrophages, suggesting a
role for the EphA2 receptor in macrophage/microbial pathogenesis (12).
February 2024 Volume 68 Issue 2 10.1128/aac.00811-23 1
Editor Helen Boucher, Tufts University - New
England Medical Center, Boston, Massachusetts, USA
Address correspondence to Theodore J. Kottom,
kottom.theodore@mayo.edu.
The authors declare no conict of interest.
See the funding table on p. 8.
Received 19 June 2023
Accepted 3 December 2023
Published 11 January 2024
Copyright © 2024 American Society for
Microbiology. All Rights Reserved.
ALW-II-41-27 is a small-molecule inhibitor that has been demonstrated to selectively
bind to the ATP-binding pocket of the EphA2 receptor kinase domain (13). Although the
compound has been shown to have EC50 values on a number of kinases in vitro at 10 uM
(including CSF1R, DDR1/2, Kit, Lck, and PDGFRα/β) (14), this and other recent studies
show EC50 eects of the inhibitor at 200 nM or less on EphA2 kinase activity, allowing
more directed and targeted therapeutic dosing (14–16). The inhibitor has been used
in the past to inhibit cancer cell growth in vitro and in vivo (17–20) and more recently
to inhibit uropathogenic bacteria adherence to bladder epithelial cells and to reduce
the oxidative stress and proinammatory host response in LPS(lipopolysaccharide)-trea
ted colonic cells (16). Therefore, we evaluated this inhibitor to determine whether it
might serve as an alternative agent to reduce proinammatory events resulting from
interactions with Pneumocystis β-glucans through the EphA2 receptor kinase signaling
pathway in macrophages in vitro (1), as well as the inhibitor’s ability to reduce yeast
β-glucan-driven proinammatory cytokine release in the lung. Measured and timed
therapeutic inhibition of EphA2 signaling may aid in mitigating harmful downstream
inammatory events that result during anti-Pneumocystis pneumonia (PCP) treatment.
We and others have shown that killing of fungal organisms as a result of this action
exposes highly inammatory β-glucan carbohydrate (21–29).
Cells [2 × 105 RAW macrophages (American Type Culture Collection)] for each
experimental condition were plated in ve wells of a 96-well microtiter plate and
incubated for 4 hours. After 4 hours, ALW-II-41-27 purchased from Sigma-Aldrich was
pre-incubated with the RAW cells for 60 minutes. Next, 100 ug/mL of Pneumocystis
carinii (Pc) β-glucans (28) was added to the wells, and the plates were centrifuged at
500 × g to synchronize the carbohydrate/macrophage interactions. Plates were then
placed at 37°C for 60 minutes. Next, cells were washed with 1× PBS, lysed, and protein
quantication determined. Total proteins in equal amounts were loaded and separated
by polyacrylamide gel electrophoresis. Finally, proteins were transferred to nylon for
Western blotting and incubated with antibodies to phospho-p38 or ERK1/2 as well as
total p38 and ERK1/2 (Cell Signaling Technology) to demonstrate equal loading control.
Protein phosphorylation kinetics were quantied with Image Studio Lite (LI-COR). All
experiments were repeated four to ve times. Activation of MAPK (mitogen-activated
protein kinase) is well documented in macrophage responses to Pneumocystis infection
(29–31). Herein, we demonstrate that the specic EphA2 receptor inhibitor ALW-II-41-27
can indeed signicantly inhibit Pc β-glucan-induced phosphorylation of both p38
(Fig. 1A) and ERK1/2 (Fig. 2) in a dose-dependent manner measured by Western blot
with similar ALW-II-41-27 concentrations previously published for cervical, endometrial,
nasopharyngeal, and colonic cells inhibitor studies (13, 15, 16, 32). Western blots for
both phospho-p38 (Fig. 1B) and phospho-ERK1/2 (Fig. 2B) were quantied by densi
tometry analysis against their respective total proteins. Next, we wanted to determine
whether ALW-II-41-27 inhibition would not only result in decreased MAPK phosphoryla
tion but also result in downstream reduced secretion of the proinammatory cytokine
TNF-alpha. To determine this, RAW macrophages were seeded as above. After 4 hours,
ALW-II-41-27 was incubated with the RAW cells for 60 minutes. Cell media were removed,
and 100 ug/mL of Pc β-glucans plus ALW-II-41-27 compound was applied to the cells,
centrifuged as described above, and incubated for 18 hours at 37°C. Supernatants were
then collected and assayed for TNF-alpha by ELISA (26). As shown in Fig. 3A, ALW-II-41-27
signicantly reduced TNF-alpha release in a dose-dependent fashion. To determine if
ALW-II-41-27 could signicantly alter the inammatory potential of macrophages already
undergoing activated proinammatory cytokine release via Pc β-glucan stimulation, we
also added ALW-II-41-27 post 60 minutes after Pc β-glucan stimulation. Similar to Fig. 3A,
we also noted signicant suppression of RAW cell TNF-alpha release in a dose response
fashion in these experiments (Fig. 3B). Next, to conrm these ndings in primary cells,
mouse lung alveolar macrophages were isolated as previously described (31). After
allowing the macrophages to bind for 2 hours, ALW-II-41-27 was incubated with the
macrophages for 60 minutes. As stated above, the supernatant was removed, and Pc
Short Form Antimicrobial Agents and Chemotherapy
February 2024 Volume 68 Issue 2 10.1128/aac.00811-23 2
β-glucans (100 ug/mL) plus ALW-II-41-27 compound was added to the cells, centrifuged
as described above, and incubated for 18 hours at 37°C . Supernatants were then
collected and assayed for TNF-alpha by ELISA as noted above. Similar to what was shown
in Fig. 3, ALW-II-41-27 addition to Pc β-glucan-induced primary mouse lung alveolar
FIG 1 (A) ALW-II-41-27 can signicantly inhibit Pc β-glucan-induced ERK1/2 phosphorylation in a dose-dependent manner. Representative blot of four separate
experiments. (B) The phospho ERK1/2 signals were quantied with Image Studio Lite software and normalized to total ERK1/2 levels. Initial analysis was rst
performed with ANOVA. If ANOVA indicated overall dierences, subsequent group analysis was then performed by a two-sample unpaired Student t test for
normally distributed variables. Error bars show SD from the mean. *P < 0.05, ns, non-signicant; ANOVA, analysis of variance.
FIG 2 (A) ALW-II-41-27 can signicantly inhibit Pc β-glucan-induced p38 phosphorylation. Representative blot of ve separate experiments. (B) The phospho p38
signals were quantied with Image Studio Lite software and normalized to total p38 levels. Similar to phospho ERK1/2 quantitation above, analysis of phospho
p38 quantication by ANOVA was rst performed. If ANOVA indicated overall dierences, subsequent group analysis was then performed by a two-sample
unpaired Student t test for normally distributed variables. Error bars show SD from the mean. ****P < 0.0001. ANOVA, analysis of variance.
Short Form Antimicrobial Agents and Chemotherapy
February 2024 Volume 68 Issue 2 10.1128/aac.00811-23 3
macrophages signicantly reduced TNF-alpha cytokine release from these cells (Fig. 4),
conrming the ability of the compound to inhibit proinammatory response in native
lung alveolar macrophages. To determine if ALW-II-41-27 could also aect TNF-alpha
release from RAW macrophages infected with live P. murina organisms, 2 × 105 cells were
plated in duplicate wells of a 96-well plate for 4 hours as above. Next, ALW-II-41-27 was
incubated with the RAW cells for 60 minutes. Finally, P. murina organisms in the presence
of ALW-II-41-27 were applied at a multiplicity of infection of 2:1 to the cell supernatant,
and the plates centrifuged at 500 × g to synchronize the fungal organism/RAW cell
interactions and incubated for 18 hours at 37°C. Supernatants were then collected
and tested for TNF-alpha release. Similar to what we reported above for Pc β-glucan
alone, TNF-alpha was signicantly induced in the presence of live fungal organisms. The
addition of 1,000 nM of ALW-II-41-27 to the media prior to the addition of the fungal
organisms signicantly suppressed TNF-alpha release from the cultured macrophages
(Fig. 5). These results suggest the exciting possibility of targeting the EphA2 tyrosine
kinase receptor pathway in those individuals with active PCP to reduce exuberant lung
inammation. On this note, as an initial proof-of-concept experiment to determine if
ALW-II-41-27 administered to mice could inhibit fungal β-glucan-driven proinammatory
cytokine response in the lung, we pre-treated mice with intraperitoneal (IP) injections of
either ALW-II-41-27 or the vehicle control 20 hours prior to the addition of Saccharomyces
cerevisiae β-glucans. After 20 hours, the yeast β-glucans were administered via intratra
cheally (IT). The following day (24 hours), the mice were sacriced, and total lung protein
lysates measured for TNF-alpha by ELISA. Remarkably, we noted that ALW-II-41-27 could
indeed signicantly reduce TNF-alpha protein levels in the lungs versus the vehicle
control group in yeast β-glucan-challenged mouse lungs (Fig. 6).
FIG 3 ALW-II-41-27 administered 60 minutes prior to (A) or 60 minutes after (B) Pc β-glucans signicantly dampens RAW 264.7 production of TNF-alpha in
vitro. Data are the ±SEM for at least four separate experiments. The TNF-alpha data analysis was initially rst performed with ANOVA. If ANOVA indicated overall
dierences, subsequent group analysis was then performed by a two-sample unpaired Student t test for normally distributed variables. Error bars show SD from
the mean. *P < 0.05, **P < 0.01, and ****P < 0.0001. ANOVA, analysis of variance.
Short Form Antimicrobial Agents and Chemotherapy
February 2024 Volume 68 Issue 2 10.1128/aac.00811-23 4
A number of reports have shown the importance of the EphA2 receptor pathway
in organism attachment and host immune recognition to microbial pathogens (12, 33–
35). Recently, we also have reported that EphA2 can bind Pneumocystis glucans and
is involved in lung epithelial cell proinammatory response to the organism’s cell wall
carbohydrate (1).
FIG 4 In primary mouse lung alveolar macrophages, ALW-II-41-27 signicantly dampens production of TNF-alpha in vitro in
the presence of Pc β-glucans. Data are the ±SEM for at least three separate experiments. Initial analysis was rst performed
with ANOVA. If ANOVA indicated overall dierences, subsequent group analysis was then performed by a two-sample
unpaired Student t test for normally distributed variables. Error bars show SD from the mean. *P < 0.05, and ****P < 0.0001.
ANOVA, analysis of variance.
Short Form Antimicrobial Agents and Chemotherapy
February 2024 Volume 68 Issue 2 10.1128/aac.00811-23 5
In vitro and in vivo data presented here suggest promising preliminary evidence
that therapeutically targeting the EphA2 receptor in PCP-infected individuals undergo
ing standard anti-PCP treatment may provide additional anti-inammatory relief as a
result of fungal killing and the release of highly proinammatory β-glucans during the
treatment of Pneumocystis pneumonia.
FIG 5 ALW-II-41-27 can signicantly inhibit P. murina-induced TNF-alpha secretion in mouse macro
phages. Data represent the mean ± SEM for at least three separate experiments. Initial analysis was rst
performed with ANOVA. If ANOVA indicated overall dierences, subsequent group analysis was then
performed by a two-sample unpaired Student t-test for normally distributed variables. Error bars depict
the SD from the mean. *P < 0.05 and **P < 0.01. ANOVA, analysis of variance.
Short Form Antimicrobial Agents and Chemotherapy
February 2024 Volume 68 Issue 2 10.1128/aac.00811-23 6
FIG 6 The eects of IP injection of ALW-II-41-27 on Saccharomyces cerevisiae β-glucan proinammatory response in the
lung. Twenty hours prior to administering 100 ug/mL of S. cerevisiae β-glucans via IT, mice were administered 0.1 mg/kg
ALW-II-41-27 or the vehicle (Methocel) control via IP. After 18 hours post drug or vehicle treatment, mice were administered
another 0.1 mg/kg of ALW-II-41-27 via IP or vehicle stated. After 2 hours, mice were administered 100 ug/mL of S. cerevisiae
β-glucans via IT administration. The following day, mice were sacriced, and total lung protein lysates (200 ug total) measured
for TNF-alpha by ELISA. Bar graph represents the results from 11 to 12 mice per group. If ANOVA indicated overall dierences,
subsequent group analysis was then performed by two-sample unpaired Student t test for normally distributed variables.
Error bars show SD from the mean. ****P < 0.0001. ANOVA, analysis of variance.
Short Form Antimicrobial Agents and Chemotherapy
February 2024 Volume 68 Issue 2 10.1128/aac.00811-23 7
AUTHOR AFFILIATIONS
1Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester,
Minnesota, USA
2Thoracic Diseases Research Unit, Mayo Clinic College of Medicine, Rochester, Minnesota,
USA
AUTHOR ORCIDs
Theodore J. Kottom http://orcid.org/0000-0003-0364-3311
Andrew H. Limper http://orcid.org/0000-0001-5671-6874
FUNDING
Funder Grant(s) Author(s)
HHS | National Institutes of Health (NIH) 1R21AI181542-01 Eva M. Carmona
AUTHOR CONTRIBUTIONS
Theodore J. Kottom, Conceptualization, Data curation, Formal analysis, Funding
acquisition, Investigation, Methodology, Writing – original draft | Eva M. Carmona, Formal
analysis, Writing – review and editing | Andrew H. Limper, Conceptualization, Formal
analysis, Funding acquisition, Supervision, Writing – original draft
REFERENCES
1. Kottom TJ, Schaefbauer K, Carmona EM, Limper AH. 2022. EphA2 is a
lung epithelial cell receptor for pneumocystis β-glucans. J Infect Dis
225:525–530. https://doi.org/10.1093/infdis/jiab384
2. Swidergall M, Solis NV, Millet N, Huang MY, Lin J, Phan QT, Lazarus MD,
Wang Z, Yeaman MR, Mitchell AP, Filler SG. 2021. Activation of EphA2-
EGFR signaling in oral epithelial cells by Candida albicans virulence
factors. PLoS Pathog 17:e1009221. https://doi.org/10.1371/journal.ppat.
1009221
3. Swidergall M, Solis NV, Lionakis MS, Filler SG. 2018. EphA2 is an epithelial
cell pattern recognition receptor for fungal β-glucans. Nat Microbiol
3:1074. https://doi.org/10.1038/s41564-018-0188-5
4. Zapata-Mercado E, Biener G, McKenzie DM, Wimley WC, Pasquale EB,
Raicu V, Hristova K. 2022. The ecacy of receptor tyrosine kinase EphA2
autophosphorylation increases with EphA2 oligomer size. J Biol Chem
298:102370. https://doi.org/10.1016/j.jbc.2022.102370
5. Wilson K, Shiuan E, Brantley-Sieders DM. 2021. Oncogenic functions and
therapeutic targeting of EphA2 in cancer. Oncogene 40:2483–2495.
https://doi.org/10.1038/s41388-021-01714-8
6. Pasquale EB. 2010. Eph receptors and ephrins in cancer: bidirectional
signalling and beyond. Nat Rev Cancer 10:165–180. https://doi.org/10.
1038/nrc2806
7. Funk SD, Orr AW. 2013. Ephs and ephrins resurface in inammation,
immunity, and atherosclerosis. Pharmacol Res 67:42–52. https://doi.org/
10.1016/j.phrs.2012.10.008
8. Swidergall M, Solis NV, Wang Z, Phan QT, Marshall ME, Lionakis MS,
Pearlman E, Filler SG. 2019. EphA2 is a neutrophil receptor for Candida
albicans that stimulates antifungal activity during oropharyngeal
infection. Cell Rep 28:423–433. https://doi.org/10.1016/j.celrep.2019.06.
020
9. Su C, Wu L, Chai Y, Qi J, Tan S, Gao GF, Song H, Yan J. 2020. Molecular
basis of EphA2 recognition by gHgL from gammaherpesviruses. Nat
Commun 11:5964. https://doi.org/10.1038/s41467-020-19617-9
10. Zhang J, Yuan J, Wang L, Zheng Z, Ran H, Liu F, Li F, Tang X, Zhang J, Ni
Q, Zou L, Huang Y, Feng S, Xia X, Wan Y. 2020. MiR-26a targets EphA2 to
resist intracellular Listeria monocytogenes in macrophages. Mol Immunol
128:69–78. https://doi.org/10.1016/j.molimm.2020.09.016
11. de Boer ECW, van Gils JM, van Gils MJ. 2020. Ephrin-Eph signaling usage
by a variety of viruses. Pharmacol Res 159:105038. https://doi.org/10.
1016/j.phrs.2020.105038
12. Bouchery T, Volpe B, Doolan R, Coakley G, Moyat M, Esser-von Bieren J,
Wickramasinghe LC, Hibbs ML, Sotillo J, Camberis M, Le Gros G, Khan N,
Williams D, Harris NL. 2022. β-glucan receptors on IL-4 activated
macrophages are required for hookworm larvae recognition and
trapping. Immunol Cell Biol 100:223–234. https://doi.org/10.1111/imcb.
12536
13. Xiang Y-P, Xiao T, Li Q-G, Lu S-S, Zhu W, Liu Y-Y, Qiu J-Y, Song Z-H, Huang
W, Yi H, Tang Y-Y, Xiao Z-Q. 2020. Y772 phosphorylation of EphA2 is
responsible for EphA2-dependent NPC nasopharyngeal carcinoma
growth by Shp2/Erk-1/2 signaling pathway. Cell Death Dis 11:709. https:/
/doi.org/10.1038/s41419-020-02831-0
14. Choi Y, Syeda F, Walker JR, Finerty PJ, Cuerrier D, Wojciechowski A, Liu Q ,
Dhe-Paganon S, Gray NS. 2009. Discovery and structural analysis of Eph
receptor tyrosine kinase inhibitors. Bioorg Med Chem Lett 19:4467–
4470. https://doi.org/10.1016/j.bmcl.2009.05.029
15. Li X, Li D, Ma R. 2022. ALW-II-41-27, an EphA2 inhibitor, inhibits
proliferation, migration and invasion of cervical cancer cells via
inhibition of the RhoA/ROCK pathway. Oncol Lett 23:129. https://doi.
org/10.3892/ol.2022.13249
16. Zeng L, Li K, Wei H, Hu J, Jiao L, Yu S, Xiong Y. 2018. A novel EphA2
inhibitor exerts benecial eects in PI-IBS in vivo and in vitro models via
Nrf2 and NF-κB signaling pathways. Front Pharmacol 9:272. https://doi.
org/10.3389/fphar.2018.00272
17. Martini G, Cardone C, Vitiello PP, Belli V, Napolitano S, Troiani T,
Ciardiello D, Della Corte CM, Morgillo F, Matrone N, Sforza V, Papaccio G,
Desiderio V, Paul MC, Moreno-Viedma V, Normanno N, Rachiglio AM,
Tirino V, Maiello E, Latiano TP, Rizzi D, Signoriello G, Sibilia M, Ciardiello
F, Martinelli E. 2019. EphA2 is a predictive biomarker of resistance and a
potential therapeutic target for improving antiepidermal growth factor
receptor therapy in colorectal cancer. Mol Cancer Ther 18:845–855.
https://doi.org/10.1158/1535-7163.MCT-18-0539
18. Ruan H, Li S, Bao L, Zhang X. 2020. Enhanced YB1/EphA2 axis signaling
promotes acquired resistance to sunitinib and metastatic potential in
renal cell carcinoma. Oncogene 39:6113–6128. https://doi.org/10.1038/
s41388-020-01409-6
19. Amato KR, Wang S, Tan L, Hastings AK, Song W , Lovly CM, Meador CB, Ye
F, Lu P, Balko JM, Colvin DC, Cates JM, Pao W, Gray NS, Chen J. 2016.
EphA2 blockade overcomes acquired resistance to EGFR kinase
inhibitors in lung cancer. Cancer Res 76:305–318. https://doi.org/10.
1158/0008-5472.CAN-15-0717
Short Form Antimicrobial Agents and Chemotherapy
February 2024 Volume 68 Issue 2 10.1128/aac.00811-23 8
20. Amato KR, Wang S, Hastings AK, Youngblood VM, Santapuram PR, Chen
H, Cates JM, Colvin DC, Ye F, Brantley-Sieders DM, Cook RS, Tan L, Gray
NS, Chen J. 2014. Genetic and pharmacologic inhibition of EPHA2
promotes apoptosis in NSCLC. J Clin Invest 124:2037–2049. https://doi.
org/10.1172/JCI72522
21. Kutty G, Davis AS, Ferreyra GA, Qiu J, Huang DW, Sassi M, Bishop L,
Handley G, Sherman B, Lempicki R, Kovacs JA. 2016. β-glucans are
masked but contribute to pulmonary inammation during Pneumocystis
pneumonia. J Infect Dis 214:782–791. https://doi.org/10.1093/infdis/
jiw249
22. Sassi M, Kutty G, Ferreyra GA, Bishop LR, Liu Y, Qiu J, Huang DW, Kovacs
JA. 2018. The major surface glycoprotein of Pneumocystis murina does
not activate dendritic cells. J Infect Dis 218:1631–1640. https://doi.org/
10.1093/infdis/jiy342
23. Evans HM, Bryant GL, Garvy BA. 2016. The life cycle stages of Pneumocys
tis murina have opposing eects on the immune response to this
opportunistic, fungal pathogen. Infect Immun 84:3195–3205. https://
doi.org/10.1128/IAI.00519-16
24. Evans HM, Garvy BA. 2018. The trophic life cycle stage of Pneumocystis
species induces protective adaptive responses without inammation-
mediated progression to pneumonia. Med Mycol 56:994–1005. https://
doi.org/10.1093/mmy/myx145
25. Evans HM, Simpson A, Shen S, Stromberg AJ, Pickett CL, Garvy BA,
Pirofski L. 2017. The trophic life cycle stage of the opportunistic fungal
pathogen Pneumocystis murina hinders the ability of dendritic cells to
stimulate CD4+ T cell responses. Infect Immun 85:e00396-17. https://doi.
org/10.1128/IAI.00396-17
26. Kottom TJ, Hebrink DM, Jenson PE, Gudmundsson G, Limper AH. 2015.
Evidence for proinammatory β-1,6 glucans in the Pneumocystis carinii
cell wall. Infect Immun 83:2816–2826. https://doi.org/10.1128/IAI.00196-
15
27. Linke MJ, Ashbaugh A, Collins MS, Lynch K, Cushion MT. 2013.
Characterization of a distinct host response prole to Pneumocystis
murina Asci during clearance of pneumocystis pneumonia. Infect
Immun 81:984–995. https://doi.org/10.1128/IAI.01181-12
28. Hahn PY, Evans SE, Kottom TJ, Standing JE, Pagano RE, Limper AH. 2003.
Pneumocystis carinii cell wall β-glucan induces release of macrophage
inammatory protein-2 from alveolar epithelial cells via a lactosylcera
mide-mediated mechanism. J Biol Chem 278:2043–2050. https://doi.
org/10.1074/jbc.M209715200
29. Wang J, Wright TW, Gigliotti F. 2011. Immune modulation as adjunctive
therapy for Pneumocystis pneumonia. Interdiscip Perspect Infect Dis
2011:918038. https://doi.org/10.1155/2011/918038
30. Tachado SD, Zhang J, Zhu J, Patel N, Cushion M, Koziel H. 2007.
Pneumocystis-mediated IL-8 release by macrophages requires
coexpression of mannose receptors and TLR2. J Leukoc Biol 81:205–211.
https://doi.org/10.1189/jlb.1005580
31. Kottom TJ, Nandakumar V, Hebrink DM, Carmona EM, Limper AH. 2020.
A critical role for CARD9 in pneumocystis pneumonia host defence. Cell
Microbiol 22:e13235. https://doi.org/10.1111/cmi.13235
32. Hudecek R, Kohlova B, Siskova I, Piskacek M, Knight A. 2021. Blocking of
EphA2 on endometrial tumor cells reduces susceptibility to Vδ1 gamma-
delta T-cell-mediated killing. Front Immunol 12:752646. https://doi.org/
10.3389/mmu.2021.752646
33. Chainarin S, Jaihan U, Tapaopong P, Kongngen P, Kunkeaw N, Cui L,
Sattabongkot J, Nguitragool W, Roobsoong W. 2022. Overexpression of
hepatocyte EphA2 enhances liver-stage infection by Plasmodium vivax.
Sci Rep 12:21542. https://doi.org/10.1038/s41598-022-25281-4
34. Shaikh MS, Islam F, Gargote PP, Gaikwad RR, Dhupe KC, Khan SL,
Siddiqui FA, Tapadiya GG, Ali SS, Dey A, Emran TB. 2022. Potential EphA2
receptor blockers involved in cerebral malaria from Taraxacum ocinale,
Tinospora cordifolia, Rosmarinus ocinalis and Ocimum basilicum: a
computational approach. Pathogens 11:1296. https://doi.org/10.3390/
pathogens11111296
35. Prakash PS, Kruse A, Vogel C, Schagdarsurengin U, Wagenlehner F. 2022.
Targeting host tyrosine kinase receptor EphA2 signaling aects
uropathogen infection in human bladder epithelial cells. Pathogens
11:1176. https://doi.org/10.3390/pathogens11101176
Short Form Antimicrobial Agents and Chemotherapy
February 2024 Volume 68 Issue 2 10.1128/aac.00811-23 9
... 33 Previously mainly studied in inhibiting cancer, ALW-II-41-27 has recently been discovered potential therapeutic effects in benign diseases. 34,35 And in our primary cell, it is observed that genetic and pharmacological targeting EphA2 significantly suppressed proliferation, migration, and invasion, consistent with Matzuk. 8 Interestingly, EphA2 inhibition did not show significant inhibitory or pro-apoptotic effects in endometriosis. ...
Article
Full-text available
Endometriosis is a benign disease with similar characteristics to tumors. Recent studies have found that the erythropoietin-producing hepatoma receptor A2 (EphA2) has the dual effect of promoting tumor and inhibiting tumor. The objective of this study was to explore the specific regulatory mechanism of EphA2 in endometriosis. The expression level of Eph protein family in endometriosis was analyzed by bioinformatics method. At the clinical level, qPCR, Western blot and immunohistochemistry were used to verify the correlation between increased EphA2 levels and endometriosis. The effects of blocking EphA2 on cell migration, invasion, proliferation and apoptosis of primary eutopic endometriotic stromal cells were explored in vitro. Our study indicated that EphA2 expression was elevated in endometriosis patients, and blocking EphA2 in vitro inhibited cell proliferation, migration and invasion through AMPK signaling pathway. Targeting EphA2 can inhibit the progression of endometriosis through the AMPK signaling pathway.
... Furthermore, our laboratory has recently shown that in mouse lung epithelial cells, the EphA2 receptor is also involved in binding Pneumocystis and that β-glucans from the organism can activate downstream signaling through EphA2 engagement, leading to release of lung epithelial IL-6 release [7]. We have also recently published that a small molecule inhibitor of the EphA2 receptor, termed ALW-II-41-27, can dampen in vitro macrophage immune proinflammatory response to Pneumocystis β-glucans [8]. These data provide initial proof of concept that timed intervention of ALW-II-41-27 during or after anti-Pneumocystis treatment may greatly improve the deleterious effects on the host caused by organism killing and release of proinflammatory carbohydrates [9]. ...
Article
Full-text available
The EphA2 receptor inhibitor ALW-II-41-27 has proven to be an effective in vitro antagonist of Pneumocystis β-glucan-induced proinflammatory signaling. This suggests its potential as a candidate for initial anti-inflammatory drug testing in the rodent model of Pneumocystis pneumonia (PCP). Initially, single-dose intraperitoneal (IP) injections of ALW-II-41-27 were administered at concentrations of 0, 10, 15, 20, and 30 mg/kg over a 24-h treatment period. Pharmacokinetics were assessed in plasma, bronchoalveolar lavage fluid (BALF), and epithelial lining fluid (ELF). Following these assessments, a final single mg/kg dosing was determined. Mice received daily IP injections of either vehicle or 20.0 mg/kg of ALW-II-41-27 for 10 days, with their weights recorded daily. On day 11, mice were weighed and euthanized. Lungs, liver, and kidneys were harvested for H&E staining and pathology scoring. Lung samples were further analyzed for proinflammatory cytokines using enzyme-linked immunosorbent assay (ELISA) and extracellular matrix production using quantitative PCR (qPCR). Postmortem blood collection was conducted for complete blood count (CBC) blood chemistry analysis. Lastly, ALW-II-41-27 was administered to mice prior to fungal β-glucans challenge to determine in vivo effects on lung inflammation. This report describes the PK assessment of ALW-II-41-27 given via IP in C57BL/6 mice. After PK data were generated, we tested ALW-II-41-27 at 20 mg/kg IP in mice and noted no significant changes in daily or final weight gain. ELISA results of proinflammatory cytokines from lung tissues showed no major differences in the respective groups. qPCR analysis of extracellular matrix transcripts were statistically similar. Examination and pathology scoring of H&E slides from lung, liver, and kidney in all groups and subsequent pathology scoring showed no significant toxicity. Blood chemistry and CBC analyses revealed no major abnormalities. Additionally, administering ALW-II-41-27 before intratracheal inoculation of fungal β-glucans, known to induce a strong proinflammatory response in the lungs, significantly reduced lung tissue IL-1β levels. In our initial general safety and toxicology assessments, ALW-II-41-27 displayed no inherent safety concerns in the analyzed parameters. These data support broader in vivo testing of the inhibitor as a timed adjunct therapy to the deleterious proinflammatory host immune response often associated with anti-Pneumocystis therapy.
Article
Full-text available
The liver is the first destination of malaria parasites in humans. After reaching the liver by the blood stream, Plasmodium sporozoites cross the liver sinusoid epithelium, enter and exit several hepatocytes, and eventually invade a final hepatocyte host cell. At present, the mechanism of hepatocyte invasion is only partially understood, presenting a key research gap with opportunities for the development of new therapeutics. Recently, human EphA2, a membrane-bound receptor tyrosine kinase, was implicated in hepatocyte infection by the human malaria parasite Plasmodium falciparum and the rodent parasite Plasmodium yoelii, but its role is not known for Plasmodium vivax, a major human parasite whose liver infection poses a specific challenge for malaria treatment and elimination. In this study, the role of EphA2 in P. vivax infection was investigated. It was found that surface expression of several recombinant fragments of EphA2 enhanced the parasite infection rate, thus establishing its role in P. vivax infection. Furthermore, a new permanent cell line (EphA2Extra-HC04) expressing the whole extracellular domain of EphA2 was generated. This cell line supports a higher rate of P. vivax infection and is a valuable tool for P. vivax liver-stage research.
Article
Full-text available
Cerebral malaria (CM) is a severe manifestation of parasite infection caused by Plasmodium species. In 2018, there were approximately 228 million malaria cases worldwide, resulting in about 405,000 deaths. Survivors of CM may live with lifelong post-CM consequences apart from an increased risk of childhood neurodisability. EphA2 receptors have been linked to several neurological disorders and have a vital role in the CM-associated breakdown of the blood–brain barrier. Molecular docking (MD) studies of phytochemicals from Taraxacum officinale, Tinospora cordifolia, Rosmarinus officinalis, Ocimum basilicum, and the native ligand ephrin-A were conducted to identify the potential blockers of the EphA2 receptor. The software program Autodock Vina 1.1.2 in PyRx-Virtual Screening Tool and BIOVIA Discovery Studio visualizer was used for this MD study. The present work showed that blocking the EphA2 receptor by these phytochemicals prevents endothelial cell apoptosis by averting ephrin-A ligand-expressing CD8+ T cell bioadhesion. These phytochemicals showed excellent docking scores and binding affinity, demonstrating hydrogen bond, electrostatic, Pi-sigma, and pi alkyl hydrophobic binding interactions when compared with native ligands at the EphA2 receptor. The comparative MD study using two PDB IDs showed that isocolumbin, carnosol, luteolin, and taraxasterol have better binding affinities (viz. −9.3, −9.0, −9.5, and −9.2 kcal/mol, respectively). Ocimum basilicum phytochemicals showed a lower docking score but more binding interactions than native ligands at the EphA2 receptor for both PDB IDs. This suggests that these phytochemicals may serve as potential drug candidates in the management of CM. We consider that the present MD study provides leads in drug development by targeting the EphA2 receptor in managing CM. The approach is innovative because a role for EphA2 receptors in CM has never been highlighted.
Article
Full-text available
Urinary tract infections (UTIs) affect a major proportion of the world population but have limited non-antibiotic-based therapeutic and preventative strategies against UTIs. Facultative intracellular uropathogens such as strains of uropathogenic E. coli, K. pneumoniae, E. faecalis, E. cloacae are well-known uropathogens causing UTIs. These pathogens manipulate several host-signaling pathways during infection, which contributes to recurrent UTIs and inappropriate antibiotic application. Since host cell receptor tyrosine kinases (RTKs) are critical for the entry, survival and replication of intracellular pathogens, we investigated whether different uropathogens require host EPHA2 receptors for their intracellular survival using a cell culture model of intracellular infection in human bladder epithelial cells (BECs). Infection of BECs with seven different uropathogens enhanced the expression levels and activation of EPHA2. The significance of EPHA2 signaling for uropathogen infection was investigated by silencing EPHA2 expression using RNA interference or by inhibiting the kinase activity of EPHA2 using small-molecule compounds such as dasatinib or ALW-II-41-27. Both preventive and therapeutic tyrosine kinase inhibition significantly reduced the intracellular bacterial load. Thus, our results demonstrate the involvement of host cell EPHA2 receptor during intracellular uropathogen infection of BECs, and targeting RTK activity is a viable non-antibiotic therapeutic strategy for managing recurrent UTIs.
Article
Full-text available
The receptor tyrosine kinase (RTK) EphA2 is expressed in epithelial and endothelial cells and controls the assembly of cell-cell junctions. EphA2 has also been implicated in many diseases, including cancer. Unlike most RTKs, which signal predominantly as dimers, EphA2 readily forms higher order oligomers upon ligand binding. Here, we investigated if a correlation exists between EphA2 signaling properties and the size of the EphA2 oligomers induced by multiple ligands, including the widely used ephrinA1-Fc ligand, the soluble monomeric m-ephrinA1, and novel engineered peptide ligands. We used Fluorescence Intensity Fluctuation (FIF) spectrometry to characterize the EphA2 oligomer populations induced by the different ligands. Interestingly, we found that different monomeric and dimeric ligands induce EphA2 oligomers with widely different size distributions. Our comparison of FIF brightness distribution parameters and EphA2 signaling parameters reveals that the efficacy of EphA2 phosphorylation on tyrosine 588, an autophosphorylation response contributing to EphA2 activation, correlates with EphA2 mean oligomer size. However, we found that other characteristics, such as the efficacy of AKT inhibition and ligand bias coefficients, appear to be independent of EphA2 oligomer size. Taken together, this work highlights the utility of FIF in RTK signaling research and demonstrates a quantitative correlation between the architecture of EphA2 signaling complexes and signaling features.
Article
Full-text available
Recent studies have shown that the Eph receptor A2 (EphA2) and its inhibitor ALW-II-41-27 could regulate various cellular processes in several types of cancer. However, the manner in which ALW-II-41-27 affects the development of cervical cancer (CC) remains unknown. The present study aimed to evaluate the role of ALW-II-41-27 in inhibiting the proliferation, invasion and migration of human papilloma virus-positive CC cells and to verify whether Ras homolog family member A (RhoA)/Rho-associated protein kinase (ROCK) may be a crucial pathway involved in this process. Reverse transcription-quantitative PCR and western blotting analyses indicated an upregulation of EphA2 expression in CC cell lines (HeLa and CaSki). Furthermore, the results from MTT and colony formation assays indicated that ALW-II-41-27 inhibited cell proliferation. Results from wound healing and Transwell assays further demonstrated the inhibitory effect of ALW-II-41-27 on CaSki and HeLa cell migration and invasion, respectively. Furthermore, ALW-II-41-27 inhibited the protein expression of GTP-RhoA and ROCK1 in CaSki and HeLa cells. In addition, the ALW-II-41-27-induced inhibition of the biological function of CaSki and HeLa cells was promoted by cell co-culture with RhoA and ROCK inhibitors. Taken together, the present findings revealed that ALW-II-41-27 inhibited CC cell proliferation, migration and invasion by blocking the RhoA/ROCK pathway. These findings provide further insight into the mechanism of CC progression and significant information for the development of potential therapeutic targets for CC.
Article
Full-text available
Recent advances in the field of host immunity against parasitic nematodes have revealed the importance of macrophages in trapping tissue migratory larvae. Protective immune mechanisms against the rodent hookworm Nippostrongylus brasiliensis (Nb) are mediated, at least in part, by IL‐4‐activated macrophages that bind and trap larvae in the lung. However, it is still not clear how host macrophages recognize the parasite. We utilized an in vitro co‐culture system of bone marrow‐derived macrophages and Nb infective larvae to screen for the possible ligand‐receptor pair involved in macrophage attack of larvae. Competitive binding assays revealed an important role for β‐glucan recognition in the process. We further identified a role for CD11b and the non‐classical pattern recognition receptor ephrin‐A2 (EphA2), but not the highly expressed β‐glucan dectin‐1 receptor, in this process of recognition. This work raises the possibility that parasitic nematodes synthesize β‐glucans and identifies CD11b and Ephrin‐A2 as important pattern recognition receptors involved in the host recognition of these evolutionary old pathogens. To our knowledge, this is the first time that EphA2 has been implicated in immune responses to a helminth.
Article
Full-text available
Background Endometriosis is a common gynecological disease characterized by the presence of endometrial tissue outside the uterus causing chronic inflammation, severe pain, and infertility. However, the innate immunity of gamma-delta (γδ) T lymphocytes in endometriosis has not been characterized. Women with endometriosis present numerous endocrine and immune dysfunctions and elevated risk for endometrial, ovarian, and breast cancers. The tyrosine kinase EphA2 is often overexpressed in cancer including endometrial carcinoma. Methods We analyzed Vδ1 and Vδ2 γδ T cells in peripheral blood and paired peritoneal fluid samples in endometriosis patients (n = 19) and compared the counts with that of age- and sex-matched healthy donors (n = 33) using flow cytometry. Vδ1 and Vδ2 T cells isolated from healthy donors were used against KLE, RL-95, and Ishikawa endometrial tumor cells in 4 h flow cytometric cytotoxicity assays. The EphA2 blocking studies were performed using antibody, small-molecule inhibitor ALW-II-41-27, and the CRISPR/Cas9. Results We determined Vδ1 T cells substantially reduced in patients’ peripheral blood (p < 0.01) and peritoneal fluid (p < 0.001). No differences were found for circulating Vδ2 T cells compared with peritoneal fluid samples. We observed inherent cytotoxic reactivity of Vδ1 and Vδ2 γδ T lymphocytes against endometrial tumor cells. Importantly, we found reduced specific lysis of EphA2-positive cell lines KLE and RL-95 by Vδ1 T cells in the EphA2 antibody blocking studies and by the EphA2 inhibitor. Furthermore, Vδ1 T-cell-mediated killing was significantly decreased in RL-95 cell EPHA2 knockout. Finally, potent cytolytic activity exerted by Vδ1 T cells was significantly reduced in EPHA2 knockouts in renal A-498 and colon HT-29 carcinoma cell lines. Conclusions We determined variable levels of Vδ1 and Vδ2 γδ T cells in endometriosis patients. We observed inherent cytotoxic reactivity of γδ T-cell subsets against endometrial cell lines. Specifically, we found that blocking of EphA2 expression resulted in significant inhibition of endometrial tumor killing mediated by Vδ1 γδ T cells. These results suggest that EphA2 is involved in tumor cell lysis and contributes to susceptibility to Vδ1 γδ T cells cytotoxic reactivity.
Article
Full-text available
During oropharyngeal candidiasis (OPC), Candida albicans invades and damages oral epithelial cells, which respond by producing proinflammatory mediators that recruit phagocytes to foci of infection. The ephrin type-A receptor 2 (EphA2) detects β-glucan and plays a central role in stimulating epithelial cells to release proinflammatory mediators during OPC. The epidermal growth factor receptor (EGFR) also interacts with C. albicans and is known to be activated by the Als3 adhesin/invasin and the candidalysin pore-forming toxin. Here, we investigated the interactions among EphA2, EGFR, Als3 and candidalysin during OPC. We found that EGFR and EphA2 constitutively associate with each other as part of a heteromeric physical complex and are mutually dependent for C. albicans-induced activation. Als3-mediated endocytosis of a C. albicans hypha leads to the formation of an endocytic vacuole where candidalysin accumulates at high concentration. Thus, Als3 potentiates targeting of candidalysin, and both Als3 and candidalysin are required for C. albicans to cause maximal damage to oral epithelial cells, sustain activation of EphA2 and EGFR, and stimulate pro-inflammatory cytokine and chemokine secretion. In the mouse model of OPC, C. albicans-induced production of CXCL1/KC and CCL20 is dependent on the presence of candidalysin and EGFR, but independent of Als3. The production of IL-1α and IL-17A also requires candidalysin but is independent of Als3 and EGFR. The production of TNFα requires Als1, Als3, and candidalysin. Collectively, these results delineate the complex interplay among host cell receptors EphA2 and EGFR and C. albicans virulence factors Als1, Als3 and candidalysin during the induction of OPC and the resulting oral inflammatory response.
Article
Pneumocystis spp. interaction with myeloid cells is well known, especially in macrophages. Contrary, how the organism binds to lung epithelial cells is incompletely understood. Ephrin type-A receptor (EphA2), has been previously identified as a lung epithelial pattern recognition receptor (PRR) that binds to fungal β-glucans. Herein, we also report that EphA2 can also bind Pneumocystis β-glucans, both in isolated forms and also on exposed surfaces of the organism. Furthermore, binding of Pneumocystis β-glucans resulted in phosphorylation of the EphA2 receptor, which has been shown to be important for downstream proinflammatory response. Indeed, we also show that IL-6 cytokine is significantly increased when lung epithelial cells are exposed to Pneumocystis β-glucans, and that this response could be blocked with preincubation with a specific antibody to EphA2. Our study presents yet another Pneumocystis lung epithelial cell receptor with implications for initial colonization and possible therapeutic intervention.
Article
More than 25 years of research and preclinical validation have defined EphA2 receptor tyrosine kinase as a promising molecular target for clinical translation in cancer treatment. Molecular, genetic, biochemical, and pharmacological targeting strategies have been extensively tested in vitro and in vivo, and drugs like dasatinib, initially designed to target SRC family kinases, have been found to also target EphA2 activity. Other small molecules, therapeutic targeting antibodies, and peptide-drug conjugates are being tested, and more recently, approaches harnessing antitumor immunity against EphA2-expressing cancer cells have emerged as a promising strategy. This review will summarize preclinical studies supporting the oncogenic role of EphA2 in breast cancer, lung cancer, glioblastoma, and melanoma, while delineating the differing roles of canonical and noncanonical EphA2 signaling in each setting. This review also summarizes completed and ongoing clinical trials, highlighting the promise and challenges of targeting EphA2 in cancer.