monocytes undergo functional reprogramming to generate

9
Research Article Monocytes Undergo Functional Reprogramming to Generate Immunosuppression through HIF-1α Signaling Pathway in the Late Phase of Sepsis Li-Li Li, Bing Dai , Yu-Han Sun, and Ting-Ting Zhang Department of Respiratory and Critical Care Medicine, The First Aliated Hospital, China Medical University, Shenyang 110001, China Correspondence should be addressed to Bing Dai; [email protected] Received 11 June 2019; Revised 8 October 2019; Accepted 20 November 2019; Published 7 February 2020 Academic Editor: Elaine Hatanaka Copyright © 2020 Li-Li Li et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Severe pneumonia with sepsis is characterized by a dysregulated inammatory response of endotoxin. In our study, we attempted to investigate the roles of the immune guardian cells (monocytes) in the immune-inammatory response of severe pneumonia- induced sepsis. We performed analysis in the blood samples of human and animals with ELISA, western blot, ow cytometry (FCM) methods, etc. Results showed that the proinammatory status shifted to hypoinammatory phases during the sepsis process. In a clinical study, the levels of IL-1β, IL-6, TNF-α, etc., except for IL-10, were inhibited in the late phase of sepsis, while, in an animal study, the immune suppression status was attenuated with administration of the adenovirus Ade-HIF-1α. Conversely, the amount of IL-10 was lower in the adenovirus Ade-HIF-1α group compared with the sepsis model group and the Ade-control group. Moreover, in the clinical study, the programmed cell death-ligand 1 (PD-L1) was overexpressed in monocytes in the late phase of sepsis, while the expression of proteins HIF-1α and STAT3 was decreased in the late phase of sepsis. However, in the animal study, we found that the HIF-1α factor facilitated the inammatory response. The expression of the proteins HIF-1α and STAT3 was increased, and the PD-L1 protein was decreased with the adenovirus Ade-HIF-1α administration compared with the rats without Ade-HIF-1α injection and with the Ade-control injection. Additionally, the proteins HIF-1α and STAT3 were coregulated at transcriptional levels during the inammatory responses of sepsis. Taken together, monocytes undergo reprogramming to generate immunosuppression through the HIF-1α signaling pathway in the late phase of sepsis. 1. Introduction Severe pneumonia with sepsis caused the highest mortality in intensive care units worldwide due to endogenous endotoxin. Data revealed that there are nearly 5.3 million deaths from severe pneumonia-induced sepsis every year [1]. Sepsis contained two stages: hyperinammatory and hypoinam- matory phases. During the hyperinammatory stage, the immune cells are triggered, such as the immune guardian cells: monocytes and neutrophils, which in turn release abun- dant inammatory cytokines (IL-1β, IL-6, and TNF-α), che- mokines (CXCL1), myeloperoxidases, and proteases to induce subsequent adaptive immune responses and induce the activated neutrophil degranulation [2, 3]. In the case of the hypoinammatory phase, the immune system is dysregu- lated likely owing to the impairment of the phagocytic capac- ity of monocytes and monocyte-derived macrophages [4]. Studies showed that the expression of major histocompatibil- ity class II antigens and the complement receptor-1 (CR1 and CD35) was reduced [5, 6] in monocytes, the production of inammatory cytokines TNF-α and IL-12 was decreased and the release of anti-inammatory cytokines IL-10 and PGE2 was enhanced, etc. [7, 8], all of which induced the pro- liferation and function of T cells and natural killer (NK) cells in the adaptive response [9]. However, some researchers had identied that the predominant immunosuppressed charac- teristic was that the monocytes cocommunicate with T cells in the sepsis process [10]. The monocytes, a kind of antigen- Hindawi Mediators of Inflammation Volume 2020, Article ID 4235909, 9 pages https://doi.org/10.1155/2020/4235909

Upload: others

Post on 20-Apr-2022

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Monocytes Undergo Functional Reprogramming to Generate

Research ArticleMonocytes Undergo Functional Reprogramming to GenerateImmunosuppression through HIF-1α Signaling Pathway in theLate Phase of Sepsis

Li-Li Li, Bing Dai , Yu-Han Sun, and Ting-Ting Zhang

Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, China Medical University,Shenyang 110001, China

Correspondence should be addressed to Bing Dai; [email protected]

Received 11 June 2019; Revised 8 October 2019; Accepted 20 November 2019; Published 7 February 2020

Academic Editor: Elaine Hatanaka

Copyright © 2020 Li-Li Li et al. This is an open access article distributed under the Creative Commons Attribution License, whichpermits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Severe pneumonia with sepsis is characterized by a dysregulated inflammatory response of endotoxin. In our study, we attempted toinvestigate the roles of the immune guardian cells (monocytes) in the immune-inflammatory response of severe pneumonia-induced sepsis. We performed analysis in the blood samples of human and animals with ELISA, western blot, flow cytometry(FCM) methods, etc. Results showed that the proinflammatory status shifted to hypoinflammatory phases during the sepsisprocess. In a clinical study, the levels of IL-1β, IL-6, TNF-α, etc., except for IL-10, were inhibited in the late phase of sepsis,while, in an animal study, the immune suppression status was attenuated with administration of the adenovirus Ade-HIF-1α.Conversely, the amount of IL-10 was lower in the adenovirus Ade-HIF-1α group compared with the sepsis model group and theAde-control group. Moreover, in the clinical study, the programmed cell death-ligand 1 (PD-L1) was overexpressed inmonocytes in the late phase of sepsis, while the expression of proteins HIF-1α and STAT3 was decreased in the late phase ofsepsis. However, in the animal study, we found that the HIF-1α factor facilitated the inflammatory response. The expression ofthe proteins HIF-1α and STAT3 was increased, and the PD-L1 protein was decreased with the adenovirus Ade-HIF-1αadministration compared with the rats without Ade-HIF-1α injection and with the Ade-control injection. Additionally, theproteins HIF-1α and STAT3 were coregulated at transcriptional levels during the inflammatory responses of sepsis. Takentogether, monocytes undergo reprogramming to generate immunosuppression through the HIF-1α signaling pathway in the latephase of sepsis.

1. Introduction

Severe pneumonia with sepsis caused the highest mortality inintensive care units worldwide due to endogenous endotoxin.Data revealed that there are nearly 5.3 million deaths fromsevere pneumonia-induced sepsis every year [1]. Sepsiscontained two stages: hyperinflammatory and hypoinflam-matory phases. During the hyperinflammatory stage, theimmune cells are triggered, such as the immune guardiancells: monocytes and neutrophils, which in turn release abun-dant inflammatory cytokines (IL-1β, IL-6, and TNF-α), che-mokines (CXCL1), myeloperoxidases, and proteases toinduce subsequent adaptive immune responses and inducethe activated neutrophil degranulation [2, 3]. In the case of

the hypoinflammatory phase, the immune system is dysregu-lated likely owing to the impairment of the phagocytic capac-ity of monocytes and monocyte-derived macrophages [4].Studies showed that the expression of major histocompatibil-ity class II antigens and the complement receptor-1 (CR1 andCD35) was reduced [5, 6] in monocytes, the production ofinflammatory cytokines TNF-α and IL-12 was decreasedand the release of anti-inflammatory cytokines IL-10 andPGE2 was enhanced, etc. [7, 8], all of which induced the pro-liferation and function of T cells and natural killer (NK) cellsin the adaptive response [9]. However, some researchers hadidentified that the predominant immunosuppressed charac-teristic was that the monocytes cocommunicate with T cellsin the sepsis process [10]. The monocytes, a kind of antigen-

HindawiMediators of InflammationVolume 2020, Article ID 4235909, 9 pageshttps://doi.org/10.1155/2020/4235909

Page 2: Monocytes Undergo Functional Reprogramming to Generate

presenting cells (APC), acted as instigators of T cell suppres-sion in adaptive response by mediating the expression ofinhibitory coreceptors such as programmed cell death pro-tein 1 (PD-1) and cytotoxic T lymphocyte-associated protein4 (CTLA4) [10, 11]. The researches of Avendano-Ortiz et al.[12] and Shalova et al. [13] showed that hypoxia-induciblefactor-1α (HIF-1α) regulated functional reprogramming ofmonocytes in sepsis to suppress T cells with inhibitory cor-eceptors, cytokines, and chemokines. It was also shown byTsukamoto et al. [14] that the myeloid-derived suppressorcells (MDSCs) were more suppressive in nature by upregu-lating the expression of PD-L1 to impair antigen-specific Tcell priming and IgG production in sepsis. Hence, based onbioinformatic analysis of the GSE46955 data [13], we con-ducted this study to investigate the inflammatory responsecharacteristics of the functional disability in monocytesthrough the transcription activator hypoxia-inducible fac-tor-1α (HIF-1α) in mediating the protein signal transducerand activator of transcription 3 (STAT3) during the severepneumonia with sepsis.

2. Materials and Methods

2.1. Data Collection. Data extraction of GSE46955 wasobtained from the Gene Expression Omnibus (GEO;http://www.ncbi.nlm.nih.gov/geo/) database. From the data,peripheral blood samples were collected from gram-negativesepsis patients during sepsis (sepsis) and following their recov-ery (recovery) status as well as the healthy donor (control).The platform used in the GSE46955 data was the GPL6104Illumina HumanRef-8 v2.0 expression beadchip.

2.2. Human Blood Samples. Peripheral blood samples of 30patients diagnosed with severe pneumonia with sepsis werecollected in the early phases and late phase. Then, the sam-ples in each phase were equally divided into two groups. 30blood samples of the healthy participants for the controlgroup were also collected. Besides, the diagnostic criteria ofearly phase sepsis were patients with symptoms of chills, highfever, poor appetite, fatigue, limb joint soreness, etc., whilethe patients showed symptoms of respiratory failure, delir-ium, etc., in the late sepsis phase. Additionally, permissionto carry out this study was obtained from the Ethical Com-mittee of our Hospital, and informed consent was obtainedfrom all the patients participating in the study.

2.3. Animal Sepsis Model. Eighty SD rats aged 8-10 weekswere purchased from the SiBeiFu Laboratory (Animal Tech-nology Company, China) and equally divided into fourgroups. The rats in one group were used as the control andthe rats in the other three groups were infected with S. pneu-moniae for more than fifteen days until they had been diag-nosed to have pneumonia-induced sepsis. The diagnosticcriteria of early sepsis were rats with chills and shortness ofbreath, and the rats showed coma and were slightly breathlessin the late phase of sepsis. Besides, one group of rats from thethree experimental groups in the late phase of sepsis were tailvein-injected with the adenovirus Ade-HIF-1α (175μL, at aconcentration of 1:5 × 106 PFU) for 5 days of treatment; also,

the other sepsis model rats were injected with the sameamount of adenovirus control plasmid for 5 days (GemmaBiotechnology Co., Ltd., Shanghai). Thereafter, collection ofblood was done from the eyeball and tail vein in the SD ratsin the late phase of sepsis and normal control group. Six lungtissues from each group were also obtained.

2.4. ELISA Assay. The blood samples obtained both fromthe patients and the animals were centrifugated at 1500 gfor 10min, two times. Then, the serum of the blood sampleswas dividedly analyzed with ELISA assay kits as per themanufacturer’s protocol to quantify the concentration ofIL-1β (catalog:70-EK101B-96, homo; catalog:70-EK201B/3-96, mus; MultiSciences, China), IL-18 (catalog: 70-EK118-48, homo; catalog: 70-EK218-96, mus; MultiSciences, China),IL-6 (catalog: 70-EK106/2-96, homo; catalog: 70-EK206/3-96,mus; MultiSciences, China), IL-10 (catalog: 70-EK110/2-96,homo; catalog: 70-EK210/3-96, mus; MultiSciences, China),TNF-α (catalog: 70-EK182-96, homo; catalog: 70-EK282/3-96, mus; MultiSciences, China), CCL3 (catalog: 70-EK161-96, homo; catalog: 70-EK261/2-96, mus; MultiSciences,China), and CCL5 (catalog: 70-EK1129-96, homo; catalog:70-EK2129/2-96, mus; MultiSciences, China) in triplicates.

2.5. Western Blot. Lymphocytes were obtained from theblood samples of the patients and the animals with aFicoll-Hypaque Solution of humans (catalog: p8900, Solar-bio, China) and rats (catalog: P8620, Solarbio, China), thenthe monocytes were isolated from the lymphocytes withthe Dynabeads® FlowComp™ Human CD14 kit (catalog:11367D, Invitrogen, supplementary file (available here)).Later, they were lysed in 1mL of RIPA buffer (Beyotime)for a duration of 20 minutes on ice; besides, the PMSF, a pro-tease inhibitor cocktail, was also mixed in the RIPA buffer.Then, the extracted protein was quantified with a BCA kit(Pierce, Rockford, IL). Afterwards, separation of proteinswas carried out with sodium dodecyl sulfate polyacrylamidegel electrophoresis (SDS-PAGE), then the protein gel wastransferred to a PVDF membrane (Bio-Rad, Hercules, CA).After that, the protein membranes were blocked with 5%nonfat milk for 1 hour, then the protein membranes under-went incubation overnight at 4°C with primary antibodySTAT3 (1 : 500, catalog: 4904S, CST), HIF-1α (1 : 500, catalog:36169S, CST), PD-L1 (1 : 500, catalog: 13684S-homo/29122S-mus, CST), and GAPDH (1 : 1000, catalog: 5174S, CST).Thereafter, the membranes were washed for 3 times inTBST/1% Tween-20, then they were incubated with rabbitpolyclonal antibody at 4°C for 60min. Besides, GAPDHwas used as a control. Finally, an ECL detection instrument(Thermo Fisher Scientific) plus a chemiluminescent sub-strate were added to visualize the immunoreactive bandswith Bio-RAP.

2.6. Flow Cytometry (FCM). The monocytes were obtainedfrom peripheral blood samples of the patients with theFicoll-Hypaque Solution of human (catalog: p8900, Solarbio,China) and the Dynabeads® FlowComp™ Human CD14 kit(catalog: 11367D, Invitrogen); the details of the methodwere listed in the supplementary file. Then, the obtained

2 Mediators of Inflammation

Page 3: Monocytes Undergo Functional Reprogramming to Generate

monocytes were placed in RPMI-1640 medium with FBS(Thermo Fisher Scientific, USA). Afterwards, the expressionof CD274 was analyzed by FCM. The brief methods arelisted: the monocytes were fixed and labeled with FITC-conjugated anti-human CD274 antibody (catalog: MA5-

16848, Invitrogen), anti-mouse CD274 antibody (catalog:558065, BD Pharmingen™), and allophycocyanin- (APC-)conjugated anti-human CD14 antibody (catalog: 17-0149-42, Invitrogen). Thereafter, the cells were washed twice andresuspended in PBS for flow cytometry analysis. Finally, the

IL-1𝛽 IL-18 IL-6 IL-10 TNF-𝛼0

50100400800

12002000400060008000

10000

####

####

NCSepsis-lateSepsis-early

## ##

##

CCL3 CCL5

Cyto

kine

leve

ls (p

g/m

L)

⁎⁎⁎⁎

⁎⁎

⁎⁎

⁎⁎⁎⁎

⁎⁎

(a) In clinical

IL-18 IL-6 IL-10 TNF-𝛼 CCL3 CCL50

300

600

900

#

&

&

Cyto

kine

leve

ls (p

g/m

L)

IL-1𝛽0

NCAde-controlSepsisov-HIF-1𝛼

⁎⁎

⁎⁎

⁎⁎⁎⁎

⁎⁎

⁎⁎

⁎⁎⁎⁎⁎⁎⁎⁎⁎

⁎⁎

⁎⁎⁎⁎

⁎⁎

⁎⁎

⁎⁎

⁎⁎⁎⁎

(b) In animal study

Figure 2: Its cytokine production in the blood serum. (a) In the clinical study, the higher levels of TNF-α, IL-1β, IL-6, IL-18, CCL3, and CCL5except for IL-10 in the early phase of sepsis compared with the late phase of sepsis and the healthy participants, n = 30. ∗∗P < 0:01 representedthe significant difference; ##P < 0:01: compared with the healthy participants, the difference was of significance. (b) Ade-HIF-1α injectionincreased the production of TNF-α, IL-1β, IL-6, IL-18, CCL3, and CCL5 except for IL-10 in the late stage of the sepsis rat model. ∗∗P <0:01: compared with the normal control group, the difference was of significance; #P < 0:05: compared with the sepsis rat model groupand the adenovirus control group; &P < 0:05: compared with the sepsis rat model group. Besides, data were presented as mean ± standarddeviation, and data was repeated in triplicate.

Pathwayhsa05152: tuberculosishsa04060: cytokine-cytokine receptor interactionhsa05323: rheumatoid arthritishsa04668: TNF signaling pathwayhsa04064: NF-kappa B signaling pathwayhsa05132: salmonella infectionhsa05418: fluid shear stress and atherosclerosisM159: PID AMB2 neutrophils pathwayM159: PID TNF pathwayhsa04630: Jak-STAT signaling pathwayM264: PID toll endogenous pathwayM255: PID HIF1 TF pathwayM165: PID syndecan 4 pathwayM145: PID P53 downstream pathwayM92: PID angiopoietin receptor pathwayM15743: ST GA12 pathwayhsa05202: transciptional misregulation in cancerhsa04370: VEGF signaling pathwayhsa04216: ferroptosishsa00062: fatty acid elongationM252: PID IL8 CXCR1 pathwayM5880: NABA ECM affiliatedM167: PID AP1 pathwayhsa04142: lysosomehsa00640: propanoate metabolismM256: PID TAP63 pathwayhsa04610: complement and coagulation cascadeshsa00770: panthotehnate and CoA biosynthesisM254: PID MYC repress pathwayhsa00900: terpenoid backbone biosynthesis

0 2 4 6 8 10–log10 (P)

12 14 16

10–2

10–3

10–4

10–5

10–10

10–20

(a)

Biological processGO:0002237: response to molecule of bacterial originGO:0031349: positive regulation of defense responseGO:0002366: leukocyte activation involved in immune responseGO:0022407: regulation of cell-cell adhesionGO:0050900: leukocyte migrationGO:0009636: response to toxic substanceGO:0032729: positive regulation of interferon-gamma productionGO:0002699: positive regulation of immune effector processGO:0031663: lipopolysaccharide-mediated signaling pathwayGO:0030335: positive regulation of cell migrationGO:0002285: lymphocyte activation involded in immune responseGO:0071887: leukocyte apoptotic processGO:0032103: positive regulation of response to external stimulus GO:0070555: response to interleukin-1GO:0043065: positive regulation of apoptotic processGO:0001776: leukocyte homeostasisGO:0071706: tumor necrosis factor superfamiuly cytokine productionGO:0097190: apoptotic signaling pathwayGO:0043900: regulation of multi-organism processGO:0042060: wound healing

–log10 (P)20151050 25

10–2

10–3

10–4

10–5

10–10

10–20

(b)

Cellular componentGO:0005764: lysosomeGO:0030667: secretory granule membraneGO:0045121: membrane raftGO:0098552: side of membraneGO:0070820: tertiary granuleGO:0042629: mast cell granuleGO:0005770: late endosomeGO:0042613: MHC class II protein complexGO:0031092: platelet alpha granule membraneGO:0005925: focal adhesionGO:0016324: apical plasma membraneGO:0000139: Golgi membraneGO:0031228: intrinsic component of Golgi membraneGO:0001891: phagocytic cupGO:0044291: cell-cell contact zoneGO:0098862: cluster of actin-based cell projectionsGO:0071437: invadopodiumGO:0005938: cell cortexGO:0005788: endoplasmic reticulum lumenGO:0005740: mitochondrial envelopeGO:0098636: protein complex involved in cell adhesionGO:0005811: lipid dropletGO:0030027: lamellipodiumGO:0005769: early endosomeGO:0005884: actin filamentGO:0031225: anchored component of membraneGO:0035580: specific granule lumen

0 2 4–log10 (P)

6 8 10

10–2

10–3

10–4

10–5

10–10

10–20

(c)

Molecular functionGO:0005125: cytokine activityGO:0046982: protein heterodimerization activityGO:0031726: CCR1 chemokine receptor bindingGO:0019955: cytokine bindingGO:0008289: lipid bindingGO:0099516: ion antiporter activityGO:0051019: mitogen-activated protein kinase bindingGO:0015037: peptide disulfide oxidoreductase activityGO:0023026: MHC class II protein complex bindingGO:0016773: phosphotransferase activity, alcohol group as acceptorGO:0019900: kinase bindingGO:0050839: cell adhesion molecule bindingGO:0030674: protein binding, bridgingGO:0038187: pattern recognition receptor activityGO:0008429: phosphatidylethanolamine bindingGO:0090482: vitamin transmembrane transporter activityGO:0035035: histone acetyltransferase bindingGO:0019904: protein domain specific bindingGO:0051015: actin filament bindingGO:0000977: RNA polymerase II regulatory region sequence-specific DNA bindingGO:0031593: polyubiquitin modification-dependent protein binding GO:0050700: CARD domain bindingGO:0032182: ubiquitin-like protein bindingGO:0046915: transitioin metal ion transmembrane tranporter activityGO:0070851: growth factor receptor bindingGO:0005402: carbohydrate:cation symporter activityGO:0051219: phosphoprotein bindingGO:0043531: ADP bindingGO:0004745: retinol dehydrogenase activityGO:0030246: carbohydrate binding

0 2 4 6–log10 (P)

10–2

10–3

10–4

10–5

10–10

10–20

12108 14 16

(d)

Figure 1: Gene Set Enrichment Analysis of the dataset GSE46955 from the GEO database with Geo2R and DAVID software tools. KEGG andreactome pathway analysis of the major signaling pathways (a). Gene ontology analysis of the differentially expressed genes, the majorbiological process (b), the major cellular component (c), and the major molecular function (d).

3Mediators of Inflammation

Page 4: Monocytes Undergo Functional Reprogramming to Generate

expression of PD-L1 on the monocytes was analyzed with theFACSCalibur (Bioscience, BD, USA) instrument.

2.7. Immunofluorescence (IF). Monocytes were obtainedfrom the patients’ blood samples in the primary sepsis stage,late sepsis stage, and healthy control. Then, the cells werefixed with 4% polyformaldehyde for 30min and perme-abilized with 0.1% TritonX-100 for 3min; afterwards, theywere blocked with 10% BSA for 60min at room temperature.Finally, the cells were incubated overnight with PD-L1 (cat-alog:13684S, CST) and HIF-1α (catalog: 36169S, CST) at

4°C. Besides, the nucleus DNA was stained with DAPI for5min (catalog: D9542, Sigma, USA). The images were thenvisualized under a confocal microscope (Leica scanningmicroscope, Germany).

2.8. Coimmunoprecipitation (Co-IP) Assay. The proteinswere extracted from the collected blood samples of thepatients with a protein extraction RIPA buffer with PMSF(Beyotime, China). The protein A/G Sepharose (Santa CruzBiotechnology) was preincubated with anti-HIF-1α (catalog:36169S, CST) or anti-STAT3 (catalog: 4904S, CST) antibody

NC

Seps

is-la

te

Seps

is-ea

rl y

HIF-1𝛼

STAT3

GAPDH

PD-L1

(a)

PD-L1 HIF-1𝛼 STAT30

1

2

3

4

Rela

tive p

rote

in le

vels

(fold

of c

ontr

ol)

#

##

NCSepsis-lateSepsis-early

(b)

NC

Ade

-con

trol

Seps

is

ov-H

IF-1𝛼

HIF-1𝛼

STAT3

GAPDH

PD-L1

(c)

0.0

0.5

1.0

1.5

Rela

tive p

rote

in le

vels

(fold

of c

ontr

ol)

###

##

#

PD-L1 HIF-1𝛼 STAT3

NCAde-controlSepsisov-HIF-1𝛼

⁎⁎⁎

⁎⁎

(d)

Figure 3: Its expression of proteins in the monocytes. (a) The protein bands of HIF-1α, STAT3, and PD-L1 in the clinical study. (b) Thedownregulated expression of proteins HIF-1α and STAT3 in the late phase of sepsis compared with the primary phase of sepsis; #P < 0:05represented significant difference. While the upregulation of the immune checkpoint protein PD-L1 expressed on the monocytes in thelate phase of sepsis, #P < 0:05 represented significant difference vs. the primary phase of sepsis. ∗P < 0:05 of proteins HIF-1α and STAT3in the primary phase of sepsis versus the normal control group. (c) The protein bands of HIF-1α, STAT3, and PD-L1 in the sepsis ratmodel study. (d) Upregulated expression of proteins HIF-1α and STAT3 and downregulated protein PD-L1 expression in the sepsis ratmodel with Ade-HIF-1α injection; besides the highly expressed protein PD-L1 in the sepsis rats model, ##P < 0:05 of PD-L1 and HIF-1αversus the normal control group and ov-HIF-1α adenovirus-injected group, ∗P < 0:05 of PD-L1 versus the normal control group; ∗∗P <0:05 of HIF-1α and STAT3 versus the normal control group, sepsis rat model group, and adenovirus control group; #P < 0:05 of STAT3versus the normal control group and ov-HIF-1α adenovirus-injected group; data were presented as mean ± standard deviation, andanalysis was replicated in triplicate.

4 Mediators of Inflammation

Page 5: Monocytes Undergo Functional Reprogramming to Generate

for 60min at 4°C and IgG antibody as a control. Then, wesuspended the bead-antibody complexes with protein lysateand washed the beads with an extraction buffer 3 times.The samples were centrifuged at 3000 g to obtain the immu-neprecipitates. Then, the immuneprecipitates were analyzedwith the western blot.

2.9. Statistical Analysis. The results were analyzed in the one-way ANOVAmethod with SPSS 19.0 statistical software. Theresults were considered statistically significant at P < 0:05.The differences presented in graphs were analyzed withGraphPad Prism 6.0 software.

3. Results

3.1. Bioinformatic Analysis of the Data GSE46955. Gene SetEnrichment Analysis of the mRNA expression profiles wascarried out for the differentially expressed genes (P < 0:05)in monocytes of the gram-negative bacteria-induced sepsispatients from the GEO database (GSE46955). Data showedthat the major signaling pathways were the interaction ofthe inflammatory cytokines, TNF signaling pathway, JAK-STAT signaling pathway, etc. (Figure 1(a)), while the GeneOntology of the biological processes was mainly a responseto bacterial, positive regulation of the defense response, leu-kocyte activation in immune response, etc. (Figure 1(b));the cellular components were mainly involved in the cell orgranule membrane, etc. (Figure 1(c)); besides, the molecularfunctions were mainly about cytokine or chemokine activity,etc. (Figure 1(d)).

3.2. Immunosuppression of Monocytes via HIF-1α SignalingPathway. The analysis of inflammatory cytokine generationfrom blood samples of patients and the normal controlshowed that during the early phases of sepsis, there was aproinflammatory status with higher levels of TNF-α, IL-1β,IL-6, IL-18, CCL3, and CCL5, compared with the late phaseof sepsis except for the production of IL-10 (Figure 2(a)),while, in the animal study, the administration of adenovirusAde-HIF-1α alleviated the immune suppression status inthe late stage of sepsis and results showed that TNF-α, IL-1β, IL-6, IL-18, CCL3, and CCL5 levels were higher in theAde-HIF-1α group than in the Ade-control group, the sepsismodel group without adenovirus injection, and the normalcontrol group (Figure 2(b)), while the amount of IL-10 wasincreased more obviously in the sepsis rat model group com-pared with the Ade-control group (Figure 2(b)). Addition-ally, in the clinical study, the western blot analysis of theproteins HIF-1α and STAT3 showed that the expression ofprotein STAT3 was significantly reduced in the late phaseof sepsis compared with the primary phase (Figures 3(a)and 3(b)). Meanwhile, in the animal study, data presentedthat administration of Ade-HIF-1α resulted in an increasedexpression of HIF-1α and STAT3 compared with the sepsisrat model group and the Ade-control group (Figures 3(c)and 3(d)). Conversely, in the clinical study, the expressionof protein PD-L1 was upregulated in the late phase of sepsis,while, in the animals’ study, the protein PD-L1 expressionwas higher in the sepsis rat model group compared with the

Ade-control group and the Ade-HIF-1α group (Figure 3).Interestingly, with the coimmunoprecipitation assay, resultsshowed that proteins HIF-1α and STAT3 coregulated at thetranscriptional level in the immune process of sepsis(Figures 4(a) and 4(b)). Therefore, monocytes undergoimmunosuppression in the sepsis late phase through theHIF-1α signaling pathway.

3.3. Overexpression of Immune Checkpoint Protein PD-L1.As we had found that the monocytes undergo immunosup-pression through the HIF-1α signaling pathway (Figure 3),the further study of FCM analysis identified that PD-L1expressed on the monocytes was higher in the late phase ofsepsis compared with the early phase (Figure 5). Addition-ally, the immunofluorescence results displayed that the pro-tein HIF-1α expressed on the nuclear and PD-L1 expressedon the monocyte membrane (Figure 6). Therefore, mono-cytes undergo immunosuppression in the late phase of sep-sis through the HIF-1α signaling pathway, which inducedthe upregulation of protein PD-L1 expressed on the mono-cytes (Figure 7).

4. Discussion

Based on this study, we further confirmed that sepsis was anuncontrolled inflammatory response syndrome. During thesepsis process progression, it shifted to the immunosuppres-sion status. Results showed that the primary phase of sepsiswas associated with hyperinflammation with higher amountsof proinflammatory cytokine secretion. Conversely, therewas an increased production of anti-inflammatory cytokineIL-10 in the late phase of sepsis. This conclusion is in consis-tent with the finding of Shalova et al. [13]. Moreover, we alsofound that the HIF-1α was activated in hypoxic condi-tions during the primary phase in sepsis, which then coacti-vated the signal transducer and activator of transcription 3(STAT3) to engage in the inflammatory process. HIF-1protein is a heterodimer consisting of two subunits: HIF-a

HIF-1𝛼120 kD110 kD100 kD90 kD80 kD70 kD

STAT3

NC

Seps

is ea

rly

Input

IgG

Co-IP

Seps

is la

te

NC

NC

Seps

is ea

rly

Seps

is la

te

120 kD110 kD100 kD90 kD

80 kD

70 kD

HIF-1𝛼

STAT3

(a)

(b)

Figure 4: Coimmunoprecipitation assay of the proteins HIF-1α andSTAT3. (a, b) In clinical blood samples, the proteins HIF-1α andSTAT3 were coregulated at transcriptional levels.

5Mediators of Inflammation

Page 6: Monocytes Undergo Functional Reprogramming to Generate

Early sepsis

0

FSC-

H

FITC-CD2740 102 103 104 105

Late sepsis

(a)

Cell

coun

ts

1500

1000

500

Sepsis-early Sepsis-late0

(b)

Figure 5: (a) Flow cytometry analyzed the expression of PD-L1 in the blood samples of sepsis patients (n = 30); the dark line represented theearly phase of sepsis, while the blue line represented the late phase of sepsis. (b) ∗P < 0:05 stands for the significant difference.

PD-L1

NC

Sepsis-early

Sepsis-late

DAPI MergeHIF-𝛼

(a)

PD-L1

#

200

150

100

50

0

Cel

l mea

nflu

ores

cenc

e int

ensit

y (M

FI)

HIF-𝛼

NCSepsis-lateSepsis-early

⁎⁎

⁎⁎

(b)

Figure 6: Its immunofluorescence analysis of proteins HIF-1α and PD-L1 (200x) in the clinical study. (a) The protein HIF-1α (red) is locatedin the nuclear, the immune checkpoint protein PD-L1 (green) is expressed on the monocyte membrane, and DAPI was used to stain thenucleus (blue). (b) Reducing the mean fluorescence intensity (MFI) of protein HIF-1α in the late phase of sepsis, ∗∗P < 0:05 of HIF-1α andPD-L1 versus the normal control group represented significant difference, while increasing the fluorescence intensity (MFI) of protein PD-L1 in the late phase of sepsis versus the early phase, #P < 0:05 of PD-L1 versus normal control group represented significant difference.

6 Mediators of Inflammation

Page 7: Monocytes Undergo Functional Reprogramming to Generate

and HIF-1b. Under hypoxia condition, the nonhydroxylatedHIF-1α is not degraded. Then, the unmodified proteindimerizes with HIF-1b and p300 (or CBP) binding to HIF-1α to activate the transcription of HIF-1 target genes in itsdownstream [15, 16]. In a tumor study, researchers Niuet al. showed that STAT3 activation was proposed to beupstream of HIF-1α and regulated HIF-1α transcription inits pathway [17]. Hence, in sepsis, STAT3 also mediatedHIF-1α transcriptional activity in its signaling pathway.Furthermore, in our clinical study, results also showed thatthe proinflammatory response was suppressed in the latephase of sepsis. Data revealed it not only inhibiting the pro-inflammatory cytokine/chemokine production, but alsoupregulating the expression of protein PD-L1. However, inthe late stage sepsis rat model study, results identified thathypoxia-inducible factor-1α adenovirus injection alleviatedthe immunosuppression status along with higher secretion ofproinflammatory cytokines/chemokines and lower expressionof protein PD-L1. Hence, the immune checkpoint proteinPD-L1 expressed on the monocyte membrane also partici-pated in the sepsis immunosuppression response. Dissatis-fied, in our study, we did not identify which gene or proteindirectly regulated the expression of PD-L1 in the HIF-1αdownstream signaling pathway.

Immune checkpoints (ICs) played pivotal roles inimmune surveillance in cancer [18]. They were ligands oflymphocyte receptors engaging in modulating the duration

and initiation of adaptive immune response [19]. Therefore,it is possible that the induction of immune tolerance insepsis inhibited the T cell effector response through PD-L1in monocytes, which was in line with the work of Changet al. [20] and Zhang et al. [21]. It was declared that PD-L1was a sepsis phenotype playing roles in the communicationamong monocytes and T lymphocytes. Therefore, furtherstudies are required to reveal the mystery of PD-L1 in thecommunication among monocytes and T lymphocytes. Inaddition, a study by Deng et al. [22] claimed that the corecircadian clock gene, BMAL1, upregulates the pyruvatekinase M2 (PKM2) to prevent the development of sepsisby coregulating programmed cell death-ligand 1 (PD-L1) inmacrophages. Hence, this is also a new approach for us todirect our further study.

The inflammatory response through the HIF-1α signal-ing pathway is mainly due to IL-6/STAT3 axis and NF-κB.Studies indicated that the axis NF-κB2/p100 [23] or thephosphorylated NF-κB [24, 25] and the NF-κBp65/RelB het-erodimers [26] played pivotal roles in endotoxin toleranceduring sepsis in human monocytes. Furthermore, the studyof Qin et al., clearly declared that SIRT5 competed withSIRT2 to interact with NF-κBp65 to increase acetylation ofp65, which promoted the production of cytokines againstthe progression of sepsis [27]. Hence, it is possible that theprotein NF-κB also engages in the sepsis immune responsein the HIF-1α signaling pathway, which also awaits further

LPS IL-6

IL-6RTLR4

IFN-rR

IFN-r

MonocytesT cells Cytokines

Inflammatory cytokines

Anaerobicmetabolism

PD-L1

Hypoxia TMP1

Bcl-2

STAT3

STAT3

HIF-1𝛼IκBsNFκB

NFκB

IκBs degradation

PATPCytoCtBid

Bid

P

Monocyte cell membrane

Inflammatory cytokines

Figure 7: The inflammatory response through the HIF-1α signaling pathway. The inflammatory cytokine IL-6 and other inflammatorysignals bind with the receptors expressed on the immune cells (monocytes, etc.) to activate the downstream signals. In our study, theproteins HIF-1α and STAT3 were activated to induce inflammatory response in the early phase of sepsis. While, in the late phase ofsepsis, the immune checkpoint protein PD-L1 expressed on the monocyte membrane was upregulated to generate immunosuppression.

7Mediators of Inflammation

Page 8: Monocytes Undergo Functional Reprogramming to Generate

study. Taken together, based on our study, the immunecheckpoint protein PD-L1 overexpressed in monocytes dur-ing the late phase of sepsis contributed to immunosuppres-sion in severe pneumonia-induced sepsis through the HIF-1α signaling pathway.

5. Conclusion

Monocytes undergo immunosuppression in the late phase ofsepsis through the HIF-1α signaling pathway, which thusinhibited the production of proinflammatory cytokines andinduced the upregulation of protein PD-L1 expressed onthe monocytes.

Data Availability

The data used to support the findings of this study are allincluded within the article.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors’ Contributions

Li LL and Dai B designed the experiments, Sun YH carriedout the experiments, and Zhang TT analyzed the experimen-tal results. Li LL wrote the manuscript and Dai B approvedthe manuscript.

Supplementary Materials

The protocols of isolating monocytes. (SupplementaryMaterials)

References

[1] D. B. Danahy, S. M. Anthony, I. J. Jensen et al., “Polymicrobialsepsis impairs bystander recruitment of effector cells toinfected skin despite optimal sensing and alarming functionof skin resident memory CD8 T cells,” PLoS Pathogens,vol. 13, no. 9, article e1006569, 2017.

[2] M. Takehara, S. Seike, Y. Sonobe et al., “Clostridium perfrin-gens α-toxin impairs granulocyte colony-stimulating factorreceptor-mediated granulocyte production while triggeringseptic shock,” Communications Biology, vol. 2, p. 45, 2019.

[3] L. Jin, S. Batra, and S. Jeyaseelan, “Diminished neutrophilextracellular trap (NET) formation is a novel innate immunedeficiency induced by acute ethanol exposure in polymicrobialsepsis, which can be rescued by CXCL1,” PLoS Pathogens,vol. 13, no. 9, article e1006637, 2017.

[4] H. Minasyan, “Sepsis and septic shock: pathogenesis and treat-ment perspectives,” Journal of Critical Care, vol. 40, pp. 229–242, 2017.

[5] C. del Fresno, F. García-Rio, V. Gómez-Piña et al., “Potentphagocytic activity with impaired antigen presentation identi-fying lipopolysaccharide-tolerant human monocytes: demon-stration in isolated monocytes from cystic fibrosis patients,”Journal of Immunology, vol. 182, no. 10, pp. 6494–6507, 2009.

[6] H. Amdahl, K. Haapasalo, L. Tan et al., “Staphylococcal pro-tein Ecb impairs complement receptor-1 mediated recognition

of opsonized bacteria,” PLoS One, vol. 12, no. 3, articlee0172675, 2017.

[7] M. Nalos, S. Huang, R. Sluyter et al., “"Host tissue damage"signal ATP impairs IL-12 and IFNγ secretion in LPS stimu-lated whole human blood,” Intensive Care Medicine, vol. 34,no. 10, pp. 1891–1897, 2008.

[8] P. Revathikumar, J. Estelius, U. Karmakar et al., “Correction:Microsomal prostaglandin E synthase-1 gene deletion impairsneuro-immune circuitry of the cholinergic anti-inflammatorypathway in endotoxaemic mouse spleen,” PLoS One, vol. 13,no. 4, p. e0196806, 2018.

[9] S. Hiraki, S. Ono, M. Kinoshita et al., “Neutralization of IL-10restores the downregulation of IL-18 receptor on natural killercells and Interferon-γ production in septic Mice, thus leadingto an improved survival,” Shock, vol. 37, no. 2, pp. 177–182,2012.

[10] I. Jensen, F. V. Sjaastad, T. S. Griffith, and V. P. Badovinac,“Sepsis-induced T cell immunoparalysis: the ins and outs ofimpaired T cell immunity,” Journal of Immunology, vol. 200,no. 5, pp. 1543–1553, 2018.

[11] E. A. Fallon, B. M. Biron-Girard, C.-S. Chung et al., “A novelrole for coinhibitory receptors/checkpoint proteins in theimmunopathology of sepsis,” Journal of Leukocyte Biology,vol. 103, no. 6, pp. 1151–1164, 2018.

[12] J. Avendaño-Ortiz, C. Maroun-Eid, A. Martín-Quirós et al.,“PD-L1 overexpression during endotoxin tolerance impairsthe adaptive immune response in septic patients via HIF1α,”The Journal of Infectious Diseases, vol. 217, no. 3, pp. 393–404, 2018.

[13] I. N. Shalova, J. Y. Lim, M. Chittezhath et al., “Human mono-cytes undergo functional re-programming during sepsis medi-ated by hypoxia-inducible factor-1α,” Immunity, vol. 42, no. 3,pp. 484–498, 2015.

[14] H. Tsukamoto, S. Kozakai, Y. Kobayashi et al., “Impaired anti-gen‐specific lymphocyte priming in mice after Toll‐like recep-tor 4 activation via induction of monocytic myeloid‐derivedsuppressor cells,” European Journal of Immunology, vol. 49,no. 4, pp. 546–563, 2019.

[15] G. L. Semenza, “Defining the role of hypoxia-inducible factor 1in cancer biology and therapeutics,” Oncogene, vol. 29, no. 5,pp. 625–634, 2010.

[16] P. van Uden, N. S. Kenneth, R. Webster, H. A. Müller,S. Mudie, and S. Rocha, “Evolutionary Conserved Regulationof HIF-1β by NF-κB,” The Biochemical Journal, vol. 7, no. 1,article e1001285, 2011.

[17] G. Niu, J. Briggs, J. Deng et al., “Signal transducer and activatorof transcription 3 is required for hypoxia-inducible Factor-1αRNA expression in both tumor cells and tumor-associatedmyeloid cells,” Molecular Cancer Research, vol. 6, no. 7,pp. 1099–1105, 2008.

[18] A. Akinleye and Z. Rasool, “Immune checkpoint inhibitors ofPD-L1 as cancer therapeutics,” Journal of Hematology &Oncology, vol. 12, no. 1, p. 92, 2019.

[19] M. Feng, W. Jiang, B. Y. S. Kim, C. C. Zhang, Y. X. Fu, and I. L.Weissman, “Phagocytosis checkpoints as new targets for can-cer immunotherapy,” Nature Reviews Cancer, vol. 19, no. 10,pp. 568–586, 2019.

[20] K. Chang, C. Svabek, C. Vazquez-Guillamet et al., “Targetingthe programmed cell death 1: programmed cell death ligand1 pathway reverses T cell exhaustion in patients with sepsis,”Critical Care, vol. 18, no. 1, article R3, 2014.

8 Mediators of Inflammation

Page 9: Monocytes Undergo Functional Reprogramming to Generate

[21] Y. Zhang, J. Li, J. Lou et al., “Upregulation of programmeddeath-1 on T cells and programmed death ligand-1 on mono-cytes in septic shock patients,” Critical Care, vol. 15, no. 1,p. R70, 2011.

[22] W. Deng, S. Zhu, L. Zeng et al., “The circadian clock controlsimmune checkpoint pathway in sepsis,” Cell Reports, vol. 24,no. 2, pp. 366–378, 2018.

[23] C. Cubillos-Zapata, E. Hernández-Jiménez, V. Toledano et al.,“NFκB2/p100 is a key factor for endotoxin tolerance in humanmonocytes: a demonstration using primary human monocytesfrom patients with sepsis,” Journal of Immunology, vol. 193,no. 8, pp. 4195–4202, 2014.

[24] A. J. Hoogendijk, M. I. Garcia-Laorden, L. A. van Vught et al.,“Sepsis patients display a reduced capacity to activate nuclearfactor-κB in multiple cell types,” Critical Care Medicine,vol. 45, no. 5, pp. e524–e531, 2017.

[25] A. Zwergal, M. Quirling, B. Saugel et al., “C/EBPβ blocks p65phosphorylation and thereby NF-κB-mediated transcriptionin TNF-tolerant cells,” The Journal of Immunology, vol. 177,no. 1, pp. 665–672, 2006.

[26] B. K. Yoza, J. Y.-Q. Hu, S. L. Cousart, L. M. Forrest, and C. E.McCall, “Induction of RelB participates in endotoxin toler-ance,” The Journal of Immunology, vol. 177, no. 6, pp. 4080–4085, 2006.

[27] K. Qin, C. Han, H. Zhang, T. Li, N. Li, and X. Cao, “NAD+

dependent deacetylase sirtuin 5 rescues the innate inflamma-tory response of endotoxin tolerant macrophages by promot-ing acetylation of p65,” Journal of Autoimmunity, vol. 81,pp. 120–129, 2017.

9Mediators of Inflammation