Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2017

Open Access 01.12.2017 | Review

Metabolism-associated danger signal-induced immune response and reverse immune checkpoint-activated CD40+ monocyte differentiation

verfasst von: Jin Dai, Pu Fang, Jason Saredy, Hang Xi, Cueto Ramon, William Yang, Eric T. Choi, Yong Ji, Wei Mao, Xiaofeng Yang, Hong Wang

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2017

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Adaptive immunity is critical for disease progression and modulates T cell (TC) and antigen-presenting cell (APC) functions. Three signals were initially proposed for adaptive immune activation: signal 1 antigen recognition, signal 2 co-stimulation or co-inhibition, and signal 3 cytokine stimulation. In this article, we propose to term signal 2 as an immune checkpoint, which describes interactions of paired molecules leading to stimulation (stimulatory immune checkpoint) or inhibition (inhibitory immune checkpoint) of an immune response. We classify immune checkpoint into two categories: one-way immune checkpoint for forward signaling towards TC only, and two-way immune checkpoint for both forward and reverse signaling towards TC and APC, respectively. Recently, we and others provided evidence suggesting that metabolic risk factors (RF) activate innate and adaptive immunity, involving the induction of immune checkpoint molecules. We summarize these findings and suggest a novel theory, metabolism-associated danger signal (MADS) recognition, by which metabolic RF activate innate and adaptive immunity. We emphasize that MADS activates the reverse immune checkpoint which leads to APC inflammation in innate and adaptive immunity. Our recent evidence is shown that metabolic RF, such as uremic toxin or hyperhomocysteinemia, induced immune checkpoint molecule CD40 expression in monocytes (MC) and elevated serum soluble CD40 ligand (sCD40L) resulting in CD40+ MC differentiation. We propose that CD40+ MC is a novel pro-inflammatory MC subset and a reliable biomarker for chronic kidney disease severity. We summarize that CD40:CD40L immune checkpoint can induce TC and APC activation via forward stimulatory, reverse stimulatory, and TC contact-independent immune checkpoints. Finally, we modeled metabolic RF-induced two-way stimulatory immune checkpoint amplification and discussed potential signaling pathways including AP-1, NF-κB, NFAT, STAT, and DNA methylation and their contribution to systemic and tissue inflammation.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13045-017-0504-1) contains supplementary material, which is available to authorized users.
Abkürzungen
Ab
Antibody
Ag
Antigen
AP-1
Activator protein 1
APC
Antigen-presenting cell
BC
B cell
BCR
B cell receptor
BTLA
B and T lymphocyte attenuator
CKD
Chronic kidney disease
CpG
C, a cytosine triphosphate deoxynucleotide
CRTAM
Cytotoxic and regulatory T cell molecule
CTL
Cytotoxic T lymphocytes
CTLA-4
Cytotoxic T lymphocyte-associated protein 4
CVD
Cardiovascular disease
DAMP
Danger-associated molecular patterns
DC
Dendritic cell
DNAM-1
DNAX accessory molecule-1
EC
Endothelial cell
EPC
Epithelial cell
ERK
Extracellular signal-regulated kinase
FIB
Fibroblast
Foxp3
Forkhead box P3
GITR
Glucocorticoid-induced TNFR-related protein
GITRL
GITR ligand
HHcy
Hyperhomocysteinemia
HVEM
Herpes virus entry mediator
ICOS
Inducible T cell co-stimulator
IFN
Interferon
Ig
Immunoglobulin
IKK
I-κB kinase
IL
Interleukin
I-κBs
I-κB proteins
JNK
JUN amino-terminal kinase
LAT
Linker for activation of T cells
LPS
Lipopolysaccharide
MADS
Metabolism-associated danger signal
MAPK
Mitogen-activated protein kinase
MC
Monocyte
MHC
Major histocompatibility complex
Macrophage
NECL2
Nectin-like protein 2
NF-κB
Nuclear factor κB
NK
Natural killer
NLR
Nucleotide-binding and oligomerization domain-like receptors
Pam3CSK4
Tripalmitoyl-S-glycero-Cys-(Lys)4
PAMP
Pathogen-associated molecular patterns
PBMC
Peripheral blood mononuclear cells
PD-1
Programmed cell death protein 1
PD-L
PD ligand
PLCγ1
Phospholipase C gamma 1
Poly(I:C)
Polyinosinic-polycytidylic acid
PRR
Pattern recognition receptor
RF
Risk factors
sCD40L
Soluble CD40 ligand
STAT3
Signal transducers and activator of transcription-3
TC
T cell
TCR
T cell receptor
TGF-β
Transforming growth factor beta
Th
T helper cell
TIGIT
T cell immunoreceptor with Ig and ITIM domains
TIM
T cell (or transmembrane) immunoglobulin and mucin domain
TLR
Toll-like receptors
TNF
Tumor necrosis factor
TNFSF
Tumor necrosis factor superfamily
TRAF
Tumor necrosis factor receptor
Treg
Regulatory T cell
VEGF
Vascular endothelial growth factor
WBC
White blood cells
ZAP70
Zeta chain-associated protein kinase

Background

The immune system consists of innate and adaptive immunity. The classical innate immune system provides immediate and non-specific defense. It is activated by pathogens via a pathogen-associated molecular pattern (PAMP), which is recognized by pattern recognition receptors (PRR) in phagocytes (Fig. 1). Innate immunity may also be activated in response to injury, which releases a danger-associated molecular pattern (DAMP) also recognized by PRR. These two pathways are summarized as PAMP/DAMP+PRR recognition which leads to pathogen elimination, inflammatory responses, and antigen-presenting cell (APC) formation [1]. Evidence also suggests that the innate immune system targets innate T cells (TC) leading to TC activation [2, 3].
Different from innate immunity, adaptive immunity is featured by antigen (Ag) specificity, slow response, immunologic memorization, and low responsive cell ratio (Additional file 1: Table S1) [4]. Adaptive immunity comprises of cell-mediated immunity using TC and B cell (BC) humoral immunity. Each type of adaptive immunity contains three activating signals: (1) Ag recognition, (2) co-stimulation (we termed as immune checkpoint in this article), and (3) cytokine stimulation (Fig. 2). The term of immune checkpoint was initially proposed in 2009 referring to co-inhibitory immune checkpoint for TC suppression [5, 6] and was expanded in 2012 to include co-stimulatory immune checkpoint for TC activation [7]. The concept of immune checkpoint has been extensively studied in recent years and summarized in Table 1. It has become evident that the immune checkpoint plays an important regulatory role in adaptive immunity and determines the fate of the immune cell towards activation or suppression.
Table 1
Immune checkpoint families and paired molecules
https://static-content.springer.com/image/art%3A10.1186%2Fs13045-017-0504-1/MediaObjects/13045_2017_504_Tab1_HTML.gif
Immune checkpoints are classified as one-way immune checkpoint and two-way immune checkpoint based on signal 2 direction (forward only or both forward and reverse) and are further divided into stimulatory and inhibitory checkpoints. Listed are a few major immune checkpoint families. For example, in the CD28 family, CTLA-4 receptor binds to ligand B7-1(CD80) or B7-2(CD86) which are enriched in TC, BC, DC, MC, and MØ. The black frame emphasizes the two-way immune checkpoint to be focused. Words in the parentheses are aliases of the receptor and ligands
Abbreviations: AC apoptotic cell, BC B cell, BTLA B and T lymphocyte attenuator, CTLA-4 cytotoxic T lymphocyte-associated protein 4, CRTAM cytotoxic and regulatory T cell molecule, DC dendritic cell, DNAM-1 DNAX accessory molecule-1, EC endothelial cell, EPC epithelial cell, FIB fibroblast, GITR glucocorticoid-induced TNFR-related protein, GITRL GITR ligand, HVEM herpes virus entry mediator, ICOS inducible T cell co-stimulator, MC monocyte, macrophage, MAC mast cell, MYC myeloid cell, NKC natural killer cell, NE neuronal, NECL2 nectin-like protein 2, PA platelet, PS phosphatidylserine, PD-1 programmed cell death protein 1, PD-L PD ligand, SMC smooth muscle cell, TC T cell, TNFSF tumor necrosis factor superfamily, TIM T cell (or transmembrane) immunoglobulin and mucin domain, TIGIT T cell immunoreceptor with Ig and ITIM domains
Increased knowledge in immune checkpoints established advances in cancer medicine. For example, immune checkpoint molecule cytotoxic T lymphocyte-associated protein 4 (CTLA-4)-immunoglobulin (Ig) competes with CD28 to bind to CD80/CD86 and causes CTLA-4:CD80/CD86-induced TC suppression [8]. Antibodies against immune checkpoints, CTLA-4 (ipilimumab) and programmed cell death protein 1 (PD-1) (pembrolizumab and nivolumab), block CTLA-4:B7 and PD-1:PD-L1-induced TC suppression and thus enhance TC-dependent immune reaction [911]. These antibodies resulted in clinical regression of melanoma, non-small cell lung cancer, and other cancers [911]. Immune checkpoint therapy has also proven beneficial for inflammatory diseases such as rheumatoid arthritis and psoriasis using strategies to alleviate inflammation by engaging the inhibitory immune checkpoint [12, 13]. Immune checkpoint therapy for metabolic disease has not yet been realized, but it is an important consideration to balance TC responses and modulate immune checkpoints in contemplating therapies for metabolic disease.
The initial definition of immune checkpoints refers to receptor:ligand reaction towards TC suppression, also referred as co-inhibitory immune checkpoint. The immune checkpoint concept gradually evolved to incorporate co-stimulatory immune checkpoint and the identification of a reverse function of immune checkpoint towards APC [7, 14]. Recent evidence also suggests that metabolic risk factors (RF) can activate the stimulatory immune checkpoint leading to APC-related inflammatory responses [1519].
We propose a novel metabolism-associated danger signal (MADS) recognition, which promotes reverse stimulatory immune checkpoint leading to APC inflammation in both innate and adaptive immunity systems. MADS refers to intermediates and products of glucose, lipid, amino acid, nucleotide, hormone, and/or chemical metabolism, which can be recognized by the immune system via a metabolic sensor in a receptor-independent fashion.
In this article, we updated the molecular basis regulating innate and adaptive immunity. We proposed two novel nomenclatures, MADS recognition and reverse immune checkpoint, and suggested a new theory that MADS recognition regulates innate and adaptive immune response, via metabolic sensor, leading to immune cell activation and inflammation. Information described in this article should provide systemic knowledge and comprehensive insights into our understanding about immune response and immune checkpoints, especially the reverse stimulatory immune checkpoint in diseases.

Innate immunity recognizes novel MADS and regulates TC activation

Innate immunity and novel MADS recognition

The innate immune system is activated by pathogens via PAMP+PRR recognition and by injury-generated molecules via DAMP+PRR recognition (Fig. 1a). PRR are receptors presented on all immune cells and somatic cells, which bind to DAMP and PAMP to initiate inflammation [2, 3, 16, 20, 21]. Phagocytes, including macrophage (MØ), monocyte (MC), dendritic cell (DC), neutrophil, and natural killer (NK) cells, are activated by PAMP/DAMP+PRR recognitions which lead to pathogen elimination and inflammatory responses such as APC formation and cytokine generation [1]. Toll-like receptors (TLR) are a key PRR located on the cell surface and endosomes. Nucleotide binding and oligomerization domain-like receptors (NLR) are another important cytosolic-sensing DAMP receptor. In addition, transmembrane C-type lectin (TmCL), retinoid acid-inducible gene I (RIG-I), absent in melanoma 2 (AIM2), and receptor for advanced glycation end products (RAGE) are also characterized as classical DAMP-sensing receptors [22].
We and others provided evidence suggesting that metabolic RF activate innate immune systems leading to inflammatory responses. For example, lipid metabolite ox-LDL promoted NLRP3 inflammasome activation in MØ and foam cell formation [23]. Intermediate amino acid homocysteine (Hcy) induced nucleotide-binding oligomerization domain and leucine-rich repeat and pyrin domain containing protein 3 (NLRP3), causing NLRP3-containing inflammasome assembly, caspase-1 activation, and interleukin (IL)-1β cleavage/activation in EC [16]. Glucose, ceramide, islet amyloid polypeptide, and cholesterol crystals can be sensed by TLR or NLRP3-stimulating NLRP3 inflammasome complex assembly [16, 24, 25]. We [15] and others [26] demonstrated that MADS, such as Hcy or ox-LDL, induced MC activation in the absence of Ag within 48 h. Our data supported the notion that metabolism sensors mediate metabolic RF-induced inflammatory response in the innate immune system (Fig. 1a). Recently, we identified increased Hcy and a reduced ratio of S-adenosylmethionine (SAM)/S-adenosylhomocysteine (SAH), an indicator of cellular methylation, as the metabolic mediator/sensor for pro-inflammatory MC differentiation caused by uremic toxin in chronic kidney disease (CKD) [15].

Innate immunity in TC

CD4+ or CD8+ TC, including regulatory TC (Treg), express TLR and is directly involved in innate immunity (Fig. 1b). It is reported that PAMP/DAMP-TLR signaling lead to TC proliferation, inflammatory cytokine production, and glycolysis [2, 3]. Some inflammatory cytokines, such as IL-18, IL-12, IL-1β, IL-23, transforming growth factor (TGF)-β, and interferon (IFN)-α, quickly induced TC subset differentiation and proliferation and IFN-γ, IL-17, and IL-4 secretion in an Ag-independent fashion [6, 27, 28]. Super Ag caused non-specific TC activation and cytokine release [29]. In addition, lipid mediators, such as leukotrienes, are important activators for CD4+ and CD8+ TC recruitment to the site of infection and control fungal infection [30]. These evidences support the concept of innate immune response in TC via five mechanisms: PAMP/DAMP+PRR recognition, inflammatory cytokines, super Ag, and MADS recognition (Fig. 1).

Adaptive immunity recognizes MADS and regulates TC/BC activation

The major features of adaptive immunity are Ag specificity and immunologic memory which led to TC and BC activation (Additional file 1: Table S1). It was initially proposed that TC and BC activation involve three signals: signal 1 Ag recognition, signal 2 co-stimulation or co-inhibition, and signal 3 cytokine stimulation (Fig. 2). In this article, we termed signal 2 as the immune checkpoint which is in agreement with Dr. Pardoll’s suggestion in 2012 [7]. We defined immune checkpoint as interactions of paired molecules leading to either stimulatory or inhibitory immune response in TC and BC (other cells as well).

TC immunity (cell-mediated immunity) (Fig. 2a)

The discovery of TC receptors (TCR) led to defining TC activation signal 1, Ag recognition. Moreover, TC activation signal 2, immune checkpoint, was found to be essential for complete TC activation. For example, CD28 monoclonal antibody administration with simultaneously stimulating TCR leads to complete TC activation [5]. Signal 3, cytokine stimulation, is also involved in TC activation [31]. CD8+ TC’s response to virus was shown to be IFN-α dependent. We proposed a novel signal 4 because metabolic RF,, such as uremic toxin and hyperhomocysteinemia (HHcy), activated CD40:CD40L co-stimulatory immune checkpoint and increased serum soluble CD40L (sCD40L) levels [15].
Signal 1 (Ag recognition) is a vital immune process and determines the specificity of TC response. Ag is presented by major histocompatibility complexes (MHC) on the surface of an APC, then engaging with Ag-specific TCR on naïve TC contributing to TC activation/proliferation.
Signal 2 (immune checkpoint) plays a key role in regulating TC activation, differentiation, effector function, and deletion. Signal 2 was initially defined as co-stimulation and expanded to include co-inhibitory pathways [32]. In this article, we propose to term the co-stimulatory and co-inhibitory pathways collectively as the immune checkpoint. Immune checkpoint initially described co-inhibitory signal 2 in Topalian et al.’s papers [33] based on the discovery of T cell function restraint in normal physiologic settings and tumors [34]. This terminology was recently used to describe as a regulatory switch towards either stimulatory or inhibitory pathways [7]. Following Ag recognition or metabolic stimulation, an immune checkpoint ligand on APC binds to its receptor on TC determining TC activation or suppression. For example, CD28:B7 co-stimulatory immune checkpoint is essential for TC expansion and differentiation [35].
Signal 3 (cytokine stimulation) mediates cytokine-induced TC expansion and differentiation. For example, IL-12 and IFN-α/β, along with Ag and immune checkpoint, enhanced CD8+ TC clonal expansion [36]. The combination of IL-1β and IL-6 induced T helper (Th)-17 cell differentiation from human naïve TC (CD4+CD45RA+CCR7+CD25), in the presence of anti-CD3 (signal 1) and anti-CD28 (signal 2) antibodies [13]. IL-1β enhanced Th1, Th2, and Th17 cell proliferation with Ag stimulation in IL-1R1−/−Rag1−/− mouse [37].
Signal 4 (MADS recognition) is a novel signal we proposed based on our and other’s recent findings [15, 26]. Metabolic RF stimulates the expression of immune checkpoint molecules via a metabolic sensor, which in turn activates APC or TC and increases inflammatory cytokine production. We reported that uremic toxin, HHcy, and S-adenosylhomocysteine (SAH) increased CD40+ MC and sCD40L levels during a chronic time frame of CKD patients [15]. CD40:sCD40L molecular pair further promoted pro-inflammatory CD40+ MC and intermediate MC differentiation in 3 days. Moreover, studies in human subjects support that signal 4 MADS recognition may be involved in TC-related adaptive immunity in metabolic disorders [38]. The levels of sCD40L were found to be increased in subjects with metabolic syndrome and hypertension and negatively related to insulin sensitivity [39]. In addition, glucose sustains TC growth and proliferation upon TCR-dependent TC activation [40].

BC immunity (humoral immunity) (Fig. 2b)

BC immunity involves the same four signals which leads to antibody production and BC activation [41]. Signal 1 (Ag recognition) is the engagement of Ag with Ag-specific BC receptor (BCR). Signal 2 (immune checkpoint) is the ligation of immune checkpoint molecular pairs. Signal 3 (cytokine stimulation) describes Ag- and immune checkpoint-associated inflammatory cytokine regulation in BC activation. We proposed signal 4 (MADS recognition) for BC activation because the CD40:CD40L immune checkpoint is involved in BC activation [42] and sCD40L is induced in metabolic disease including CKD, HHcy, hypertension, hyperglycemia, and dyslipidemia [15, 39, 43].

Immune checkpoint regulates TC and APC activation

Immune checkpoints are molecular pairs (receptor:ligand) interactions regulating immune response towards TC and APC, also termed signal 2 (Fig. 2). We classified the immune checkpoint into two categories: one-way immune checkpoint for forward signaling towards TC only, and two-way immune checkpoint for both forward and reverse signaling towards TC and APC, respectively (Fig. 3). Each category can be further divided into stimulatory and inhibitory immune checkpoints. The stimulatory immune checkpoint turns up the immune system leading to immune cell proliferation or activation, while the inhibitory immune checkpoint turns down the immune system leading to immune cell suppression or death (Fig. 3a).

One-way immune checkpoint

One-way immune checkpoint refers to forward signaling only towards TC. It is dual-functional as it can modulate cell fate for proliferation or death (Fig. 3b).
Forward stimulatory molecular pairs promote TC proliferation, cytokine production, differentiation, cytotoxic function, memory formation, and survival. A well-described forward stimulatory molecular pair is CD28:B7. Interaction of CD28:B7 results in distinct phosphorylation, transcriptional activation, and cytokine and chemokine production that are essential for TC expansion and differentiation [35]. Metabolic product ceramide is involved in the forward stimulatory immune checkpoint in TCR-dependent TC activation at multiple levels [44].
The forward inhibitory molecular pair ligation in the one-way immune checkpoint leads to TC tolerance, exhaustion, apoptosis, cell cycle arrest, and effector function inhibition. For example, CD8+ tumor-infiltrating lymphocytes exhibit high proliferation and IL-2/tumor necrosis factor (TNF)-α production in TC immunoreceptor with Ig and ITIM domain (TIGIT)−/− mice [45], indicating TIGIT inhibited the effector function and proliferation of CD8+ TC.

Two-way immune checkpoint

The two-way immune checkpoint is bi-directional, towards both TC and APC. Similar as the one-way immune checkpoint, it is also dual-functional as it modulates cell fate for proliferation or death (Fig. 3c).
The stimulatory molecular pairs in the two-way immune checkpoint activate TC and APC. CD40:CD40L is one of the best-described stimulatory pairs in the two-way immune checkpoint. CD40 binds to its ligand CD40L, which is usually transiently expressed on TC [46] and modulates effector function and differentiation of TC. This is seen in CD40−/−APOE−/− mice as they have lower effector memory CD4+/CD8+ TC in the spleen [47]. Ligation of CD40L on TC with CD40 on BC promoted BC Ig isotype switching, which was associated with X-linked hyper IgM syndrome in humans [48]. Moreover, metabolic RF cholesterol crystal is required for TCR nanoclustering in TC, which enhances the avidity of the TCR-antigen interaction [49]. Reversely, cholesterol crystals trigger pro-inflammatory cytokine secretion from APC MØ [50].
The inhibitory molecular pairs in the two-way immune checkpoint lead to TC and APC suppression or death. The ligation of PD-1 and PD-L1 results in TC inactivation, IL-12 reduction, antitumor immunity suppression, and tumor progression [51]. Thus, PD-1:PD-L1 immune checkpoint therapy using PD-1 antibodies (pembrolizumab and nivolumab) achieved great success in melanoma, bladder cancer, and gastric cancer therapy [911]. Further, PD-1 delivered inhibitory signals through B7-H1 on APC [52]. Again, metabolic RF cholesterol sulfate inhibited TCR signaling [53] as well as sterologenesis in APC fibroblasts [54].

Immune checkpoint family and paired molecules

Representative paired immune checkpoint molecules (receptor:ligand) are summarized in Table 1 and listed according to immune checkpoint direction (one-way and two-way) and function (stimulatory and inhibitory). The classification of immune checkpoint families is determined by the checkpoint receptor component. Most immune checkpoint receptors are members of immunoglobulin superfamily (IgSF) and tumor necrosis factor receptor superfamily (TNFRSF), which can be further divided into specific subfamilies based on the primary amino acid sequence, protein structure, and function [52]. Notably, the majority of immune checkpoint ligands are expressed on multiple immune cells.
IgSF checkpoint receptor superfamily contains CD28, B7, CD226, TC (or transmembrane) immunoglobulin, mucin domain (TIM), and CD2/signaling lymphocytic activation molecule (SLAM) subfamilies, which participate in forward stimulatory and forward inhibitory immune checkpoints. For example, CD28 subfamily including CD28 and CD278 (inducible TC co-stimulator, ICOS) transduce stimulatory response. Other members in the CD28 subfamily, such as CTLA-4, PD-1, PD-1 homologue (PD-1H), and B and T lymphocyte attenuator (BTLA), transduce inhibitory response.
TNFRSF checkpoint receptor superfamily contains Type-V, Type-L, Type-s, and orphan subfamilies and recognizes TNF superfamily (TNFSF) molecules [52]. The common feature of TNFRSF:TNFSF is bi-directional (both forward and reverse immune checkpoint) [52].
The Type-L subfamily, also called conventional TNFRSF immune checkpoint receptors, has the most members in TNFRSF, but only CD40, herpes virus entry mediator (HVEM), death receptor 3 (DR3), and lymphotoxin-β receptor (LTBR) have a co-stimulatory function, while CD120a, CD120b, and CD95 have apoptosis function on TC [52, 55]. The Type-V subfamily, also called divergent, is the only family where all members have co-stimulatory function, including 4-1BB (CD137), OX40 (CD134), CD27, CD30, and glucocorticoid-induced TNFR-related protein (GITR) [55]. Among the Type-s subfamily, transmembrane activator and CAML interactor (TACI), B cell-activating factor receptor (BAFFR), and B cell maturation protein (BCMA) have the function of B cell activation, survival, and differentiation [52, 55]. The function of the orphan subfamily remains unclear, except that the receptor expressed in lymphoid tissues (RELT) has some evidences of stimulating TC proliferation [56].
We list six pairs of TNFRSF:TNFSF molecules in Table 1: CD40:CD40L, 4-1BB (CD137):4-1BBL, OX40 (CD134):OX40L, CD27:CD70, CD357 (GITR):GITRL, and CD30:CD30L, and discuss their characterizations in the following section.

Two-way stimulatory immune checkpoint induces tissue and systemic inflammation

Emerging evidences suggested that the two-way stimulatory immune checkpoint is critical for TC activation and APC inflammation. We summarized recent studies elucidating two-way stimulatory immune checkpoint with immune cell responses in human and mouse disease models (Tables 2 and 3).
Table 2
Two-way stimulatory immune checkpoint induces tissue and systemic inflammation (human study)
Checkpoint (receptor:ligand)
Disease
Functional change
PMID#
Year
Forward immune checkpoint (towards TC)
 CD40:CD40L
ACS
Lesion CD40L+ TC↑
12732389
2003
Bladder tumor
Peripheral T cell↓ to agonistic IgG1 chimeric α-CD40 Ab
25589626
2015
 CD137:CD137L
AS
CD137↑ in lesion CD8+ TC
18285570
2008
Head/neck cancer
CD4+/CD8+ TC proliferation maker Ki67↑ to CD137 agonist
27496866
2016
 CD134:CD134L
ACS
CD134+ CD4+CD28null TC express INF-γ/TNF-α,
22282196
2012
Colorectal cancer
CD134+ CD8+ TC infiltration enhances prognostic
26439988
2015
 CD27:CD70
NSCLC
Higher CD4/CD8 TC ratio in CD70+ tumor cell
25951351
2015
 CD30:CD30L
Lymphoma
CD30+ Th2 proliferation↓ by CD30L
12855655
2003
 GITR:GITRL
Liver tumor
Tumor-infiltrating Treg-suppressive capacity↓ by GITRL
26587321
2015
Reverse-immune checkpoint (towards APC)
 CD40:CD40L
CVD + CKD
CD40+ MC↑, MC inflammatory markers↑
27992630
2016
Cancer
CD40L mediate TIS-TC induced inflammatory MC/MØ
25375372
2014
 CD137:CD137L
AAS
CD137L+ CD14+ MC↑
24899613
2014
MM
CD137 inhibit MM cell proliferation and induce death
20520765
2010
 CD134:CD134L
ACS
OX40L+ MC↑
22282196
2012
MLC
OX40L+ MLC-DC express higher CD80+/CD86+/HLA-DR
14984494
2004
 CD27:CD70
Lymphoma
Anti-CD70 mediate MØ phagocytosis
17038522
2007
 CD30:CD30L
Lymphoma
Anti-CD30 mediate MØ phagocytosis
17909075
2007
 GITR:GITRL
AS
Plaque MØ activation↑
17067317
2006
Stimulatory immune checkpoint has been described in human and mouse metabolic diseases. For example, CD40:CD40L two-way stimulatory immune checkpoint was activated and associated with increased lesion CD40L+ TC in ACS and CD40+ MC in CKD in humans. CD40 KO reduced spleen effector memory CD4+/CD8+ TC and blood Ly6C+ MC and aorta M1 MΦ. Similar functional change was observed for immune checkpoint CD40:CD40L CD137:CD137L and OX40:OX40L. ↑ refers to higher population/expression level/activity. ↓ refers to lower population/expression level/activity
Abbreviation: AAS acute atherothrombotic stroke; CAR chimeric antigen receptor; CKD chronic kidney disease; CVD cardiovascular disease; CA carcinoma; EC endothelial cell; GITR glucocorticoid-induced TNFR-related protein; GITRL GITR ligand; KPC KrasLSL-G12D/+, Trp53LSL-R172H/+, Pdx1-Cre; MC monocyte; macrophage; mAb agonist monoclonal antibodies; MM multiple myeloma; MLC myeloid leukemia cell; NSCLC non-small cell lung cancer; NKC natural killer cell; SCID severe combined immunodeficient; TC T cell; Treg regulatory T cell; TG transgene; TIS tumor induce Senescent; VEGF vascular endothelial growth factor
Table 3
Two-way stimulatory immune checkpoint induces tissue and systemic inflammation (mouse study)
Checkpoint (receptor:ligand)
Genotype
Disease
Functional change
PMID#
Year
Forward immune checkpoint (towards TC)
 CD40:CD40L
CD40−/−APOE−/−
AS
Spleen memory CD4+/CD8+ TC↓
20100871
2007
Rag1−/−
Lung CA
Tumor Th1/Th17↑, Treg/Th2↓ to CD40 agonist
25651850
2015
 CD137:CD137L
APOE−/−
AS
Lesion CD3+/CD8+ TC↑ to CD137 agonist
18285570
2008
NSG
Osteosarcoma
Ameliorate CAR TC exhaustion to CD137
25939063
2015
 CD134:CD134L
LDLR−/−
AS
Lesion CD3+ TC↓ to α-OX40L
24068673
2013
C57BL/6
Mammary CA
Blood effector/memory TC↑ to α-OX40+Dribbles
27874054
2016
 CD27:CD70
CD70−/−LDLR−/−
AS
Spleen Treg↓
27786334
2017
CD27−/−
Solid tumor
Spleen Treg↓, tumor CD3+ TC infiltration↑
22628427
2012
 CD30:CD30L
LDLR−/−
AS
Adventitial CD3+ TC↓ to α-CD30L
23087358
2012
C57BL/6
Fibrosarcoma
CD30+ Vδ TC drives cancer progression
27384869
2016
 GITR:GITRL
LDLR−/−GITRLTG
AS
Lymph node Terg/effector memory CD4+ TC↑
27444204
2016
Reverse-immune checkpoint (towards APC)
 CD40:CD40L
CD40−/−APOE−/−
AS
Blood Ly6C+ MC↓, aorta M1 MØ↓
20100871
2007
KPC
Pancreatic CA
Tumor M1 MØ↑ to CD40 agonist
21436454
2011
 CD137:CD137L
CD137−/−APOE−/−
AS
Aorta CD11b+ MC/MØ↓
25059229
2014
C57BL/6
Liver CA
Tumor iNOS-positive MØ↑ to CD137 agonist
24789574
2014
 CD134:CD134L
OX40L−/−APOE−/−
AS
VEGF-induced angiogenesis↓
20584752
2010
C57BL/6
Sarcoma
Tumor M1 MØ↑ to CD134 agonist
22578109
2012
 CD27:CD70
CD70TG APOE−/−
AS
Circulating MC viability↓
20505312
2010
SCID
Lymphoma
Delete MØ reduce survival to α-CD70
17038522
2007
 CD30:CD30L
LDLR−/−
AS
Lesion MØ/BC no affect
23087358
2012
SCID
Lymphoma
Delete MØ reduce survival to α-CD30
17909075
2007
GITR:GITRL
C57BL/6
Liver tumor
Tumor M1 MØ↑ to GITR agonist+sunitinib
26239999
2016
Stimulatory immune checkpoint has been described in human and mouse metabolic diseases. For example, CD40:CD40L two-way stimulatory immune checkpoint was activated and associated with increased lesion CD40L+ TC in ACS and CD40+ MC in CKD in humans. CD40 KO reduced spleen effector memory CD4+/CD8+ TC and blood Ly6C+ MC and aorta M1 MΦ. Similar functional change was observed for immune checkpoint CD40:CD40L CD137:CD137L and OX40:OX40L
Abbreviation: AAS acute atherothrombotic stroke; CAR chimeric antigen receptor; CKD chronic kidney disease; CVD cardiovascular disease; CA carcinoma; EC endothelial cell; GITR glucocorticoid-induced TNFR-related protein; GITRL GITR ligand; KPC KrasLSL-G12D/+, Trp53LSL-R172H/+, Pdx1-Cre; MC monocyte; macrophage; mAb agonist monoclonal antibodies; MM multiple myeloma; MLC myeloid leukemia cell; NSCLC non-small cell lung cancer; NKC natural killer cell; SCID severe combined immunodeficient; TC T cell; Treg regulatory T cell; TG transgene; TIS tumor induce Senescent; VEGF vascular endothelial growth factor

CD40:CD40L two-way immune checkpoint

CD40:CD40L is the first discovered stimulatory molecular pair of TNFRSF:TNFSF. CD40 is not only expressed on immune cells (BC, MC, MØ, DC) but also on a variety of somatic cells such as endothelial cell (EC), smooth muscle cell (SMC), fibroblast, and platelet [57]. CD40 was initially discovered as a surface receptor on BC binding to CD40L on TC causing TC polyclonal activation and BC proliferation/differentiation [46]. CD40L is the sole ligand for CD40 and is also known as CD154. CD40L has two forms, membrane-bound CD40L and sCD40L. Membrane-bound CD40L is expressed on activated TC, MC, MØ, platelet, mast cell, and EC [58]. sCD40L circulates in the blood and is mainly produced by platelets [59]. The CD40:CD40L two-way immune checkpoint promotes atherosclerosis and inhibits tumor progress and has been used as a cancer immunotherapy target [6062]. sCD40L is significantly elevated in patients with cardiovascular disease (CVD) and CKD [15] and proposed as an independent predictor and biomarker for cardiovascular events after acute coronary syndrome and plaque vulnerability [63]. CD40:CD40L interactions stimulate the expression of inflammatory cytokines, adhesion molecules, chemokines, matrix degrading enzymes, and platelet tissue factor. CD40−/−ApoE−/− mice exhibited 55% plaque reduction and less lipid-containing, collagen-rich, stable plaque, and improved reendothelialization [64]. Similarly, anti-CD40L antibody induced a stable lesion with lipid-poor, collagen-rich plaque in ApoE−/− mice [65]. CD40-RNAi-lentivirus prevented plaque progression in ApoE−/− mice [66].

CD40:CD40L forward immune checkpoint

The influence of CD40:CD40L forward immune checkpoint towards TC is well established. TC presents at all stages of atherosclerotic lesion. The major subset of TC in atherosclerotic plaques is Th1 CD4+ TC. CD40−/−ApoE−/− mice have a lower effector memory CD4+/CD8+ TC in the spleen [47]. Anti-CD40L antibody reduced TC content in mouse atheroma [67]. Moreover, the CD40:CD40L immune checkpoint inhibited Treg activation, as CD40L−/− bone marrow reconstitution in LDLR−/− mice led to increased Treg [68], and agonistic CD40 antibody reduced Treg in Lewis lung cancer mouse model [69].

CD40:CD40L reverse immune checkpoint

Large amount of evidence described the impact of CD40:CD40L reverse stimulatory immune checkpoint towards APC. In the absence of CD40L on TC, BC only secrete IgM and cannot switch to other Igs (IgG, IgE, IgA). CD40L on TC binds to CD40 on MØ and leads to MØ activation and secretion of matrix metalloproteinase (MMP), pro-inflammatory cytokines (Il-12, TNF-α, IL-1β, IL-6, and IL-8), and platelet tissue factor. Similarly, CD40L gene mutation caused X-linked hyper IgM syndrome which is characterized by low or absent levels of IgG, IgE, and IgA but normal or elevated serum levels of IgM [48]. MC-derived DC from patients with coronary artery disease (CAD) expressed higher CD40 which was associated with smoking history, higher C-reactive protein, and lower high-density lipoprotein cholesterol (HDL-C) [70]. We reported that CD40+ MC was increased in patients with CVD and further elevated in patients with CVD+CKD. Anti-CD40L antibody significantly reduced MØ in mice [67]. CD40−/−ApoE−/− mice exhibited lower pro-inflammatory Ly6C+ MC in blood and M1 MØ in the aorta [47]. Moreover, CD40 agonist activated antitumor MØ infiltration and resulted pancreatic cancer regression in mice [71].

CD137 (4-1BB):CD137L (4-1BBL) two-way immune checkpoint

CD137 is mainly expressed on activated CD4+ TC and also on BC, MC, DC, and EC, while CD137L is constitutively expressed on APC and activated TC [72]. Soluble CD137 (sCD137) is elevated in human acute coronary syndrome (ACS) and atherothrombotic stroke [73, 74] and has been suggested as a prognostic biomarker for acute atherosclerotic disease. The CD137:CD137L immune checkpoint promotes vascular inflammation as CD137−/−ApoE−/− and CD137−/−LDLR−/− mice had reduced atherosclerotic lesions and inflammation [75] and anti-CD137 antibody decreased atherosclerosis lesion in ApoE−/− mice [76].

CD137:CD137L forward immune checkpoint

The CD137:CD137L forward immune checkpoint promotes TC activation. CD137 is expressed predominantly in CD8+ TC and occasionally in CD4+ TC in human atherosclerotic lesions and associated with pro-inflammatory factor release such as TNF-α, IL-1β, and IFN-γ. CD137 agonist induced CD8+ TC infiltration in mouse atherosclerotic lesions and promoted the progression of atherosclerosis [76]. In peripheral blood mononuclear cells (PBMC), antibody against CD137 decreased TNF-α and IFN-γ production from CD4+CD28null TC which expresses higher levels of CD137 compared with CD4+CD28+ TC [77]. The CD137:CD137L checkpoint also enhances tumor immunity, as CD137 agonist promoted CD4+ and CD8+ TC proliferation in patients with head and neck cancer [78].

CD137:CD137L reverse immune checkpoint

Recent research emphasized the role of the CD137:CD137L reverse stimulatory immune checkpoint on MC and MØ differentiation. Cross-linking of CD137L by CD137 on human PBMC induced IL-6, IL-8, IL-12, TNF-α, and IFN-γ production and inflammatory DC differentiation [79]. Circulating CD137L+CD14+ MC was increased in patients with acute ischemic atherosclerotic stroke [74]. CD137−/−ApoE−/− mice have lower MC and MØ in the aorta [80]. Anti-CD137 monoclonal antibody induced iNOS-positive MØ differentiation in hepatoma tissue in mice [81].

CD134(OX40):CD134L(OX40L) two-way immune checkpoint

CD134 is mainly expressed on activated CD4+ TC, CD8+ TC, and memory TC, while CD134L is expressed on mature APC, activated TC, and EC [82]. The levels of sOX40L were significantly increased in patients with ACS [83]. Anti-CD134L antibody significantly reduced atherosclerotic lesion in LDLR−/− mice [84, 85].

CD134:CD134L forward immune checkpoint

Similar as CD137, CD134 is highly expressed in CD4+CD28null TC. CD134 also regulates Treg function by suppressing Treg generation from naïve TC and effector TC in mice [86]. CD134L induced INF-γ CD4+ TC proliferation in cultured splenocytes from ApoE−/− mice [87]. Antibody against CD134 decreased TNF-α and IFN-γ production in CD4+CD28null TC derived from PBMC from ACS patients [77]. Anti-CD134L antibody reduced the populations of circulating CD4+CD134+ TC, CD4+ TC and CD8+ TC, and lesion CD3+ TC in LDLR−/− mice [84]. Anti-CD134 antibody combined with autophagosomes (DRibbles) induced memory and effector TC proliferation and differentiation and promoted tumor regression in mice [88]. Elevating CD134+ CD8+ TC infiltration in colorectal cancer prolonged overall survival in humans [89].

CD134:CD134L reverse immune checkpoint

Even though circulating MC expressed the highest level of CD134L in ACS patients [77], the atherogenic role of CD134:CD134L may not be mediated by MC and MØ. Anti-CD134L antibody had no effect on both M1 MØ and M2 MØ in ApoE−/− mice [87]. CD134:CD134L may participate in BC Ig isotype switch, as blocking the CD134:CD134L immune checkpoint using anti-CD134L antibody increased anti-ox-LDL IgM, a protective IgM, in LDLR−/− mice [85]. Moreover, agonistic CD134 antibody increased M2 MØ in tumor. M2 MØ produced higher IL-10 and chemokine (C-C motif) ligand (CCL)-17 and lower IL-12-b and IL-23 compared to M1 MØ, which limited the efficacy of CD134 agonist therapy in mice [90, 91].

CD27:CD70 two-way immune checkpoint

In contrast to CD134 and CD137, CD27 is expressed on naïve TC, BC, and NK cells and upregulated on activated TC, while CD70 is expressed on APC and activated TC [92]. Evidence for the role of CD27:CD70 in atherosclerosis is conflicting as ruptured atherosclerotic plaques expressed higher CD70 than those in stable lesions [93], and CD70 transgenic mice attenuated atherosclerotic development [94].

CD27:CD70 forward immune checkpoint

CD27 promotes activated TC proliferation and survival. CD27+ Treg is reduced in myocardial infarction patients, and this subset has high suppressive potential [95]. CD70 deficiency reduced spleen Treg in ApoE−/− mice [93] and CD27 deficiency reduced Treg in solid tumor in mice [96], suggesting that CD27:CD70 may have an immunosuppressive role in atherosclerosis and tumor growth.

CD27:CD70 reverse immune checkpoint

The CD27:CD70 reverse stimulatory immune checkpoint towards APC may be protective for atherosclerosis. CD70 transgenic mice displayed increased MC apoptosis [94]. CD70 promoted ox-LDL efflux in MØ [93] while engineered anti-CD70 increased MØ phagocytosis and prolonged the survival in lymphoma mice [97].

CD30:CD30L two-way immune checkpoint

CD30 is expressed on activated TC and BC, while CD30L is expressed on APC and activated TC [98]. CD30 was originally recognized as a cancer-associated surface antigen in TC. The CD30:CD30L two-way immune checkpoint promotes atherosclerosis and tumor and is a therapeutic target for both diseases. The CD30 antibody is used to treat Hodgkin’s lymphoma, anaplastic large cell lymphoma, and other cancers [99]. A few studies demonstrated that the CD30:CD30L blockade delayed the development of atherosclerosis.

CD30:CD30L forward immune checkpoint

CD30 primarily promotes CD4+ TC activation. Anti-CD30L treatment reduced CD4+ TC counts but had no effect on CD8+ TC, Th1, Th2, Th17, and Treg cell numbers in LDLR−/− mice [100]. Recombinant CD30L inhibited CD30+ Th2 lymphoma cell proliferation [101].

CD30:CD30L reverse immune checkpoint

CD30:CD30L may not affect APC function in atherosclerosis, as anti-CD30L treatment did not change BC counts, ox-LDL-specific IgM/IgG, and aortic MC numbers in LDLR−/− mice [100]. Anti-CD30 antibody enhanced MØ phagocytosis in tumor tissue and increased survival in mice [102].

CD357(GITR):CD357L(GITRL) two-way immune checkpoint

GITR is expressed on naïve TC, increased on activated TC, and is also present on BC and NK cells, while GITR ligand (GITRL) is expressed on APC [103]. GITR:GITRL may have a protective role in atherosclerosis via regulating Treg. GITRL transgenic BM transplantation reduced atherosclerotic lesion in LDLR−/− mice [104].

GITR:GITRL forward immune checkpoint

It is known that GITR:GITRL interaction is important for CD4+ TC, CD8+ TC, and Treg differentiation and expansion. Thus, GIRT is used as a Treg marker. GITRL transgenic chimeric LDLR−/− mice displayed an increased effector TC and Treg and reduced atherosclerosis [104]. sGITRL suppressed Treg infiltration in human liver tumor [105].

GITR:GITRL reverse immune checkpoint

GITR and GITRL are mainly expressed in MØ in plaques. However, the protective role of GITR:GITRL in atherosclerosis is controversial. Anti-GITR mAb induced human MC and MØ activation, MMP-9, and pro-inflammatory cytokine expression, which may promote atherosclerosis and plaque instability [106]. Agonistic GITR antibody promoted M1 MØ differentiation in mice liver tumor [107].

Molecular mechanisms underlying CD40:CD40L two-way immune checkpoint amplification

We summarized two molecular signaling pathways previously reported for the CD40:CD40L immune checkpoint: forward stimulatory immune checkpoint towards TC and reverse stimulatory immune checkpoint towards APC (Fig. 4a, b). In addition, we propose a novel pathway, the TC contact-independent immune checkpoint (Fig. 4c) based on our recent discoveries [15].
We found that metabolic RF, such as uremic toxin and HHcy, induced circulating sCD40L and CD40+ MC in CKD patients. Also, both sCD40L and HHcy promoted inflammatory CD40+ MC and intermediate MC differentiation in cultured human PBMC [15]. Other metabolic RF, such as triazolopyrimidine, inhibited CD40-associated MC activation [108]. A mechanistic study showed that SAH-related DNA hypomethylation is responsible for CD40+ MC differentiation in human PBMC [15]. We were the first to establish a direct mechanistic link between HHcy and increased cellular SAH and to propose that SAH-related hypomethylation is a key biochemical mechanism for HHcy-induced CVD in EC [109111]. We believe that Hcy and SAH function as metabolic sensors and are responsible for DNA hypomethylation and APC activation.

CD40:CD40L forward stimulatory immune checkpoint (Fig. 4a)

The CD40:CD40L forward stimulatory immune checkpoint follows signal 1 (Ag recognition) and leads to TC activation. During this process, MHC presents Ag to TCR, which triggers the assembling of TCR, CD3, and TCRζ chain. The subsequent CD40:CD40L immune checkpoint interaction amplifies the activation of three transduction pathways via the recruitment of zeta chain-associated protein kinase of 70 kDa (ZAP-70) and phosphorylation of linker for activation of T cells (LAT), RAS mitogen-activated protein kinase (MAPK) pathway, calcium-calcineurin pathway, and nuclear factor κB (NF-κB) pathway [112].

CD40:CD40L reverse stimulatory immune checkpoint (Fig. 4b)

In APC, the CD40:CD40L reverse stimulatory immune checkpoint is associated with proliferation of MC, MØ, BC, SMC, and tumor cells, and inflammatory molecular production. CD40 can bind to TNF receptor-associated factor (TRAF1-3/5-6) and activate three TNF signaling, including signal transducers and activator of transcription-3 (STAT3), NF-κB, and activator protein 1 (AP-1) pathways in cell type and TRAF member-dependent manner. For example, STAT3 can be activated by CD40:TRAF2/3 ligation via JAK in BC [113]; NF-κB can be activated by CD40:TRAF1-3/5-6 interaction via IKK/I-κB in BC and MC; and AP-1 can be activated by CD40:TRAF6 via MAPK in MC and MØ [114]. Moreover, TRAF1/2/3/5 activation is linked to NF-κB, MAPK/p38, and JUN amino-terminal kinase (JNK) pathway, while TRAF6 activates NF-κB, protein kinase B, and STAT3 pathway [113]. CD40:TRAF6 has a critical role in promoting atherosclerosis, as attenuated atherosclerosis and reduced Ly6C+ MC and M1 MØ were observed in CD40−/−TRAF6−/−ApoE−/− but not in CD40−/−TRAF2/3/5−/−ApoE−/− mice [47].

CD40:CD40L TC contact-independent immune checkpoint (Fig. 4c)

This is a novel pathway we proposed based on our and others discoveries [15, 115]. We demonstrated that metabolic RF, such as uremic toxin and HHcy, and sCD40L promoted inflammatory CD40+ MC and intermediate MC differentiation in culture human PBMC in the absence of TC [15]. We hypothesize that metabolic RF promote pro-inflammatory MC differentiation via metabolic sensors, such Hcy and SAH and DNA hypomethylation. This is based on evidence from mediation analysis showing increased plasma Hcy and SAH levels and consequential reduction of SAM/SAH ratio, a recognized indicator of methylation status, and from mechanistic studies showing Hcy-suppressed DNA methylation in CD40 promoter and folic acid, a methylation rescue reagent, reversed CD40+ MC differentiation in human PBMC [15]. We were the first to establish a direct mechanistic link between Hcy and increased cellular SAH with hypomethylation and to propose hypomethylation as a key biochemical mechanism for HHcy-induced CVD [109111]. Our discoveries suggested that the TC contact-independent immune checkpoint is a critical mechanism for systemic and tissue inflammatory response in metabolic disorders.

CD40+ MC is a novel and stronger inflammatory MC subset

MC heterogeneity has been widely acknowledged. MC expresses various receptors, which sense the environment stimulation and mediate cell differentiation towards inflammatory or anti-inflammatory subsets. MC is the most invasive immune cells which can transmigrate into tissue causing tissue inflammation and repair. In humans, MC are divided into three functionally distinct subsets according to the surface marker CD14 and CD16 [116]. CD14 is used as a marker for human MC. The common MC subsets by nomenclature is classified as (1) classical MC (CD14++CD16 phagocytic MC), (2) intermediate MC (CD14++CD16+ pro-inflammatory MC), and (3) non-classical MC (CD14+CD16++ patrolling MC) [116]. However, such human MC classification is not in harmony, as further increased expression of CD16, an inflammatory marker, is associated with anti-inflammatory function in non-classical MC subsets.
Our recent findings resolved the above controversy in MC subset classification and presented CD40+ MC as a novel and stronger pro-inflammatory MC subset compared with the nomenclature-defined intermediate MC (Table 4) [15]. By examining the expression of nine inflammatory markers in three nomenclature-defined MC subsets and CD40+ MC [15], we discovered that CD40+ MC expressed higher levels of TC activation receptor CD86, chemokine receptor CCR2, and expressed similar levers of other inflammatory surface markers than that on nomenclature-defined intermediate MC (Table 4). In contrast, CD40 MC exhibited much lower levels of TC activation receptor HLA-DR, adhesion receptor CD49d, and chemokine receptor CX3CR1 than that on common nomenclature-recognized anti-inflammatory (patrolling) non-classical MC subset.
Table 4
CD40+ MC is a novel and stronger pro-inflammatory MC subset compared with intermediated MC
Inflammatory surface maker
Pro-inflammatory MC
Anti-inflammatory MC
CD40+ MC (CD40+CD14+)
Intermediate MC (CD14++CD16+)
CD40 MC (CD40CD14+)
Non-classical MC (CD14+CD16++)
TC activation receptor
 CD86
+++
++
+
+
 CD80
+
+
+
+
 LA-DR
++
++
+
+++
Adhesion receptor
 CD62L
++
++
++
+
 CD11b
+++
+++
+
+
 CD49d
+++
+++
+
+++
Chemokine receptor
 CCR2
+++
+
++
+
 CCR5
+++
+++
+
+
 CX3CR1
+++
+++
++
+++
Inflammatory features of human CD40+ MC were characterized using nine inflammatory surface makers by flow cytometry analysis (experimental details in Yang et al. [15]). WBC from healthy subjects were isolated and stained with anti-CD14, anti-CD16, and anti-CD40 antibodies and co-stained with surface markers for TC activation (CD86, CD80, and HLA-DR), adhesion receptors (CD62L, CD11b, and CD49d), and chemokine receptors (CCR2, CCR5, and CX3CR1). Compared with the previously established inflammatory intermediate MC subset, CD40+ MC expressed higher levels of CD86 and CCR2 and similar levels of other inflammatory markers. CD40 MC expressed much lower levels of HLA-DR, CD49d, and CX3CR1 and similar levels of other inflammatory markers compared with non-classic MC
Abbreviations: MC monocyte, TC T cell, WBC white blood cells
On the other hand, classically defined pro-inflammatory intermediate MC expressed lower levels of inflammatory markers CCR2, HLA-DR, and CD62L compared with classical (phagocytic) and non-classical (patrolling) MC [15]. This is inconsistent with the inflammatory feature of these MC subsets.

CD40+ MC is a reliable biomarker of CKD severity

CKD is considered as a metabolic complication. Patients with CKD have 10 to 30 times higher cardiovascular mortality than the general population, and 50% of deaths in end-stage CKD were due to CVD [117]. MC is the key player in the development of atherosclerosis. Intermediate MC was elevated in patients with CVD compared with healthy subjects [15] and in patients with ST-elevation myocardial infarction. Its population is positively correlated with cardiovascular events, such as cardiovascular death, acute myocardial infarction, and non-hemorrhagic stroke [118]. However, there are a few contradictory dilemmas regarding the molecular marker and biological function of the currently defined three MC subsets [15]. For example, (1) the intermediate CD14++CD16+ (pro-inflammatory) MC expresses very high levels of anti-inflammatory marker CX3CR1 and (2) high levels and absence of CD16 were presented in CD14+CD16++ (phagocytic) and CD14++CD16 (patrolling) MC, respectively, both displaying an anti-inflammatory function. Therefore, there is no strong consensus to use the intermediate CD14++CD16+ MC as a reliable biomarker for the severity of CVD and metabolic disease.
Currently, CKD severity is determined by estimated glomerular filtration rate (eGFR) which is a prediction parameter calculated using blood creatinine, age, race, gender, and other factors. We believe CD40+ MC is a more accurate and reliable biomarker for CKD and CVD [15]. As shown in Fig. 5, CD40+ MC subset was elevated in patients with CVD and CVD+CKD compared with healthy subjects and increased with the elevation of CKD severity (Fig. 5a). Similarly, sCD40L was also elevated in patients with CVD and CVD+CKD compared to healthy subjects (Fig. 5b). CD40+ intermediated MC subset was elevated in patients with CVD and CVD+CKD compared with healthy subjects and increased with the elevation of CKD severity (Fig. 5c). However, intermediate MC subset was elevated in CVD patients but not further increased in CVD+CKD patients (Fig. 5d). Future studies will further analyze the relationship of CD40+ MC with different subtypes of CKD; such studies should allow us to better define CD40+ MC as a diagnosis and prognosis biomarker for CKD.

Conclusion

A novel metabolic response was incorporated into the immune system framework, providing an extensive overview of current knowledge in immune checkpoint theory (Fig. 6). This metabolic response is a novel MADS recognition pattern, mediating metabolic RF-induced innate and adaptive immune response. We propose the MADS recognition as signal 4 in adaptive immunity. MADS recognition induces immune checkpoint molecule expression via metabolic sensor leading to amplification of signal 2 two-way stimulatory immune checkpoint amplification. The forward immune checkpoint leads to TC activation. The reverse immune checkpoint leads to APC activation. Metabolic RF, such as uremic toxin or HHcy, was demonstrated to induce CD40 expression in MC and to elevate circulating sCD40L resulting in CD40+ MC differentiation via metabolic sensor. We defined CD40+ MC as a novel and stronger pro-inflammatory MC subset, compared with intermediate MC, and a reliable biomarker for CKD severity. Our studies supported the notion that MADS recognition amplify stimulatory immune checkpoint leading to TC activation and APC inflammation, respectively, which results in systemic and tissue inflammation. Furthermore, we propose the CD40:CD40L immune checkpoint as a therapeutic target for metabolic disease, CVD, and cancer.

Acknowledgements

Not applicable.

Funding

This work was supported in part by the following National Institutes of Health (NIH) grant numbers: HL82774, HL110764, HL117654, DK104116, HL131460, HL130233, and DK113775 (HW); HL9445, HL108910, and HL116917 (XFY), HL131460 (HW/EC/XFY), by American Heart Association (AHA) grant 17SDG33671051 (PF), by the National Science Foundation of China grant number 81200208 (JD), by the Medical and Health Technology Plan of Zhejiang Province grant number 2017KY508 (JD), and by the National Science Foundation of China grant numbers 81330004 and 91639204 (JY).

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Yang XF, Yin Y, Wang H. Vascular inflammation and atherogenesis are activated via receptors for PAMPs and suppressed by regulatory T cells. Drug Discov Today Ther Strateg. 2008;5(2):125–42.PubMedPubMedCentralCrossRef Yang XF, Yin Y, Wang H. Vascular inflammation and atherogenesis are activated via receptors for PAMPs and suppressed by regulatory T cells. Drug Discov Today Ther Strateg. 2008;5(2):125–42.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Peng G, Guo Z, Kiniwa Y, Voo KS, Peng W, Fu T, Wang DY, Li Y, Wang HY, Wang RF. Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science. 2005;309(5739):1380–4.PubMedCrossRef Peng G, Guo Z, Kiniwa Y, Voo KS, Peng W, Fu T, Wang DY, Li Y, Wang HY, Wang RF. Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science. 2005;309(5739):1380–4.PubMedCrossRef
3.
Zurück zum Zitat Gerriets VA, Kishton RJ, Johnson MO, Cohen S, Siska PJ, Nichols AG, Warmoes MO, de Cubas AA, MacIver NJ, Locasale JW, et al. Foxp3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression. Nat Immunol. 2016;17(12):1459–66.PubMedCrossRef Gerriets VA, Kishton RJ, Johnson MO, Cohen S, Siska PJ, Nichols AG, Warmoes MO, de Cubas AA, MacIver NJ, Locasale JW, et al. Foxp3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression. Nat Immunol. 2016;17(12):1459–66.PubMedCrossRef
4.
Zurück zum Zitat Hansson GK, Libby P, Schonbeck U, Yan ZQ. Innate and adaptive immunity in the pathogenesis of atherosclerosis. Circ Res. 2002;91(4):281–91.PubMedCrossRef Hansson GK, Libby P, Schonbeck U, Yan ZQ. Innate and adaptive immunity in the pathogenesis of atherosclerosis. Circ Res. 2002;91(4):281–91.PubMedCrossRef
5.
Zurück zum Zitat Jenkins MK, Taylor PS, Norton SD, Urdahl KB. CD28 delivers a costimulatory signal involved in antigen-specific IL-2 production by human T cells. J Immunol. 1991;147(8):2461–6.PubMed Jenkins MK, Taylor PS, Norton SD, Urdahl KB. CD28 delivers a costimulatory signal involved in antigen-specific IL-2 production by human T cells. J Immunol. 1991;147(8):2461–6.PubMed
6.
Zurück zum Zitat Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H. Interleukin-18 is a unique cytokine that stimulates both Th1 and Th2 responses depending on its cytokine milieu. Cytokine Growth Factor Rev. 2001;12(1):53–72.PubMedCrossRef Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H. Interleukin-18 is a unique cytokine that stimulates both Th1 and Th2 responses depending on its cytokine milieu. Cytokine Growth Factor Rev. 2001;12(1):53–72.PubMedCrossRef
7.
Zurück zum Zitat Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455–65.PubMedPubMedCentralCrossRef Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455–65.PubMedPubMedCentralCrossRef
8.
9.
Zurück zum Zitat Ma W, Gilligan BM, Yuan J, Li T. Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy. J Hematol Oncol. 2016;9(1):47.PubMedPubMedCentralCrossRef Ma W, Gilligan BM, Yuan J, Li T. Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy. J Hematol Oncol. 2016;9(1):47.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Wang J, Yuan R, Song W, Sun J, Liu D, Li Z. PD-1, PD-L1 (B7-H1) and tumor-site immune modulation therapy: the historical perspective. J Hematol Oncol. 2017;10(1):34.PubMedPubMedCentralCrossRef Wang J, Yuan R, Song W, Sun J, Liu D, Li Z. PD-1, PD-L1 (B7-H1) and tumor-site immune modulation therapy: the historical perspective. J Hematol Oncol. 2017;10(1):34.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Ceeraz S, Nowak EC, Burns CM, Noelle RJ. Immune checkpoint receptors in regulating immune reactivity in rheumatic disease. Arthritis Res Ther. 2014;16(5):469.PubMedPubMedCentralCrossRef Ceeraz S, Nowak EC, Burns CM, Noelle RJ. Immune checkpoint receptors in regulating immune reactivity in rheumatic disease. Arthritis Res Ther. 2014;16(5):469.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol. 2007;8(9):942–9.PubMedCrossRef Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol. 2007;8(9):942–9.PubMedCrossRef
14.
Zurück zum Zitat Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27(4):450–61.PubMedPubMedCentralCrossRef Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27(4):450–61.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Yang J, Fang P, Yu D, Zhang L, Zhang D, Jiang X, Yang WY, Bottiglieri T, Kunapuli SP, Yu J, et al. Chronic kidney disease induces inflammatory CD40+ monocyte differentiation via homocysteine elevation and DNA hypomethylation. Circ Res. 2016;119(11):1226–41.PubMedCrossRef Yang J, Fang P, Yu D, Zhang L, Zhang D, Jiang X, Yang WY, Bottiglieri T, Kunapuli SP, Yu J, et al. Chronic kidney disease induces inflammatory CD40+ monocyte differentiation via homocysteine elevation and DNA hypomethylation. Circ Res. 2016;119(11):1226–41.PubMedCrossRef
16.
Zurück zum Zitat Xi H, Zhang Y, Xu Y, Yang WY, Jiang X, Sha X, Cheng X, Wang J, Qin X, Yu J, et al. Caspase-1 inflammasome activation mediates homocysteine-induced pyrop-apoptosis in endothelial cells. Circ Res. 2016;118(10):1525–39.PubMedPubMedCentralCrossRef Xi H, Zhang Y, Xu Y, Yang WY, Jiang X, Sha X, Cheng X, Wang J, Qin X, Yu J, et al. Caspase-1 inflammasome activation mediates homocysteine-induced pyrop-apoptosis in endothelial cells. Circ Res. 2016;118(10):1525–39.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Zhang D, Jiang X, Fang P, Yan Y, Song J, Gupta S, Schafer AI, Durante W, Kruger WD, Yang X, et al. Hyperhomocysteinemia promotes inflammatory monocyte generation and accelerates atherosclerosis in transgenic cystathionine beta-synthase-deficient mice. Circulation. 2009;120(19):1893–902.PubMedPubMedCentralCrossRef Zhang D, Jiang X, Fang P, Yan Y, Song J, Gupta S, Schafer AI, Durante W, Kruger WD, Yang X, et al. Hyperhomocysteinemia promotes inflammatory monocyte generation and accelerates atherosclerosis in transgenic cystathionine beta-synthase-deficient mice. Circulation. 2009;120(19):1893–902.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Zhang D, Fang P, Jiang X, Nelson J, Moore JK, Kruger WD, Berretta RM, Houser SR, Yang X, Wang H. Severe hyperhomocysteinemia promotes bone marrow-derived and resident inflammatory monocyte differentiation and atherosclerosis in LDLr/CBS-deficient mice. Circ Res. 2012;111(1):37–49.PubMedPubMedCentralCrossRef Zhang D, Fang P, Jiang X, Nelson J, Moore JK, Kruger WD, Berretta RM, Houser SR, Yang X, Wang H. Severe hyperhomocysteinemia promotes bone marrow-derived and resident inflammatory monocyte differentiation and atherosclerosis in LDLr/CBS-deficient mice. Circ Res. 2012;111(1):37–49.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Fang P, Zhang D, Cheng Z, Yan C, Jiang X, Kruger WD, Meng S, Arning E, Bottiglieri T, Choi ET, et al. Hyperhomocysteinemia potentiates hyperglycemia-induced inflammatory monocyte differentiation and atherosclerosis. Diabetes. 2014;63(12):4275–90.PubMedPubMedCentralCrossRef Fang P, Zhang D, Cheng Z, Yan C, Jiang X, Kruger WD, Meng S, Arning E, Bottiglieri T, Choi ET, et al. Hyperhomocysteinemia potentiates hyperglycemia-induced inflammatory monocyte differentiation and atherosclerosis. Diabetes. 2014;63(12):4275–90.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Mai J, Virtue A, Shen J, Wang H, Yang XF. An evolving new paradigm: endothelial cells–conditional innate immune cells. J Hematol Oncol. 2013;6:61.PubMedPubMedCentralCrossRef Mai J, Virtue A, Shen J, Wang H, Yang XF. An evolving new paradigm: endothelial cells–conditional innate immune cells. J Hematol Oncol. 2013;6:61.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Li YF, Ren LN, Guo G, Cannella LA, Chernaya V, Samuel S, Liu SX, Wang H, Yang XF. Endothelial progenitor cells in ischemic stroke: an exploration from hypothesis to therapy. J Hematol Oncol. 2015;8:33.PubMedPubMedCentralCrossRef Li YF, Ren LN, Guo G, Cannella LA, Chernaya V, Samuel S, Liu SX, Wang H, Yang XF. Endothelial progenitor cells in ischemic stroke: an exploration from hypothesis to therapy. J Hematol Oncol. 2015;8:33.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Wang X, Li YF, Nanayakkara G, Shao Y, Liang B, Cole L, Yang WY, Li X, Cueto R, Yu J, et al. Lysophospholipid receptors, as novel conditional danger receptors and homeostatic receptors modulate inflammation-novel paradigm and therapeutic potential. J Cardiovasc Transl Res. 2016;9(4):343–59.PubMedCrossRef Wang X, Li YF, Nanayakkara G, Shao Y, Liang B, Cole L, Yang WY, Li X, Cueto R, Yu J, et al. Lysophospholipid receptors, as novel conditional danger receptors and homeostatic receptors modulate inflammation-novel paradigm and therapeutic potential. J Cardiovasc Transl Res. 2016;9(4):343–59.PubMedCrossRef
23.
Zurück zum Zitat Liu W, Yin Y, Zhou Z, He M, Dai Y. OxLDL-induced IL-1 beta secretion promoting foam cells formation was mainly via CD36 mediated ROS production leading to NLRP3 inflammasome activation. Inflammation research: official journal of the European Histamine Research Society [et al]. 2014;63(1):33–43.CrossRef Liu W, Yin Y, Zhou Z, He M, Dai Y. OxLDL-induced IL-1 beta secretion promoting foam cells formation was mainly via CD36 mediated ROS production leading to NLRP3 inflammasome activation. Inflammation research: official journal of the European Histamine Research Society [et al]. 2014;63(1):33–43.CrossRef
24.
Zurück zum Zitat Wang L, Fu H, Nanayakkara G, Li Y, Shao Y, Johnson C, Cheng J, Yang WY, Yang F, Lavallee M, et al. Novel extracellular and nuclear caspase-1 and inflammasomes propagate inflammation and regulate gene expression: a comprehensive database mining study. J Hematol Oncol. 2016;9(1):122.PubMedPubMedCentralCrossRef Wang L, Fu H, Nanayakkara G, Li Y, Shao Y, Johnson C, Cheng J, Yang WY, Yang F, Lavallee M, et al. Novel extracellular and nuclear caspase-1 and inflammasomes propagate inflammation and regulate gene expression: a comprehensive database mining study. J Hematol Oncol. 2016;9(1):122.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Li X, Wang S, Zhu R, Li H, Han Q, Zhao RC. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFkappaB-TLR signaling pathway. J Hematol Oncol. 2016;9:42.PubMedPubMedCentralCrossRef Li X, Wang S, Zhu R, Li H, Han Q, Zhao RC. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFkappaB-TLR signaling pathway. J Hematol Oncol. 2016;9:42.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Sutton CE, Lalor SJ, Sweeney CM, Brereton CF, Lavelle EC, Mills KH. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity. 2009;31(2):331–41.PubMedCrossRef Sutton CE, Lalor SJ, Sweeney CM, Brereton CF, Lavelle EC, Mills KH. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity. 2009;31(2):331–41.PubMedCrossRef
28.
Zurück zum Zitat Goplen NP, Saxena V, Knudson KM, Schrum AG, Gil D, Daniels MA, Zamoyska R, Teixeiro E. IL-12 signals through the TCR to support CD8 innate immune responses. J Immunol. 2016;197(6):2434–43.PubMedCrossRef Goplen NP, Saxena V, Knudson KM, Schrum AG, Gil D, Daniels MA, Zamoyska R, Teixeiro E. IL-12 signals through the TCR to support CD8 innate immune responses. J Immunol. 2016;197(6):2434–43.PubMedCrossRef
29.
Zurück zum Zitat Kim HK, Falugi F, Missiakas DM, Schneewind O. Peptidoglycan-linked protein A promotes T cell-dependent antibody expansion during Staphylococcus aureus infection. Proc Natl Acad Sci U S A. 2016;113(20):5718–23.PubMedPubMedCentralCrossRef Kim HK, Falugi F, Missiakas DM, Schneewind O. Peptidoglycan-linked protein A promotes T cell-dependent antibody expansion during Staphylococcus aureus infection. Proc Natl Acad Sci U S A. 2016;113(20):5718–23.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Secatto A, Rodrigues LC, Serezani CH, Ramos SG, Dias-Baruffi M, Faccioli LH, Medeiros AI. 5-Lipoxygenase deficiency impairs innate and adaptive immune responses during fungal infection. PLoS One. 2012;7(3):e31701.PubMedPubMedCentralCrossRef Secatto A, Rodrigues LC, Serezani CH, Ramos SG, Dias-Baruffi M, Faccioli LH, Medeiros AI. 5-Lipoxygenase deficiency impairs innate and adaptive immune responses during fungal infection. PLoS One. 2012;7(3):e31701.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Kolumam GA, Thomas S, Thompson LJ, Sprent J, Murali-Krishna K. Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection. J Exp Med. 2005;202(5):637–50.PubMedPubMedCentralCrossRef Kolumam GA, Thomas S, Thompson LJ, Sprent J, Murali-Krishna K. Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection. J Exp Med. 2005;202(5):637–50.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Flemming A. T cells: successful checkpoint blockade requires positive co-stimulation. Nat Rev Immunol. 2017;17(4):215.PubMedCrossRef Flemming A. T cells: successful checkpoint blockade requires positive co-stimulation. Nat Rev Immunol. 2017;17(4):215.PubMedCrossRef
34.
Zurück zum Zitat Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28(19):3167–75.PubMedPubMedCentralCrossRef Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28(19):3167–75.PubMedPubMedCentralCrossRef
35.
37.
Zurück zum Zitat Ben-Sasson SZ, Hu-Li J, Quiel J, Cauchetaux S, Ratner M, Shapira I, Dinarello CA, Paul WE. IL-1 acts directly on CD4 T cells to enhance their antigen-driven expansion and differentiation. Proc Natl Acad Sci U S A. 2009;106(17):7119–24.PubMedPubMedCentralCrossRef Ben-Sasson SZ, Hu-Li J, Quiel J, Cauchetaux S, Ratner M, Shapira I, Dinarello CA, Paul WE. IL-1 acts directly on CD4 T cells to enhance their antigen-driven expansion and differentiation. Proc Natl Acad Sci U S A. 2009;106(17):7119–24.PubMedPubMedCentralCrossRef
38.
39.
Zurück zum Zitat Gateva A, Assyov Y, Tsakova A, Kamenov Z. Soluble CD40L is associated with insulin resistance, but not with glucose tolerance in obese nondiabetic patients. Arch Physiol Biochem. 2016;122(3):161–5.PubMedCrossRef Gateva A, Assyov Y, Tsakova A, Kamenov Z. Soluble CD40L is associated with insulin resistance, but not with glucose tolerance in obese nondiabetic patients. Arch Physiol Biochem. 2016;122(3):161–5.PubMedCrossRef
40.
Zurück zum Zitat Palmer CS, Ostrowski M, Balderson B, Christian N, Crowe SM. Glucose metabolism regulates T cell activation, differentiation, and functions. Front Immunol. 2015;6:1.PubMedPubMedCentralCrossRef Palmer CS, Ostrowski M, Balderson B, Christian N, Crowe SM. Glucose metabolism regulates T cell activation, differentiation, and functions. Front Immunol. 2015;6:1.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Gauld SB, Merrell KT, Cambier JC. Silencing of autoreactive B cells by anergy: a fresh perspective. Curr Opin Immunol. 2006;18(3):292–7.PubMedCrossRef Gauld SB, Merrell KT, Cambier JC. Silencing of autoreactive B cells by anergy: a fresh perspective. Curr Opin Immunol. 2006;18(3):292–7.PubMedCrossRef
43.
Zurück zum Zitat Schonbeck U, Gerdes N, Varo N, Reynolds RS, Horton DB, Bavendiek U, Robbie L, Ganz P, Kinlay S, Libby P. Oxidized low-density lipoprotein augments and 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors limit CD40 and CD40L expression in human vascular cells. Circulation. 2002;106(23):2888–93.PubMedCrossRef Schonbeck U, Gerdes N, Varo N, Reynolds RS, Horton DB, Bavendiek U, Robbie L, Ganz P, Kinlay S, Libby P. Oxidized low-density lipoprotein augments and 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors limit CD40 and CD40L expression in human vascular cells. Circulation. 2002;106(23):2888–93.PubMedCrossRef
44.
Zurück zum Zitat Adam D, Heinrich M, Kabelitz D, Schutze S. Ceramide: does it matter for T cells? Trends Immunol. 2002;23(1):1–4.PubMedCrossRef Adam D, Heinrich M, Kabelitz D, Schutze S. Ceramide: does it matter for T cells? Trends Immunol. 2002;23(1):1–4.PubMedCrossRef
45.
Zurück zum Zitat Kurtulus S, Sakuishi K, Ngiow SF, Joller N, Tan DJ, Teng MW, Smyth MJ, Kuchroo VK, Anderson AC. TIGIT predominantly regulates the immune response via regulatory T cells. J Clin Invest. 2015;125(11):4053–62.PubMedPubMedCentralCrossRef Kurtulus S, Sakuishi K, Ngiow SF, Joller N, Tan DJ, Teng MW, Smyth MJ, Kuchroo VK, Anderson AC. TIGIT predominantly regulates the immune response via regulatory T cells. J Clin Invest. 2015;125(11):4053–62.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y, Noelle RJ. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol Rev. 2009;229(1):152–72.PubMedCrossRef Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y, Noelle RJ. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol Rev. 2009;229(1):152–72.PubMedCrossRef
47.
Zurück zum Zitat Lutgens E, Lievens D, Beckers L, Wijnands E, Soehnlein O, Zernecke A, Seijkens T, Engel D, Cleutjens J, Keller AM, et al. Deficient CD40-TRAF6 signaling in leukocytes prevents atherosclerosis by skewing the immune response toward an antiinflammatory profile. J Exp Med. 2010;207(2):391–404.PubMedPubMedCentralCrossRef Lutgens E, Lievens D, Beckers L, Wijnands E, Soehnlein O, Zernecke A, Seijkens T, Engel D, Cleutjens J, Keller AM, et al. Deficient CD40-TRAF6 signaling in leukocytes prevents atherosclerosis by skewing the immune response toward an antiinflammatory profile. J Exp Med. 2010;207(2):391–404.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Hill A, Chapel H. X-linked immunodeficiency. The fruits of cooperation. Nature. 1993;361(6412):494.PubMedCrossRef Hill A, Chapel H. X-linked immunodeficiency. The fruits of cooperation. Nature. 1993;361(6412):494.PubMedCrossRef
49.
Zurück zum Zitat Molnar E, Swamy M, Holzer M, Beck-Garcia K, Worch R, Thiele C, Guigas G, Boye K, Luescher IF, Schwille P, et al. Cholesterol and sphingomyelin drive ligand-independent T-cell antigen receptor nanoclustering. J Biol Chem. 2012;287(51):42664–74.PubMedPubMedCentralCrossRef Molnar E, Swamy M, Holzer M, Beck-Garcia K, Worch R, Thiele C, Guigas G, Boye K, Luescher IF, Schwille P, et al. Cholesterol and sphingomyelin drive ligand-independent T-cell antigen receptor nanoclustering. J Biol Chem. 2012;287(51):42664–74.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Ostrand-Rosenberg S, Horn LA, Haile ST. The programmed death-1 immune-suppressive pathway: barrier to antitumor immunity. J Immunol. 2014;193(8):3835–41.PubMedPubMedCentralCrossRef Ostrand-Rosenberg S, Horn LA, Haile ST. The programmed death-1 immune-suppressive pathway: barrier to antitumor immunity. J Immunol. 2014;193(8):3835–41.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Wang F, Beck-Garcia K, Zorzin C, Schamel WW, Davis MM. Inhibition of T cell receptor signaling by cholesterol sulfate, a naturally occurring derivative of membrane cholesterol. Nat Immunol. 2016;17(7):844–50.PubMedPubMedCentralCrossRef Wang F, Beck-Garcia K, Zorzin C, Schamel WW, Davis MM. Inhibition of T cell receptor signaling by cholesterol sulfate, a naturally occurring derivative of membrane cholesterol. Nat Immunol. 2016;17(7):844–50.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Williams ML, Rutherford SL, Feingold KR. Effects of cholesterol sulfate on lipid metabolism in cultured human keratinocytes and fibroblasts. J Lipid Res. 1987;28(8):955–67.PubMed Williams ML, Rutherford SL, Feingold KR. Effects of cholesterol sulfate on lipid metabolism in cultured human keratinocytes and fibroblasts. J Lipid Res. 1987;28(8):955–67.PubMed
55.
Zurück zum Zitat Croft M, Duan W, Choi H, Eun SY, Madireddi S, Mehta A. TNF superfamily in inflammatory disease: translating basic insights. Trends Immunol. 2012;33(3):144–52.PubMedCrossRef Croft M, Duan W, Choi H, Eun SY, Madireddi S, Mehta A. TNF superfamily in inflammatory disease: translating basic insights. Trends Immunol. 2012;33(3):144–52.PubMedCrossRef
56.
Zurück zum Zitat Sica GL, Zhu G, Tamada K, Liu D, Ni J, Chen L. RELT, a new member of the tumor necrosis factor receptor superfamily, is selectively expressed in hematopoietic tissues and activates transcription factor NF-kappaB. Blood. 2001;97(9):2702–7.PubMedCrossRef Sica GL, Zhu G, Tamada K, Liu D, Ni J, Chen L. RELT, a new member of the tumor necrosis factor receptor superfamily, is selectively expressed in hematopoietic tissues and activates transcription factor NF-kappaB. Blood. 2001;97(9):2702–7.PubMedCrossRef
57.
Zurück zum Zitat Jansen MF, Hollander MR, van Royen N, Horrevoets AJ, Lutgens E. CD40 in coronary artery disease: a matter of macrophages? Basic Res Cardiol. 2016;111(4):38.PubMedPubMedCentralCrossRef Jansen MF, Hollander MR, van Royen N, Horrevoets AJ, Lutgens E. CD40 in coronary artery disease: a matter of macrophages? Basic Res Cardiol. 2016;111(4):38.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Rizvi M, Pathak D, Freedman JE, Chakrabarti S. CD40-CD40 ligand interactions in oxidative stress, inflammation and vascular disease. Trends Mol Med. 2008;14(12):530–8.PubMedCrossRef Rizvi M, Pathak D, Freedman JE, Chakrabarti S. CD40-CD40 ligand interactions in oxidative stress, inflammation and vascular disease. Trends Mol Med. 2008;14(12):530–8.PubMedCrossRef
59.
Zurück zum Zitat Andre P, Nannizzi-Alaimo L, Prasad SK, Phillips DR. Platelet-derived CD40L: the switch-hitting player of cardiovascular disease. Circulation. 2002;106(8):896–9.PubMedCrossRef Andre P, Nannizzi-Alaimo L, Prasad SK, Phillips DR. Platelet-derived CD40L: the switch-hitting player of cardiovascular disease. Circulation. 2002;106(8):896–9.PubMedCrossRef
60.
Zurück zum Zitat Schiza A, Wenthe J, Mangsbo S, Eriksson E, Nilsson A, Totterman TH, Loskog A, Ullenhag G. Adenovirus-mediated CD40L gene transfer increases Teffector/Tregulatory cell ratio and upregulates death receptors in metastatic melanoma patients. J Transl Med. 2017;15(1):79.PubMedPubMedCentralCrossRef Schiza A, Wenthe J, Mangsbo S, Eriksson E, Nilsson A, Totterman TH, Loskog A, Ullenhag G. Adenovirus-mediated CD40L gene transfer increases Teffector/Tregulatory cell ratio and upregulates death receptors in metastatic melanoma patients. J Transl Med. 2017;15(1):79.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Ngiow SF, Young A, Blake SJ, Hill GR, Yagita H, Teng MW, Korman AJ, Smyth MJ. Agonistic CD40 mAb-driven IL12 reverses resistance to anti-PD1 in a T-cell-rich tumor. Cancer Res. 2016;76(21):6266–77.PubMedCrossRef Ngiow SF, Young A, Blake SJ, Hill GR, Yagita H, Teng MW, Korman AJ, Smyth MJ. Agonistic CD40 mAb-driven IL12 reverses resistance to anti-PD1 in a T-cell-rich tumor. Cancer Res. 2016;76(21):6266–77.PubMedCrossRef
63.
Zurück zum Zitat Helseth R, Weiss TW, Opstad TB, Siegbahn A, Solheim S, Freynhofer MK, Huber K, Arnesen H, Seljeflot S. Associations between circulating proteins and corresponding genes expressed in coronary thrombi in patients with acute myocardial infarction. Thromb Res. 2015;136(6):1240–4.PubMedCrossRef Helseth R, Weiss TW, Opstad TB, Siegbahn A, Solheim S, Freynhofer MK, Huber K, Arnesen H, Seljeflot S. Associations between circulating proteins and corresponding genes expressed in coronary thrombi in patients with acute myocardial infarction. Thromb Res. 2015;136(6):1240–4.PubMedCrossRef
64.
Zurück zum Zitat Lutgens E, Gorelik L, Daemen MJ, de Muinck ED, Grewal IS, Koteliansky VE, Flavell RA. Requirement for CD154 in the progression of atherosclerosis. Nat Med. 1999;5(11):1313–6.PubMedCrossRef Lutgens E, Gorelik L, Daemen MJ, de Muinck ED, Grewal IS, Koteliansky VE, Flavell RA. Requirement for CD154 in the progression of atherosclerosis. Nat Med. 1999;5(11):1313–6.PubMedCrossRef
65.
Zurück zum Zitat Lutgens E, Cleutjens KB, Heeneman S, Koteliansky VE, Burkly LC, Daemen MJ. Both early and delayed anti-CD40L antibody treatment induces a stable plaque phenotype. Proc Natl Acad Sci U S A. 2000;97(13):7464–9.PubMedPubMedCentralCrossRef Lutgens E, Cleutjens KB, Heeneman S, Koteliansky VE, Burkly LC, Daemen MJ. Both early and delayed anti-CD40L antibody treatment induces a stable plaque phenotype. Proc Natl Acad Sci U S A. 2000;97(13):7464–9.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Wang B, Qian H, Yang H, Xu L, Xu W, Yan J. Regression of atherosclerosis plaques in apolipoprotein E-/- mice after lentivirus-mediated RNA interference of CD40. Int J Cardiol. 2013;163(1):34–9.PubMedCrossRef Wang B, Qian H, Yang H, Xu L, Xu W, Yan J. Regression of atherosclerosis plaques in apolipoprotein E-/- mice after lentivirus-mediated RNA interference of CD40. Int J Cardiol. 2013;163(1):34–9.PubMedCrossRef
67.
Zurück zum Zitat Mach F, Schonbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature. 1998;394(6689):200–3.PubMedCrossRef Mach F, Schonbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature. 1998;394(6689):200–3.PubMedCrossRef
68.
Zurück zum Zitat Slawek A, Maj T, Chelmonska-Soyta A. CD40, CD80, and CD86 costimulatory molecules are differentially expressed on murine splenic antigen-presenting cells during the pre-implantation period of pregnancy, and they modulate regulatory T-cell abundance, peripheral cytokine response, and pregnancy outcome. Am J Reprod Immunol. 2013;70(2):116–26.PubMedCrossRef Slawek A, Maj T, Chelmonska-Soyta A. CD40, CD80, and CD86 costimulatory molecules are differentially expressed on murine splenic antigen-presenting cells during the pre-implantation period of pregnancy, and they modulate regulatory T-cell abundance, peripheral cytokine response, and pregnancy outcome. Am J Reprod Immunol. 2013;70(2):116–26.PubMedCrossRef
69.
Zurück zum Zitat Zhang Y, Hu X, Hu Y, Teng K, Zhang K, Zheng Y, Hong X, Yu K, Wang Y, Liu L. Anti-CD40-induced inflammatory E-cadherin+ dendritic cells enhance T cell responses and antitumour immunity in murine Lewis lung carcinoma. J Exp Clin Cancer Res. 2015;34:11.PubMedPubMedCentralCrossRef Zhang Y, Hu X, Hu Y, Teng K, Zhang K, Zheng Y, Hong X, Yu K, Wang Y, Liu L. Anti-CD40-induced inflammatory E-cadherin+ dendritic cells enhance T cell responses and antitumour immunity in murine Lewis lung carcinoma. J Exp Clin Cancer Res. 2015;34:11.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Dopheide JF, Sester U, Schlitt A, Horstick G, Rupprecht HJ, Munzel T, Blankenberg S. Monocyte-derived dendritic cells of patients with coronary artery disease show an increased expression of costimulatory molecules CD40, CD80 and CD86 in vitro. Coron Artery Dis. 2007;18(7):523–31.PubMedCrossRef Dopheide JF, Sester U, Schlitt A, Horstick G, Rupprecht HJ, Munzel T, Blankenberg S. Monocyte-derived dendritic cells of patients with coronary artery disease show an increased expression of costimulatory molecules CD40, CD80 and CD86 in vitro. Coron Artery Dis. 2007;18(7):523–31.PubMedCrossRef
72.
73.
Zurück zum Zitat Yin Y, Pastrana JL, Li X, Huang X, Mallilankaraman K, Choi ET, Madesh M, Wang H, Yang XF. Inflammasomes: sensors of metabolic stresses for vascular inflammation. Front Biosci (Landmark Ed). 2013;18:638–49.CrossRef Yin Y, Pastrana JL, Li X, Huang X, Mallilankaraman K, Choi ET, Madesh M, Wang H, Yang XF. Inflammasomes: sensors of metabolic stresses for vascular inflammation. Front Biosci (Landmark Ed). 2013;18:638–49.CrossRef
74.
Zurück zum Zitat Yu Y, He Y, Yang TT, Jiang H, Xiang YJ, Fang LB, Hjelmstrom P, Gao XG, Liu GZ. Elevated plasma levels and monocyte-associated expression of CD137 ligand in patients with acute atherothrombotic stroke. Eur Rev Med Pharmacol Sci. 2014;18(10):1525–32.PubMed Yu Y, He Y, Yang TT, Jiang H, Xiang YJ, Fang LB, Hjelmstrom P, Gao XG, Liu GZ. Elevated plasma levels and monocyte-associated expression of CD137 ligand in patients with acute atherothrombotic stroke. Eur Rev Med Pharmacol Sci. 2014;18(10):1525–32.PubMed
75.
Zurück zum Zitat Jeon HJ, Choi JH, Jung IH, Park JG, Lee MR, Lee MN, Kim B, Yoo JY, Jeong SJ, Kim DY, et al. CD137 (4-1BB) deficiency reduces atherosclerosis in hyperlipidemic mice. Circulation. 2010;121(9):1124–33.PubMedCrossRef Jeon HJ, Choi JH, Jung IH, Park JG, Lee MR, Lee MN, Kim B, Yoo JY, Jeong SJ, Kim DY, et al. CD137 (4-1BB) deficiency reduces atherosclerosis in hyperlipidemic mice. Circulation. 2010;121(9):1124–33.PubMedCrossRef
76.
Zurück zum Zitat Olofsson PS, Soderstrom LA, Wagsater D, Sheikine Y, Ocaya P, Lang F, Rabu C, Chen L, Rudling M, Aukrust P, et al. CD137 is expressed in human atherosclerosis and promotes development of plaque inflammation in hypercholesterolemic mice. Circulation. 2008;117(10):1292–301.PubMedCrossRef Olofsson PS, Soderstrom LA, Wagsater D, Sheikine Y, Ocaya P, Lang F, Rabu C, Chen L, Rudling M, Aukrust P, et al. CD137 is expressed in human atherosclerosis and promotes development of plaque inflammation in hypercholesterolemic mice. Circulation. 2008;117(10):1292–301.PubMedCrossRef
77.
Zurück zum Zitat Dumitriu IE, Baruah P, Finlayson CJ, Loftus IM, Antunes RF, Lim P, Bunce N, Kaski JC. High levels of costimulatory receptors OX40 and 4-1BB characterize CD4+CD28null T cells in patients with acute coronary syndrome. Circ Res. 2012;110(6):857–69.PubMedCrossRef Dumitriu IE, Baruah P, Finlayson CJ, Loftus IM, Antunes RF, Lim P, Bunce N, Kaski JC. High levels of costimulatory receptors OX40 and 4-1BB characterize CD4+CD28null T cells in patients with acute coronary syndrome. Circ Res. 2012;110(6):857–69.PubMedCrossRef
78.
Zurück zum Zitat Srivastava RM, Trivedi S, Concha-Benavente F, Gibson SP, Reeder C, Ferrone S, Ferris RL. CD137 stimulation enhances cetuximab-induced natural killer: dendritic cell priming of antitumor T-cell immunity in patients with head and neck cancer. Clin Cancer Res. 2017;23(3):707–16.PubMedCrossRef Srivastava RM, Trivedi S, Concha-Benavente F, Gibson SP, Reeder C, Ferrone S, Ferris RL. CD137 stimulation enhances cetuximab-induced natural killer: dendritic cell priming of antitumor T-cell immunity in patients with head and neck cancer. Clin Cancer Res. 2017;23(3):707–16.PubMedCrossRef
79.
Zurück zum Zitat Ju S, Ge Y, Qiu H, Lu B, Qiu Y, Fu J, Liu G, Wang Q, Hu Y, Shu Y, et al. A novel approach to induce human DCs from monocytes by triggering 4-1BBL reverse signaling. Int Immunol. 2009;21(10):1135–44.PubMedCrossRef Ju S, Ge Y, Qiu H, Lu B, Qiu Y, Fu J, Liu G, Wang Q, Hu Y, Shu Y, et al. A novel approach to induce human DCs from monocytes by triggering 4-1BBL reverse signaling. Int Immunol. 2009;21(10):1135–44.PubMedCrossRef
80.
Zurück zum Zitat Jung IH, Choi JH, Jin J, Jeong SJ, Jeon S, Lim C, Lee MR, Yoo JY, Sonn SK, Kim YH, et al. CD137-inducing factors from T cells and macrophages accelerate the destabilization of atherosclerotic plaques in hyperlipidemic mice. FASEB J. 2014;28(11):4779–91.PubMedCrossRef Jung IH, Choi JH, Jin J, Jeong SJ, Jeon S, Lim C, Lee MR, Yoo JY, Sonn SK, Kim YH, et al. CD137-inducing factors from T cells and macrophages accelerate the destabilization of atherosclerotic plaques in hyperlipidemic mice. FASEB J. 2014;28(11):4779–91.PubMedCrossRef
81.
Zurück zum Zitat Gauttier V, Judor JP, Le Guen V, Cany J, Ferry N, Conchon S. Agonistic anti-CD137 antibody treatment leads to antitumor response in mice with liver cancer. Int J Cancer. 2014;135(12):2857–67.PubMedCrossRef Gauttier V, Judor JP, Le Guen V, Cany J, Ferry N, Conchon S. Agonistic anti-CD137 antibody treatment leads to antitumor response in mice with liver cancer. Int J Cancer. 2014;135(12):2857–67.PubMedCrossRef
82.
Zurück zum Zitat Webb GJ, Hirschfield GM, Lane PJ. OX40, OX40L and autoimmunity: a comprehensive review. Clin Rev Allergy Immunol. 2016;50(3):312–32.PubMedCrossRef Webb GJ, Hirschfield GM, Lane PJ. OX40, OX40L and autoimmunity: a comprehensive review. Clin Rev Allergy Immunol. 2016;50(3):312–32.PubMedCrossRef
83.
Zurück zum Zitat Dongming L, Zuxun L, Liangjie X, Biao W, Ping Y. Enhanced levels of soluble and membrane-bound CD137 levels in patients with acute coronary syndromes. Clin Chim Acta. 2010;411(5-6):406–10.PubMedCrossRef Dongming L, Zuxun L, Liangjie X, Biao W, Ping Y. Enhanced levels of soluble and membrane-bound CD137 levels in patients with acute coronary syndromes. Clin Chim Acta. 2010;411(5-6):406–10.PubMedCrossRef
84.
Zurück zum Zitat Foks AC, van Puijvelde GH, Bot I, ter Borg MN, Habets KL, Johnson JL, Yagita H, van Berkel TJ, Kuiper J. Interruption of the OX40-OX40 ligand pathway in LDL receptor-deficient mice causes regression of atherosclerosis. J Immunol. 2013;191(9):4573–80.PubMedCrossRef Foks AC, van Puijvelde GH, Bot I, ter Borg MN, Habets KL, Johnson JL, Yagita H, van Berkel TJ, Kuiper J. Interruption of the OX40-OX40 ligand pathway in LDL receptor-deficient mice causes regression of atherosclerosis. J Immunol. 2013;191(9):4573–80.PubMedCrossRef
85.
Zurück zum Zitat van Wanrooij EJ, van Puijvelde GH, de Vos P, Yagita H, van Berkel TJ, Kuiper J. Interruption of the Tnfrsf4/Tnfsf4 (OX40/OX40L) pathway attenuates atherogenesis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2007;27(1):204–10.PubMedCrossRef van Wanrooij EJ, van Puijvelde GH, de Vos P, Yagita H, van Berkel TJ, Kuiper J. Interruption of the Tnfrsf4/Tnfsf4 (OX40/OX40L) pathway attenuates atherogenesis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2007;27(1):204–10.PubMedCrossRef
86.
Zurück zum Zitat Vu MD, Xiao X, Gao W, Degauque N, Chen M, Kroemer A, Killeen N, Ishii N, Li XC. OX40 costimulation turns off Foxp3+ Tregs. Blood. 2007;110(7):2501–10.PubMedPubMedCentralCrossRef Vu MD, Xiao X, Gao W, Degauque N, Chen M, Kroemer A, Killeen N, Ishii N, Li XC. OX40 costimulation turns off Foxp3+ Tregs. Blood. 2007;110(7):2501–10.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Yan J, Su H, Xu L, Wang C. OX40-OX40L interaction promotes proliferation and activation of lymphocytes via NFATc1 in ApoE-deficient mice. PLoS One. 2013;8(4):e60854.PubMedPubMedCentralCrossRef Yan J, Su H, Xu L, Wang C. OX40-OX40L interaction promotes proliferation and activation of lymphocytes via NFATc1 in ApoE-deficient mice. PLoS One. 2013;8(4):e60854.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Yu G, Li Y, Cui Z, Morris NP, Weinberg AD, Fox BA, Urba WJ, Wang L, Hu HM. Combinational immunotherapy with Allo-DRibble vaccines and anti-OX40 co-stimulation leads to generation of cross-reactive effector T cells and tumor regression. Sci Rep. 2016;6:37558.PubMedPubMedCentralCrossRef Yu G, Li Y, Cui Z, Morris NP, Weinberg AD, Fox BA, Urba WJ, Wang L, Hu HM. Combinational immunotherapy with Allo-DRibble vaccines and anti-OX40 co-stimulation leads to generation of cross-reactive effector T cells and tumor regression. Sci Rep. 2016;6:37558.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Weixler B, Cremonesi E, Sorge R, Muraro MG, Delko T, Nebiker CA, Daster S, Governa V, Amicarella F, Soysal SD, et al. OX40 expression enhances the prognostic significance of CD8 positive lymphocyte infiltration in colorectal cancer. Oncotarget. 2015;6(35):37588–99.PubMedPubMedCentralCrossRef Weixler B, Cremonesi E, Sorge R, Muraro MG, Delko T, Nebiker CA, Daster S, Governa V, Amicarella F, Soysal SD, et al. OX40 expression enhances the prognostic significance of CD8 positive lymphocyte infiltration in colorectal cancer. Oncotarget. 2015;6(35):37588–99.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Gough MJ, Killeen N, Weinberg AD. Targeting macrophages in the tumour environment to enhance the efficacy of alphaOX40 therapy. Immunology. 2012;136(4):437–47.PubMedPubMedCentralCrossRef Gough MJ, Killeen N, Weinberg AD. Targeting macrophages in the tumour environment to enhance the efficacy of alphaOX40 therapy. Immunology. 2012;136(4):437–47.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Chai ZT, Zhu XD, Ao JY, Wang WQ, Gao DM, Kong J, Zhang N, Zhang YY, Ye BG, Ma DN, et al. microRNA-26a suppresses recruitment of macrophages by down-regulating macrophage colony-stimulating factor expression through the PI3K/Akt pathway in hepatocellular carcinoma. J Hematol Oncol. 2015;8:56.PubMedPubMedCentralCrossRef Chai ZT, Zhu XD, Ao JY, Wang WQ, Gao DM, Kong J, Zhang N, Zhang YY, Ye BG, Ma DN, et al. microRNA-26a suppresses recruitment of macrophages by down-regulating macrophage colony-stimulating factor expression through the PI3K/Akt pathway in hepatocellular carcinoma. J Hematol Oncol. 2015;8:56.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat van de Ven K, Borst J. Targeting the T-cell co-stimulatory CD27/CD70 pathway in cancer immunotherapy: rationale and potential. Immunotherapy. 2015;7(6):655–67.PubMedCrossRef van de Ven K, Borst J. Targeting the T-cell co-stimulatory CD27/CD70 pathway in cancer immunotherapy: rationale and potential. Immunotherapy. 2015;7(6):655–67.PubMedCrossRef
93.
Zurück zum Zitat Winkels H, Meiler S, Smeets E, Lievens D, Engel D, Spitz C, Burger C, Rinne P, Beckers L, Dandl A, et al. CD70 limits atherosclerosis and promotes macrophage function. Thromb Haemost. 2017;117(1):164–75.PubMedCrossRef Winkels H, Meiler S, Smeets E, Lievens D, Engel D, Spitz C, Burger C, Rinne P, Beckers L, Dandl A, et al. CD70 limits atherosclerosis and promotes macrophage function. Thromb Haemost. 2017;117(1):164–75.PubMedCrossRef
94.
Zurück zum Zitat van Olffen RW, de Bruin AM, Vos M, Staniszewska AD, Hamann J, van Lier RA, de Vries CJ, Nolte MA. CD70-driven chronic immune activation is protective against atherosclerosis. J Innate Immun. 2010;2(4):344–52.PubMedCrossRef van Olffen RW, de Bruin AM, Vos M, Staniszewska AD, Hamann J, van Lier RA, de Vries CJ, Nolte MA. CD70-driven chronic immune activation is protective against atherosclerosis. J Innate Immun. 2010;2(4):344–52.PubMedCrossRef
95.
Zurück zum Zitat Sardella G, De Luca L, Francavilla V, Accapezzato D, Mancone M, Sirinian MI, Fedele F, Paroli M. Frequency of naturally-occurring regulatory T cells is reduced in patients with ST-segment elevation myocardial infarction. Thromb Res. 2007;120(4):631–4.PubMedCrossRef Sardella G, De Luca L, Francavilla V, Accapezzato D, Mancone M, Sirinian MI, Fedele F, Paroli M. Frequency of naturally-occurring regulatory T cells is reduced in patients with ST-segment elevation myocardial infarction. Thromb Res. 2007;120(4):631–4.PubMedCrossRef
96.
Zurück zum Zitat Claus C, Riether C, Schurch C, Matter MS, Hilmenyuk T, Ochsenbein AF. CD27 signaling increases the frequency of regulatory T cells and promotes tumor growth. Cancer Res. 2012;72(14):3664–76.PubMedCrossRef Claus C, Riether C, Schurch C, Matter MS, Hilmenyuk T, Ochsenbein AF. CD27 signaling increases the frequency of regulatory T cells and promotes tumor growth. Cancer Res. 2012;72(14):3664–76.PubMedCrossRef
97.
Zurück zum Zitat McEarchern JA, Oflazoglu E, Francisco L, McDonagh CF, Gordon KA, Stone I, Klussman K, Turcott E, van Rooijen N, Carter P, et al. Engineered anti-CD70 antibody with multiple effector functions exhibits in vitro and in vivo antitumor activities. Blood. 2007;109(3):1185–92.PubMedCrossRef McEarchern JA, Oflazoglu E, Francisco L, McDonagh CF, Gordon KA, Stone I, Klussman K, Turcott E, van Rooijen N, Carter P, et al. Engineered anti-CD70 antibody with multiple effector functions exhibits in vitro and in vivo antitumor activities. Blood. 2007;109(3):1185–92.PubMedCrossRef
98.
Zurück zum Zitat Sun M, Fink PJ. A new class of reverse signaling costimulators belongs to the TNF family. J Immunol. 2007;179(7):4307–12.PubMedCrossRef Sun M, Fink PJ. A new class of reverse signaling costimulators belongs to the TNF family. J Immunol. 2007;179(7):4307–12.PubMedCrossRef
99.
Zurück zum Zitat Bartlett NL, Chen R, Fanale MA, Brice P, Gopal A, Smith SE, Advani R, Matous JV, Ramchandren R, Rosenblatt JD, et al. Retreatment with brentuximab vedotin in patients with CD30-positive hematologic malignancies. J Hematol Oncol. 2014;7:24.PubMedPubMedCentralCrossRef Bartlett NL, Chen R, Fanale MA, Brice P, Gopal A, Smith SE, Advani R, Matous JV, Ramchandren R, Rosenblatt JD, et al. Retreatment with brentuximab vedotin in patients with CD30-positive hematologic malignancies. J Hematol Oncol. 2014;7:24.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Foks AC, Bot I, Frodermann V, de Jager SC, Ter Borg M, van Santbrink PJ, Yagita H, Kuiper J, van Puijvelde GH. Interference of the CD30-CD30L pathway reduces atherosclerosis development. Arterioscler Thromb Vasc Biol. 2012;32(12):2862–8.PubMedCrossRef Foks AC, Bot I, Frodermann V, de Jager SC, Ter Borg M, van Santbrink PJ, Yagita H, Kuiper J, van Puijvelde GH. Interference of the CD30-CD30L pathway reduces atherosclerosis development. Arterioscler Thromb Vasc Biol. 2012;32(12):2862–8.PubMedCrossRef
101.
Zurück zum Zitat Willers J, Dummer R, Kempf W, Kundig T, Burg G, Kadin ME. Proliferation of CD30+ T-helper 2 lymphoma cells can be inhibited by CD30 receptor cross-linking with recombinant CD30 ligand. Clin Cancer Res. 2003;9(7):2744–54.PubMed Willers J, Dummer R, Kempf W, Kundig T, Burg G, Kadin ME. Proliferation of CD30+ T-helper 2 lymphoma cells can be inhibited by CD30 receptor cross-linking with recombinant CD30 ligand. Clin Cancer Res. 2003;9(7):2744–54.PubMed
102.
Zurück zum Zitat Oflazoglu E, Stone IJ, Gordon KA, Grewal IS, van Rooijen N, Law CL, Gerber HP. Macrophages contribute to the antitumor activity of the anti-CD30 antibody SGN-30. Blood. 2007;110(13):4370–2.PubMedCrossRef Oflazoglu E, Stone IJ, Gordon KA, Grewal IS, van Rooijen N, Law CL, Gerber HP. Macrophages contribute to the antitumor activity of the anti-CD30 antibody SGN-30. Blood. 2007;110(13):4370–2.PubMedCrossRef
103.
Zurück zum Zitat Nocentini G, Ronchetti S, Petrillo MG, Riccardi C. Pharmacological modulation of GITRL/GITR system: therapeutic perspectives. Br J Pharmacol. 2012;165(7):2089–99.PubMedPubMedCentralCrossRef Nocentini G, Ronchetti S, Petrillo MG, Riccardi C. Pharmacological modulation of GITRL/GITR system: therapeutic perspectives. Br J Pharmacol. 2012;165(7):2089–99.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Meiler S, Smeets E, Winkels H, Shami A, Pascutti MF, Nolte MA, Beckers L, Weber C, Gerdes N, Lutgens E. Constitutive GITR activation reduces atherosclerosis by promoting regulatory CD4+ T-cell responses—brief report. Arterioscler Thromb Vasc Biol. 2016;36(9):1748–52.PubMedCrossRef Meiler S, Smeets E, Winkels H, Shami A, Pascutti MF, Nolte MA, Beckers L, Weber C, Gerdes N, Lutgens E. Constitutive GITR activation reduces atherosclerosis by promoting regulatory CD4+ T-cell responses—brief report. Arterioscler Thromb Vasc Biol. 2016;36(9):1748–52.PubMedCrossRef
105.
Zurück zum Zitat Pedroza-Gonzalez A, Zhou G, Singh SP, Boor PP, Pan Q, Grunhagen D, de Jonge J, Tran TK, Verhoef C, JN IJ, et al. GITR engagement in combination with CTLA-4 blockade completely abrogates immunosuppression mediated by human liver tumor-derived regulatory T cells ex vivo. Oncoimmunology. 2015;4(12):e1051297.PubMedPubMedCentralCrossRef Pedroza-Gonzalez A, Zhou G, Singh SP, Boor PP, Pan Q, Grunhagen D, de Jonge J, Tran TK, Verhoef C, JN IJ, et al. GITR engagement in combination with CTLA-4 blockade completely abrogates immunosuppression mediated by human liver tumor-derived regulatory T cells ex vivo. Oncoimmunology. 2015;4(12):e1051297.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Kim WJ, Bae EM, Kang YJ, Bae HU, Hong SH, Lee JY, Park JE, Kwon BS, Suk K, Lee WH. Glucocorticoid-induced tumour necrosis factor receptor family related protein (GITR) mediates inflammatory activation of macrophages that can destabilize atherosclerotic plaques. Immunology. 2006;119(3):421–9.PubMedPubMedCentralCrossRef Kim WJ, Bae EM, Kang YJ, Bae HU, Hong SH, Lee JY, Park JE, Kwon BS, Suk K, Lee WH. Glucocorticoid-induced tumour necrosis factor receptor family related protein (GITR) mediates inflammatory activation of macrophages that can destabilize atherosclerotic plaques. Immunology. 2006;119(3):421–9.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Yu N, Fu S, Xu Z, Liu Y, Hao J, Zhang A, Wang B. Synergistic antitumor responses by combined GITR activation and sunitinib in metastatic renal cell carcinoma. Int J Cancer. 2016;138(2):451–62.PubMedCrossRef Yu N, Fu S, Xu Z, Liu Y, Hao J, Zhang A, Wang B. Synergistic antitumor responses by combined GITR activation and sunitinib in metastatic renal cell carcinoma. Int J Cancer. 2016;138(2):451–62.PubMedCrossRef
108.
Zurück zum Zitat Zhou L, Ismaili J, Stordeur P, Thielemans K, Goldman M, Pradier O. Inhibition of the CD40 pathway of monocyte activation by triazolopyrimidine. Clin Immunol. 1999;93(3):232–8.PubMedCrossRef Zhou L, Ismaili J, Stordeur P, Thielemans K, Goldman M, Pradier O. Inhibition of the CD40 pathway of monocyte activation by triazolopyrimidine. Clin Immunol. 1999;93(3):232–8.PubMedCrossRef
109.
Zurück zum Zitat Lee ME, Wang H. Homocysteine and hypomethylation. A novel link to vascular disease. Trends Cardiovasc Med. 1999;9(1-2):49–54.PubMedCrossRef Lee ME, Wang H. Homocysteine and hypomethylation. A novel link to vascular disease. Trends Cardiovasc Med. 1999;9(1-2):49–54.PubMedCrossRef
110.
Zurück zum Zitat Wang H, Yoshizumi M, Lai K, Tsai JC, Perrella MA, Haber E, Lee ME. Inhibition of growth and p21ras methylation in vascular endothelial cells by homocysteine but not cysteine. J Biol Chem. 1997;272(40):25380–5.PubMedCrossRef Wang H, Yoshizumi M, Lai K, Tsai JC, Perrella MA, Haber E, Lee ME. Inhibition of growth and p21ras methylation in vascular endothelial cells by homocysteine but not cysteine. J Biol Chem. 1997;272(40):25380–5.PubMedCrossRef
111.
Zurück zum Zitat Jamaluddin MD, Chen I, Yang F, Jiang X, Jan M, Liu X, Schafer AI, Durante W, Yang X, Wang H. Homocysteine inhibits endothelial cell growth via DNA hypomethylation of the cyclin A gene. Blood. 2007;110(10):3648–55.PubMedPubMedCentralCrossRef Jamaluddin MD, Chen I, Yang F, Jiang X, Jan M, Liu X, Schafer AI, Durante W, Yang X, Wang H. Homocysteine inhibits endothelial cell growth via DNA hypomethylation of the cyclin A gene. Blood. 2007;110(10):3648–55.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Halloran PF. Immunosuppressive drugs for kidney transplantation. N Engl J Med. 2004;351(26):2715–29.PubMedCrossRef Halloran PF. Immunosuppressive drugs for kidney transplantation. N Engl J Med. 2004;351(26):2715–29.PubMedCrossRef
113.
Zurück zum Zitat Smeets E, Meiler S, Lutgens E. Lymphocytic tumor necrosis factor receptor superfamily co-stimulatory molecules in the pathogenesis of atherosclerosis. Curr Opin Lipidol. 2013;24(6):518–24.PubMedCrossRef Smeets E, Meiler S, Lutgens E. Lymphocytic tumor necrosis factor receptor superfamily co-stimulatory molecules in the pathogenesis of atherosclerosis. Curr Opin Lipidol. 2013;24(6):518–24.PubMedCrossRef
114.
Zurück zum Zitat Mukundan L, Bishop GA, Head KZ, Zhang L, Wahl LM, Suttles J. TNF receptor-associated factor 6 is an essential mediator of CD40-activated proinflammatory pathways in monocytes and macrophages. J Immunol. 2005;174(2):1081–90.PubMedCrossRef Mukundan L, Bishop GA, Head KZ, Zhang L, Wahl LM, Suttles J. TNF receptor-associated factor 6 is an essential mediator of CD40-activated proinflammatory pathways in monocytes and macrophages. J Immunol. 2005;174(2):1081–90.PubMedCrossRef
115.
Zurück zum Zitat Sanguigni V, Ferro D, Pignatelli P, Del Ben M, Nadia T, Saliola M, Sorge R, Violi F. CD40 ligand enhances monocyte tissue factor expression and thrombin generation via oxidative stress in patients with hypercholesterolemia. J Am Coll Cardiol. 2005;45(1):35–42.PubMedCrossRef Sanguigni V, Ferro D, Pignatelli P, Del Ben M, Nadia T, Saliola M, Sorge R, Violi F. CD40 ligand enhances monocyte tissue factor expression and thrombin generation via oxidative stress in patients with hypercholesterolemia. J Am Coll Cardiol. 2005;45(1):35–42.PubMedCrossRef
116.
Zurück zum Zitat Yang J, Zhang L, Yu C, Yang XF, Wang H. Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomark Res. 2014;2(1):1.PubMedPubMedCentralCrossRef Yang J, Zhang L, Yu C, Yang XF, Wang H. Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomark Res. 2014;2(1):1.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Foley RN, Parfrey PS, Sarnak MJ. Clinical epidemiology of cardiovascular disease in chronic renal disease. Am J Kidney Dis. 1998;32(5 Suppl 3):S112–9.PubMedCrossRef Foley RN, Parfrey PS, Sarnak MJ. Clinical epidemiology of cardiovascular disease in chronic renal disease. Am J Kidney Dis. 1998;32(5 Suppl 3):S112–9.PubMedCrossRef
118.
Zurück zum Zitat Rogacev KS, Cremers B, Zawada AM, Seiler S, Binder N, Ege P, Grosse-Dunker G, Heisel I, Hornof F, Jeken J, et al. CD14++CD16+ monocytes independently predict cardiovascular events: a cohort study of 951 patients referred for elective coronary angiography. J Am Coll Cardiol. 2012;60(16):1512–20.PubMedCrossRef Rogacev KS, Cremers B, Zawada AM, Seiler S, Binder N, Ege P, Grosse-Dunker G, Heisel I, Hornof F, Jeken J, et al. CD14++CD16+ monocytes independently predict cardiovascular events: a cohort study of 951 patients referred for elective coronary angiography. J Am Coll Cardiol. 2012;60(16):1512–20.PubMedCrossRef
Metadaten
Titel
Metabolism-associated danger signal-induced immune response and reverse immune checkpoint-activated CD40+ monocyte differentiation
verfasst von
Jin Dai
Pu Fang
Jason Saredy
Hang Xi
Cueto Ramon
William Yang
Eric T. Choi
Yong Ji
Wei Mao
Xiaofeng Yang
Hong Wang
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2017
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-017-0504-1

Weitere Artikel der Ausgabe 1/2017

Journal of Hematology & Oncology 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.