Myeloid-derived suppressor cells (MDSCs) were initially identified in humans and mice with cancer where they profoundly suppress T cell– and NK cell–mediated antitumor immunity. Inflammation is a central feature of many pathologies and normal physiological conditions and is the dominant driving force for the accumulation and function of MDSCs. Therefore, MDSCs are present in conditions where inflammation is present. Although MDSCs are detrimental in cancer and conditions where cellular immunity is desirable, they are beneficial in settings where cellular immunity is hyperactive. Because MDSCs can be generated ex vivo, they are being exploited as therapeutic agents to reduce damaging cellular immunity. In this review, we discuss the detrimental and beneficial roles of MDSCs in disease settings such as bacterial, viral, and parasitic infections, sepsis, obesity, trauma, stress, autoimmunity, transplantation and graft-versus-host disease, and normal physiological settings, including pregnancy and neonates as well as aging. The impact of MDSCs on vaccination is also discussed.

Myeloid-derived suppressor cells (MDSCs) were first identified in patients with advanced cancer where they are profoundly immune-suppressive cells that inhibit antitumor immunity. With the notoriety of MDSCs as potent inhibitors of cancer immunotherapy and the knowledge that chronic inflammation is the dominant driving force for the development and function of MDSCs, investigators in other fields in which inflammation is also prevalent have asked whether MDSCs are induced. The answer has been a resounding “yes,” and MDSCs are now recognized as regulatory cells in many pathological settings as well as in normal physiological conditions. In contrast to cancer where MDSCs are exclusively detrimental because they are potent inhibitors of natural antitumor immunity and obstacles to cancer immunotherapies, MDSCs can be either detrimental or beneficial in noncancer settings. In addition, their potent immune-suppressive activity is being exploited in settings where cell-mediated immunity is damaging (Table I) (1–85).

Table I.
MDSCs are present in many immunological diseases and normal settings where they are either detrimental or beneficial
Disease/ConditionHuman/MouseMDSCsBeneficial/DetrimentalCommentsReferences
Cancer Human, mouse PMN-MDSCs,M-MDSCs Detrimental Prevent T cell activation; inhibit T cell function; promote protumor type 2 responses, metastasis, Tregs, stem cell expansion, angiogenesis (1–5
Bacterial infections Mouse S. aureus, mouse M-MDSCs, MDSCs Detrimental Promote biofilm formation (6, 7
TB, human MDSCs Detrimental Inhibit adaptive immunity (8
TB, human MDSCs Beneficial Can be converted to M1 macrophages and clear TB (9
Pneumonia, mouse and human Human/mouse M-MDSCs Beneficial IL-10 prevents lung damage, improves survival (10, 11
Viral infections Human hepatitis B and C, human M-MDSCs Detrimental Inhibit antiviral T cells; drive induced Tregs (12–15
HIV, human M-MDSCs Detrimental Inhibit antiviral immunity; drive Tregs (16–18
SARS-CoV-2, human M-MDSCs, PMN-MDSCs Detrimental, beneficial Suppress antiviral T cells; reduce damaging inflammation (19, 20
Parasitic diseases T. cruzi, mouse MDSCs Beneficial, detrimental Resolve inflammation; prevent T. cruzi–induced myocarditis; prolong parasite survival (21, 22
Leishmania, mouse MDSCs Beneficial, detrimental Suppress T cell activation; possibly promote leishmaniasis; kill amastigote stage; protect against infection (23–25
Malaria, human and mouse PMN-MDSCs Detrimental Inhibit CD4+ and CD8+ T cells; early marker for disease; promote Th17 response (26, 27
Schistosomiasis, mouse PMN-MDSCs Detrimental Suppress antischistosome T cell responses (28
Echinococcosis, mouse MDSCs Detrimental Inhibit Th2 cells; prevent helminth clearance (29
Sepsis and postsepsis immune suppression Human M-MDSCs, MDSCs Detrimental Suppress T cell activation and cytokine production; increased mortality due to infections; lymphopenia; high levels in early sepsis predict poor prognosis (30–32
Obesity Mouse MDSCs Beneficial Decrease metabolic dysfunction (glucose tolerance; glucose serum levels) (33, 34
Mouse MDSCs Detrimental Increase cancer progression (33, 35–37
Pregnancy and neonates Pregnancy, mouse PMN-MDSCs Beneficial Facilitate implantation; maintain pregnancy (38–41
Pregnancy, human PMN-MDSCs Beneficial Protect against pre-eclampsia; predict successful pregnancy following IVF (42–45
Preterm; normal term infants, mouse and human PMN-MDSCs Detrimental Increase susceptibility to infection (46, 47
Newborns, preterm infants, mouse and human M-MDSCs Beneficial Adoptive transfer of lactoferrin/ex vivo–induced MDSCs protect against pathologic inflammation (48, 49
Transplantation; GVHD Human and mouse Adoptive transfer of M-MDSCs and PMN-MDSCs for alloHCT Beneficial Higher levels of MDSCs predict less severe GVHD; higher levels correlate with less severe GVHD; M-MDSCs increase Tregs; better survival rates; does not reduce GVL (50–57
Autoimmunity EAE, colitis, SLE, CIA, type 1 diabetes, mouse MDSCs Beneficial Adoptive transfer downregulates Th1, Th17 cytokines; promotes Tregs; MDSCs expand IL-10–producing Bregs (40, 58–66
Uveitis, mouse and human M-MDSCs Beneficial Decrease Th1 and Th17; MDSC levels correlate with remission (61, 67
EAE, mouse MDSCs Detrimental Drive Th17 cells (68, 69
MS, human MDSCs Beneficial? Increased MDSCs correlate with remission (58
SLE, human and mouse M-MDSCs, PMN-MDSCs Detrimental Drive Th17 cells; impair Treg development (70, 71
Stress and trauma Stress, mouse MDSCs Detrimental Postsurgery stress: decrease T cell function; enhance metastasis (72
Trauma, mouse MDSCs Detrimental Decrease T cell function (73, 74
Trauma, human PMN-MDSCs Beneficial IL-10 reduces trauma-associated inflammation (75
Aging Human MDSCs Detrimental Presumably decrease immunocompetence (76
Vaccination Bacillus Calmette–Guérin in mice M-MDSCs Detrimental Impair T cell priming in DLNs (77
mRNA flu vaccine in rhesus macaques M-MDSCs, PMN-MDSCs Possibly detrimental Invade injection site but not DLNs (78
Hepatitis B vaccine in obese mice MDSCs Detrimental Inhibit T cell proliferation and Ab production (79
HIV vaccine in mice MDSCs Detrimental Reduce HIV-1 GAG-specific CD8+ T cells; induced MDSCs are HIV-infected and PD-L1+ (80
Parasite-infected humans MDSCs Potentially detrimental In endemic areas of parasite infection individuals may have infection-induced MDSCs (81
T. cruzi vaccine in mice MDSCs Detrimental MDSC depletion enhances vaccine efficacy (82
Ovarian cancer vaccine in mice MDSCs Detrimental MDSC depletion yields antitumor CD8+ T cells (83
HPV-targeted vaccine for multiple HPV+ tumors in mice MDSCs Detrimental Sunitinib depletion of MDSCs increases HPV E7-specific CD8+ T cells and survival (84
DC wild-type p53 vaccine in small-cell lung cancer patients MDSCs Detrimental ALTRA depletion of MDSCs increases p53-reactive CD8+ T cells (85
Disease/ConditionHuman/MouseMDSCsBeneficial/DetrimentalCommentsReferences
Cancer Human, mouse PMN-MDSCs,M-MDSCs Detrimental Prevent T cell activation; inhibit T cell function; promote protumor type 2 responses, metastasis, Tregs, stem cell expansion, angiogenesis (1–5
Bacterial infections Mouse S. aureus, mouse M-MDSCs, MDSCs Detrimental Promote biofilm formation (6, 7
TB, human MDSCs Detrimental Inhibit adaptive immunity (8
TB, human MDSCs Beneficial Can be converted to M1 macrophages and clear TB (9
Pneumonia, mouse and human Human/mouse M-MDSCs Beneficial IL-10 prevents lung damage, improves survival (10, 11
Viral infections Human hepatitis B and C, human M-MDSCs Detrimental Inhibit antiviral T cells; drive induced Tregs (12–15
HIV, human M-MDSCs Detrimental Inhibit antiviral immunity; drive Tregs (16–18
SARS-CoV-2, human M-MDSCs, PMN-MDSCs Detrimental, beneficial Suppress antiviral T cells; reduce damaging inflammation (19, 20
Parasitic diseases T. cruzi, mouse MDSCs Beneficial, detrimental Resolve inflammation; prevent T. cruzi–induced myocarditis; prolong parasite survival (21, 22
Leishmania, mouse MDSCs Beneficial, detrimental Suppress T cell activation; possibly promote leishmaniasis; kill amastigote stage; protect against infection (23–25
Malaria, human and mouse PMN-MDSCs Detrimental Inhibit CD4+ and CD8+ T cells; early marker for disease; promote Th17 response (26, 27
Schistosomiasis, mouse PMN-MDSCs Detrimental Suppress antischistosome T cell responses (28
Echinococcosis, mouse MDSCs Detrimental Inhibit Th2 cells; prevent helminth clearance (29
Sepsis and postsepsis immune suppression Human M-MDSCs, MDSCs Detrimental Suppress T cell activation and cytokine production; increased mortality due to infections; lymphopenia; high levels in early sepsis predict poor prognosis (30–32
Obesity Mouse MDSCs Beneficial Decrease metabolic dysfunction (glucose tolerance; glucose serum levels) (33, 34
Mouse MDSCs Detrimental Increase cancer progression (33, 35–37
Pregnancy and neonates Pregnancy, mouse PMN-MDSCs Beneficial Facilitate implantation; maintain pregnancy (38–41
Pregnancy, human PMN-MDSCs Beneficial Protect against pre-eclampsia; predict successful pregnancy following IVF (42–45
Preterm; normal term infants, mouse and human PMN-MDSCs Detrimental Increase susceptibility to infection (46, 47
Newborns, preterm infants, mouse and human M-MDSCs Beneficial Adoptive transfer of lactoferrin/ex vivo–induced MDSCs protect against pathologic inflammation (48, 49
Transplantation; GVHD Human and mouse Adoptive transfer of M-MDSCs and PMN-MDSCs for alloHCT Beneficial Higher levels of MDSCs predict less severe GVHD; higher levels correlate with less severe GVHD; M-MDSCs increase Tregs; better survival rates; does not reduce GVL (50–57
Autoimmunity EAE, colitis, SLE, CIA, type 1 diabetes, mouse MDSCs Beneficial Adoptive transfer downregulates Th1, Th17 cytokines; promotes Tregs; MDSCs expand IL-10–producing Bregs (40, 58–66
Uveitis, mouse and human M-MDSCs Beneficial Decrease Th1 and Th17; MDSC levels correlate with remission (61, 67
EAE, mouse MDSCs Detrimental Drive Th17 cells (68, 69
MS, human MDSCs Beneficial? Increased MDSCs correlate with remission (58
SLE, human and mouse M-MDSCs, PMN-MDSCs Detrimental Drive Th17 cells; impair Treg development (70, 71
Stress and trauma Stress, mouse MDSCs Detrimental Postsurgery stress: decrease T cell function; enhance metastasis (72
Trauma, mouse MDSCs Detrimental Decrease T cell function (73, 74
Trauma, human PMN-MDSCs Beneficial IL-10 reduces trauma-associated inflammation (75
Aging Human MDSCs Detrimental Presumably decrease immunocompetence (76
Vaccination Bacillus Calmette–Guérin in mice M-MDSCs Detrimental Impair T cell priming in DLNs (77
mRNA flu vaccine in rhesus macaques M-MDSCs, PMN-MDSCs Possibly detrimental Invade injection site but not DLNs (78
Hepatitis B vaccine in obese mice MDSCs Detrimental Inhibit T cell proliferation and Ab production (79
HIV vaccine in mice MDSCs Detrimental Reduce HIV-1 GAG-specific CD8+ T cells; induced MDSCs are HIV-infected and PD-L1+ (80
Parasite-infected humans MDSCs Potentially detrimental In endemic areas of parasite infection individuals may have infection-induced MDSCs (81
T. cruzi vaccine in mice MDSCs Detrimental MDSC depletion enhances vaccine efficacy (82
Ovarian cancer vaccine in mice MDSCs Detrimental MDSC depletion yields antitumor CD8+ T cells (83
HPV-targeted vaccine for multiple HPV+ tumors in mice MDSCs Detrimental Sunitinib depletion of MDSCs increases HPV E7-specific CD8+ T cells and survival (84
DC wild-type p53 vaccine in small-cell lung cancer patients MDSCs Detrimental ALTRA depletion of MDSCs increases p53-reactive CD8+ T cells (85

DLN, draining lymph node; TB, tuberculosis.

Historically, MDSCs have predominantly been characterized in mouse and human cancer, but MDSCs in other settings appear to share similar key characteristics. As it has become apparent that MDSCs are involved in many immunological processes, MDSC development and function are also being explored in noncancer immune conditions. We briefly summarize the salient features of MDSCs and then focus on MDSCs in settings other than cancer. More detailed descriptions of the development and function of cancer-induced MDSCs can be found in several excellent recent reviews (1–5).

MDSCs are a diverse population of myeloid cells that span the differentiation pathway of the common myeloid progenitor cell. MDSC accumulation is initiated when NOTCH and IRF8 are downregulated and “emergency myelopoiesis” occurs. The expansion of MDSCs occurs at the expense of other cells in the common myeloid progenitor lineage.

There are two dominant phenotypes of MDSCs: monocytic (M-MDSCs) and polymorphonuclear or granulocytic (PMN-MDSCs). In mice, MDSCs are phenotypically identified as Gr1+CD11b+ cells, with M-MDSCs being CD11b+Ly6C+Ly6G and PMN-MDSCs being CD11b+Ly6G+Ly6C−/low. Neutrophils also have the latter phenotype in infectious disease settings. In humans, MDSCs are CD33+CD11b+HLA-DRlow/−, with M-MDSCs being CD14+CD15 and PMN-MDSCs being CD15+CD14CD66b+. Cells with a third phenotype termed early stage MDSCs (eMDSCs) are present in humans; these are CD33+CD11b+ and lack other myeloid markers. The absence of HLA-DR distinguishes human MDSCs from monocytes. MDSCs are primarily identified by the immune regulatory molecules they produce. M-MDSCs generate NO synthase 2 (NOS2), NO, TGF-β, and IL-1β. PMN-MDSCs contain NOX2 and reactive oxygen species (ROS, H2O2, O2). Both types of MDSCs can produce arginase 1 (Arg1), peroxynitrite (ONNO), PGE2, and IL-10. These phenotypic markers in combination with assays demonstrating functional suppression of T cells (e.g., inhibition of T cell proliferation, T cell activation including IFN-γ or IL-2 production, T cell CD3ζ downregulation, and for CD8 T cells, T cell cytotoxic activity) are the defining but not exclusive parameters of MDSCs (86) (Fig. 1).

FIGURE 1.

MDSCs are predominantly induced by proinflammatory mediators and by some noninflammatory molecules and are naturally present in many pathological and normal settings. MDSCs are also induced ex vivo and used for adoptive transfer to downregulate undesirable immune responses. Multiple proinflammatory and a few noninflammatory molecules in combination with adrenergic signaling drive the accumulation and suppressive function of MDSCs, including GM-CSF, PGE2, IL-6, VEGF, S100A8/A9, TNF-α, G-CSF, IL-1β, HMGB1, leptin, adenosine, lactoferrin, and oxidized lipids. PMN-MDSCs and M-MDSCs both use Arg1, PGE2, peroxynitrite (ONNO), and IL-10 to suppress T cell activation and function, whereas PMN-MDSCs also use hydrogen peroxide (H2O2), superoxide (O2), and NOX2, and M-MDSCs also use NO, TGF-β, and IL-1β.

FIGURE 1.

MDSCs are predominantly induced by proinflammatory mediators and by some noninflammatory molecules and are naturally present in many pathological and normal settings. MDSCs are also induced ex vivo and used for adoptive transfer to downregulate undesirable immune responses. Multiple proinflammatory and a few noninflammatory molecules in combination with adrenergic signaling drive the accumulation and suppressive function of MDSCs, including GM-CSF, PGE2, IL-6, VEGF, S100A8/A9, TNF-α, G-CSF, IL-1β, HMGB1, leptin, adenosine, lactoferrin, and oxidized lipids. PMN-MDSCs and M-MDSCs both use Arg1, PGE2, peroxynitrite (ONNO), and IL-10 to suppress T cell activation and function, whereas PMN-MDSCs also use hydrogen peroxide (H2O2), superoxide (O2), and NOX2, and M-MDSCs also use NO, TGF-β, and IL-1β.

Close modal

In the presence of inflammation in general, as well as certain noninflammatory molecules, MDSCs expand and gain increased suppressive potency. Proinflammatory cytokines, including IL-6, IL-1β, TNF-α, VEGF, GM-CSF, and G-CSF, the bioactive lipid PGE2, and the DAMP/alarmins HMGB1 and S100A8/A9, all upregulate MDSCs, as do the noninflammatory molecules adenosine (87), lactoferrin (48), and leptin (33). In combination with adrenergic signaling these molecules activate MDSCs through the STAT3 and NF-κB pathways. In the tumor microenvironment (TME), inducers are produced by tumor cells or are generated by the hypoxic TME resulting in activation of an endoplasmic reticulum stress response (reviewed in Ref. 2) (Fig. 1).

MDSCs exist in the circulation of healthy individuals at low levels. When they enter the TME or other proinflammatory locales, they undergo metabolic changes. Within the TME, MDSCs become dependent on the uptake of lipids and fatty acid oxidation (FAO). Polyunsaturated fatty acids increase the differentiation of mouse MDSCs and upregulate MDSC suppressive activity by increasing ROS production via JAK-STAT3 signaling (88). Pharmacologic inhibition of fatty acid uptake and FAO reduces mouse and human MDSC suppressive potency and reprograms MDSCs to contain more mitochondria, synthesize enzymes essential for FAO, increase the oxidation rate (89, 90), and increase the expression of fatty acid transport proteins (FATPs) (91). G-CSF and GM-CSF induce mouse and human MDSCs by upregulating the expression of lipid transport receptors such as FATP2 and fatty acid metabolic pathways via STAT3 and STAT5 signaling. Increased FATP2 expression in PMN-MDSCs facilitates the uptake of arachidonic acid, which is converted intracellularly to PGE2 (92), a dominant driver of MDSC suppressive potency (93, 94). This suppressive activity is enhanced by mast cells (95). Metabolic-driven accumulation of methylglyoxal also increases the suppressive activity of MDSCs (96) as described in MDSC suppressive mechanisms.

MDSCs circulating in blood use oxidative phosphorylation for energy generation but convert to glycolysis (Warburg effect) when they enter the hypoxic TME (97). Immunometabolic and single-cell RNA sequencing studies of MDSCs demonstrate that maturation and the acquisition of increased suppressive potency requires aerobic glycolysis and the consumption of high levels of glucose. However, MDSCs themselves consume large amounts of glucose, and their limited lifespan in vivo is likely due to insufficient quantities of glucose (6).

As alluded to above, MDSCs use multiple mechanisms to suppress adaptive and innate immunity. MDSCs are best known for their ability to inhibit T cell activation and function. They produce Arg1 and IDO and sequester cysteine, thereby limiting the availability of l-arginine, cystine, and tryptophan, amino acids essential for T activation (98–100). MDSCs also deplete T cells of essential l-arginine by their accumulation of the metabolite methylglyoxal, which they transfer to T cells. Within T cells, methylglyoxal glycates l-arginase, thereby rendering it unusable by T cells (96).

MDSCs produce ROS and NOS2 to generate peroxynitrite, which nitrates TCRs, MHC molecules, and chemokines, preventing T cell recognition of tumor Ags and blocking T cell chemotaxis (101, 102). Although MDSCs produce high levels of ROS, they are not themselves affected due to their upregulation of NF erythroid-2 (Nrf2), which controls a battery of antioxidant genes (103). L-selectin is essential for naive T cells to enter lymph nodes and become activated. MDSCs perturb this trafficking by their expression of ADAM-17, which cleaves L-selectin (104, 105). MDSCs are also potent inducers of T regulatory cells (Tregs) through their production of IDO, TGF-β, and IL-10 (106). Engagement of MDSC-expressed PD-L1 and galectin-9, ligands for the T cell inhibitory receptors PD-1 and Tim-3, respectively, drives T cell exhaustion and arrest (107, 108). Activated T cells can defend themselves against PD-L1+ MDSCs through their expression of FasL, which mediates apoptosis of Fas+ MDSCs (109). As MDSCs accumulate and differentiate, they synthesize S100A9, thereby skewing myeloid cell differentiation toward MDSCs and away from dendritic cells (DCs), resulting in diminished availability of cells for Ag processing and presentation for T cell activation (110).

MDSCs also inhibit humoral immunity. Their production of Arg1, NO, and ROS decreases B cell IgM responses and induces B cell death (111), and their synthesis of TGF-β downregulates IL-7 production and downstream STAT5 signaling, which are essential for B cell development (112).

Innate immunity is also inhibited by MDSCs. MDSCs express membrane-bound TGF-β1, which inhibits NK cell expression of NKG2D and reduces NK cell production of IFN-γ, thereby anergizing NK cells (113). MDSC production of IL-10 drives the polarization of macrophages toward an M2 phenotype, which promotes tumor progression (114).

Many of the suppressive mediators are transported in exosomes and include proteins, glycoproteins, microRNAs, and mRNAs (115, 116). MDSC activity does not involve specific receptors; however, cell-to-cell proximity is required, presumably due to concentration effects of secreted/released materials. Given the diversity of MDSCs, it is likely that their phenotype and function vary depending on the physiological setting, so that not all suppressive mechanisms are relevant in all settings. Fig. 2 summarizes the suppressive mechanisms used by MDSCs.

FIGURE 2.

Immune-suppressive mechanisms used by MDSCs. MDSCs use a variety of mechanisms involving secreted molecules, membrane-bound molecules, cytokines, and metabolic reprogramming to suppress T cells, B cells, and innate immune effector cells. T cell mechanisms are shown in yellow boxes, B cell mechanisms in purple boxes, and innate mechanisms in red boxes.

FIGURE 2.

Immune-suppressive mechanisms used by MDSCs. MDSCs use a variety of mechanisms involving secreted molecules, membrane-bound molecules, cytokines, and metabolic reprogramming to suppress T cells, B cells, and innate immune effector cells. T cell mechanisms are shown in yellow boxes, B cell mechanisms in purple boxes, and innate mechanisms in red boxes.

Close modal

PMN-MDSCs and M-MDSCs share some characteristics with neutrophils and inflammatory monocytes, respectively. Whether MDSCs are a distinct population of cells or are a subset of neutrophils or inflammatory monocytes remains controversial. MDSCs were originally defined by their phenotype, their accumulation under pathological (cancer) conditions, and their exceptional ability to inhibit T cell activation and function (117). Other myeloid cells including tumor-associated neutrophils and tumor-associated macrophages share some of these properties with MDSCs. However, studies have identified the lectin-type oxidized low-density lipoprotein receptor-1 (LOX1) (118) and FATP2 as specific markers for PMN-MDSCs (5). M-MDSCs, which phenotypically resemble inflammatory monocytes, can differentiate into tumor-associated macrophages (119) and fibrocytes (120), activities that are not associated with classical monocytes. Likewise, some of the mechanisms used by MDSCs to suppress T cell activation and function, such as cysteine deprivation (99) and l-selectin downregulation (104), are not functions of neutrophils or monocytes, and single-cell transcriptional analyses of PMN-MDSCs and tumor-associated neutrophils have identified transcriptomes specific for each cell type (121). These differences do not speak to the relationship between MDSCs, neutrophils, and inflammatory monocytes; however, they define MDSCs as a functionally distinct cell population. Many reports define MDSCs without using the full repertoire of MDSC characteristics, so there is some ambiguity as to whether the cells are full-fledged PMN-MDSCs or neutrophils, or M-MDSCs or other monocytes. In this review, MDSCs are defined as cells with the established phenotype and demonstrated immune-suppressive activity.

Some of the following studies distinguish between PMN-MDSCs and M-MDSCs; however, other studies have not separated the two populations. When studies identified PMN-MDSCs and M-MDSCs, then the specific identification is used. However, when reports do not separate the two subtypes, then we use the terminology “MDSCs.”

MDSCs expand in response to inflammation in all bacterial and viral diseases studied to date. The initial expansion of MDSCs presumably protects the host against excessive inflammation. However, when MDSCs persist, they inhibit the antipathogen function of T and NK cells, leading to reduced pathogen control and increased disease progression. Therefore, MDSCs can have both protective and detrimental functions.

Bacterial infections

MDSCs play both detrimental and beneficial roles in bacterial infection, with MDSC-derived IL-10 being a key factor. Metabolic profiling demonstrated that limited amounts of environmental glucose in invasive Staphylococcus aureus disease inhibit the differentiation of MDSCs into more mature nonsuppressive cells (6). Nonetheless, MDSCs generated in S. aureus infection produce IL-10, which supports biofilm formation (7). In contrast, M-MDSC–derived IL-10 promotes clearance of Klebsiella pneumoniae in mice and reduces inflammation, thereby improving host survival (10, 11). MDSCs also accumulate in patients infected with Mycobacterium tuberculosis as well as in individuals recently exposed to M. tuberculosis (8). The MDSCs typically inhibit immunity to M. tuberculosis. However, if their leukocyte Ig-like receptor B is blocked by Ab, then human MDSCs are converted to M1-type macrophages, which mediate the intracellular killing of M. tuberculosis (9).

Viral infections

In general, MDSCs play a detrimental role in viral infections and the generation and maintenance of antiviral immunity. M-MDSCs accumulate in blood and the liver of hepatitis B patients where they inhibit antiviral T cell responses (12). M-MDSC–derived IL-10 promotes the development of induced Tregs, which further inhibit antiviral immunity (13). These findings have been replicated in chronic hepatitis C patients (14, 15). In HIV-infected individuals, M-MDSCs accumulate in response to IL-6 and drive Treg levels via their production of IL-10 (16). The persistence of immune dysfunction in AIDS patients on combined antiretroviral therapy is thought to be due to the persistence of M-MDSCs (17).

M-MDSCs accumulate in the blood of SARS-CoV-2–infected patients with both mild and severe disease (122, 123). Higher levels of M-MDSCs and PMN-MDSCs at the time of hospital admission are predictive of severity (19), and recovery is associated with reductions in MDSCs (122). PMN-MDSCs similarly expand in patients with severe acute respiratory distress syndrome accompanying SARS-CoV-2 infection (20), and they correlate with COVID-19–associated lymphopenia. The continued accumulation of MDSCs parallels fatal disease, although it is unclear whether MDSCs contribute to fatality through their inhibition of antiviral immunity (124). COVID-19–induced MDSCs suppress T cell activation in vitro and, as for other pathogens, the anti-inflammatory and immune-suppressive functions of MDSCs work antagonistically in that they limit the damaging inflammation, but at the same time suppress antiviral immunity. Transcriptional analysis suggests that PMN-MDSCs of COVID-19 patients with severe disease have increased Arg1 expression relative to asymtomatic patients. People who die of COVID-19 have large infiltrates of Arg1+ PMN-MDSCs in their lungs, suggesting that ROS production by PMN-MDSCs may contribute to lung pathology (125). In patients with asymptomatic or mild disease, MDSCs remain elevated for at least 3 mo postrecovery (126). Given the excessive levels and persistence of MDSCs, it is questionable whether new T cells are activated in immunized patients who succumb to SARS-CoV-2 infection, or whether resolution of the disease is due to adaptive immunity induced predisease by vaccination.

Myeloid cells are key players in antiparasitic immunity with different roles in protozoan and metazoan infections. Although not extensively studied, it has been reported that MDSCs are produced in tandem with enhanced myelopoiesis during several parasitic infections (127). MDSCs are also considered to be part of “trained” immunity during fungal infections including Aspergillus (128), Candida (128–130), and Cryptococcus (131). The effects of MDSCs on immunity to complex pathogens can be detrimental in some infections where T cell–mediated immunity is protective. For example, arginase inhibitors, specifically the p38 inhibitor SB202190 or the receptor tyrosine kinase vandetanib, suppress PMN-MDSCs in Cryptococcus neoformans fungal infection and enhance protective T cell responses (131).

MDSCs, particularly M-MDSCs, play a key role in kinetoplastid infections. In mouse models they resolve inflammation during Trypanosoma cruzi acute infection (21, 22) and prevent T. cruzi infiltration of the heart causing Chagas-associated myocarditis (132). A cellular therapy with mesenchymal stem/stromal cells overexpressing the MDSC inducer G-CSF increased MDSC migration to the heart (133) where they suppressed cardiac pathology. MDSCs suppress T cell function by both Arg1 (22) and NO (134). In mouse T. cruzi infection, suppression prolongs parasite survival in heart tissue. In Chagas disease, MDSCs alter ROS and NO-mediated mechanisms of T. cruzi parasite killing (22). In contrast, MDSCs in leishmaniasis expand in response to parasite Ag (23) and suppress T cell proliferation (23, 24), possibly contributing to progressive visceral leishmaniasis (25). Surprisingly, the NO induced in MDSCs is lethal to intracellular amastigote stages and protects against propagation of infection (24). In contrast, glycyrrhizic acid inhibits Cox-2–mediated MDSC suppression and restores T cell proliferation leading to control of parasite numbers in BALB/c mice (23).

There is little information on MDSCs in apicomplexan infection. This is surprising given the burden of malaria, the recent surge in understanding Cryptosporidium with the emergence of a tractable model of study, and the historically strong focus on Toxoplasma. An initial report described the generation of PMN-MDSCs in controlled human malaria challenge with Plasmodium falciparum and their suppression of CD3/CD28-mediated T cell proliferation (26). A recent paper indicates the presence of potential MDSC populations in PBMCs during naturally acquired infections with severe P. falciparum and uncomplicated Plasmodium vivax malaria, albeit suppressive capacity was not tested (135). Splenic MDSC populations, in particular NO-expressing PMN-MDSCs, expand in C57BL/6 mice infected with Plasmodium berghei ANKA (27), a mouse model of cerebral malaria. Expansion was dependent on STAT3 signaling emanating from the IL-6 receptor and correlated with promoting Th17 responses. The significance of this finding is unclear because other studies suggest that neurologic manifestations of P. berghei ANKA in mice are independent of IL-17 (136).

Schistosoma japonicum worm Ag and egg Ag also drive differentiation of MDSCs via STAT3 (137) with ROS-dependent suppression on antischistosome T cell responses (28). Similarly, PD-L1 expression by PMN-MDSCs has been suggested to suppress antischistosome Tfh1 cells (28), potentially impacting antischistosome humoral immunity. MDSCs also expand in Echinococcus granulosus infection of BALB/c mice (138). These MDSCs use NO to suppress Th2 cell development that drives antihelminth mechanisms of clearance (29). E. granulosus–driven MDSCs have elevated transcription of VEGF and induce angiogenesis in cultured HUVECs (139). Furthermore, multiple microRNAs involved in immunoregulatory pathways (139) as well as long coding RNAs (140) are enriched in MDSCs from E. granulosus–infected mice.

Mast cells impact MDSC function in parasitic disease

Mast cells are key mediators in parasite expulsion, but they effect MDSC function, which can feed back into antiparasite immunity. In the TME, mast cells augment accumulation of M-MDSCs (141, 142), in part through histamine released during mast cell degranulation (143). However, mast cells are critical mediators of parasite expulsion, and PMN-MDSCs enhance antiparasitic immunity and have been correlated with elevated levels of the Th2 cytokines IL-4, IL-5, IL-13 (144), as well as IL-17 and the alarmin IL-33. Depletion of MDSCs by gemcitabine enhanced infection with the rat hookworm Nippostrongylus (144) and the nematode Trichinella spiralis (145). Although off-target effects of gemcitabine were assessed, there was no separation of neutrophils from PMN-MDSCs, making it difficult to separate effects of PMN-MDSCs from the protective effects of neutrophil extracellular traps produced from mature neutrophils in the Nippostrongylus model (146). This point is pertinent because of the role of mast cell tryptase in the induction of neutrophil extracellular traps (147). Nonetheless, the enhancement of IgE-mediated mast cell function, a key mechanism in mast cell activation during helminth infection, suggests how MDSCs augment immunity to intestinal helminths. This activity may also impact other infections such as malaria, where the role of mast cell degranulation promotes parasite survival in the P. berghei ANKA mouse model in some (148) but not all (149) genetic backgrounds.

Sepsis results when innate immunity overresponds to infection, and the release of proinflammatory cytokines and chemokines causes cardiovascular dysfunction and organ destruction. Hyperinflammation, cytokine release syndrome, tissue hypoxia, hypercoagulation, and endothelial cell destruction are other potentially lethal events (150). Medical advances have improved survival of patients with acute sepsis; however, sepsis survivors can have long-term complications including chronic inflammation, immune suppression, tissue wasting, and lymphopenia. A study of 74 patients with severe sepsis/septic shock detected increasing levels of MDSCs throughout the 28-d study period (151), and high levels of MDSCs during early sepsis are predictive of a poor prognosis (32). Chronic immune suppression in post–severe sepsis/septic shock patients has a poor clinical prognosis due to infection and is associated with abnormal myelopoiesis and the accumulation of MDSCs (152, 153).

Studies of postsurgical sepsis patients underscore the detrimental effects of MDSCs, particularly M-MDSCs, in suppressing immunity to infection. In 267 survivors of surgical sepsis, MDSC levels were elevated for at least 6 wk postinfection (30), suppressing Ag-driven T cell proliferation and cytokine production and leading to increased nosocomial infections. Cardiac surgery patients with elevated levels of M-MDSCs had higher incidences of postsurgery sepsis-induced immune suppression and subsequent mortality due to infection (154). Elevated levels of M-MDSCs were similarly found to predispose to sepsis-induced immune suppression and mortality due to infection in 301 septic shock patients, 50 of whom exhibited post–septic shock immune suppression (31). A single-cell RNA sequencing study of MDSCs from two 21-d postsepsis patients and two healthy controls identified common transcripts as well as transcripts unique to post sepsis MDSCs, suggesting that postsepsis MDSCs may have a distinct transcriptome (155).

Studies in mice have provided mechanistic insight into MDSCs in sepsis. A study using cecal ligation and puncture followed by polymicrobial infection demonstrated that late-phase MDSCs accumulate in the vasculature of nonlymphoid tissues, including the lungs (156). In the same model, COX2 (157) and S100A9 (158) are key drivers of MDSCs in postsepsis immune suppression, and in a mouse LPS-induced sepsis system Nrf2 is essential for MDSC survival (159). In a neonatal sepsis model (bacterial infection of 3- to 4-d-old mice) MDSCs contain elevated transcripts for NOS2, Arg1, and IL-27p28 and express TLR2, TLR4, and TLR5, which recognize multiple pathogen-associated molecular patterns of Escherichia coli (160).

The chronic presence of multiple proinflammatory mediators such as IL-6, TNF-α, PGE2, and IL-1β in obesity is reminiscent of the TME. Because these molecules are also inducers of MDSCs, it is not surprising that MDSCs are elevated in obese patients (161, 162). Mice that become obese through consumption of a high-fat diet (HFD) have elevated levels of MDSCs in their circulation and adipose tissue, and tumor growth is accelerated (33, 37, 163). Although MDSC-like cells are elevated in morbidly obese patients and the levels decrease following bariatric surgery, these cells have low levels of ROS and do not suppress T cell proliferation (164). Therefore, not all MDSCs in obese individuals have the same characteristics.

Obesity is a risk factor for cancer development and progression (165–167). The discovery of MDSCs in obese individuals led to the concept that immune suppression by MDSCs contributes to cancer risk (36), and studies in tumor-bearing obese mice, including breast (39), ovarian (35), and renal (37) cancers, support this hypothesis.

Diabetes can also be a consequence of obesity and is characterized by elevated fasting glucose levels and high insulin. HFD obese mice have elevated TNF-α in the circulation and adipose tissues, as well as elevated blood glucose, and are insulin resistant. Depletion of MDSCs further increased blood glucose levels and insulin tolerance, indicating that these cells protected the mice against more extreme metabolic dysfunction, while concomitantly enhancing tumor progression (33, 34). Leptin is produced by adipose cells in response to inflammation and is an appetite suppressant in nonobese healthy individuals. It regulates the balance between metabolism and food intake and is frequently overexpressed in obese individuals who become nonresponsive to its regulatory function. Leptin levels are elevated in the blood of HFD obese mice. Depletion of MDSCs increases leptin levels and blocking the leptin receptor in HFD obese mice lowers circulating MDSC levels. Hence, leptin serves as a driver for MDSC accumulation while MDSCs downregulate leptin levels (33).

During healthy pregnancies women are tolerant to their semiallogeneic fetus, a phenomenon known as maternal–fetal tolerance. It is likely that multiple mechanisms are responsible for maternal–fetal tolerance. MDSCs appear to be one of the key mechanisms. PMN-MDSCs are present in human cord blood, accumulate in the peripheral blood and endometrium during healthy pregnancies, and are diminished in women experiencing spontaneous abortions and miscarriages (168, 169). Following parturition, blood PMN-MDSC levels decrease. Pregnancy-associated PMN-MDSCs suppress T cell activation, contain Arg1 and NOS2, produce large amounts of ROS, and polarize CD4+ T cells toward a Th2 phenotype (170, 171). Decreases in MDSCs during early miscarriage track with declines in estrogen and progesterone and the diverting of CD4+ T cells away from a Th2 phenotype and toward a Th1 phenotype, although it is unclear whether these effects are linked to MDSC levels (172).

Studies in mice demonstrated PMN-MDSC infiltration into the uterus of pregnant females. The MDSCs are activated via STAT3 and used ROS to suppress T cell activation (40, 41). Depletion of MDSCs early in gestation caused implantation failure, enhanced T cell activation, and increased T cell migration into the uterus. Induction of MDSCs by G-CSF reversed these effects. Naive T cells of pregnant mice had reduced l-selectin and were impaired in their ability to enter lymph nodes and become activated (39). Mice that contain HIF-1α–deficient myeloid cells contain fewer uterus MDSCs, decreased implantation rates, and increased abortions, indicating that hypoxia in the uterus is critical for the development of MDSCs and successful pregnancy (38).

PMN-MDSC deficiency contributes to pre-eclampsia in women. PMN-MDSCs express the HLA-G receptors ILT2 and ILT4, which are increased during pregnancy. A soluble form of HLA-G increases PMN-MDSC suppressive potency by activating STAT3 (44). Mouse studies using Qa-2, the mouse analog of human HLA-G, demonstrated that Qa-2 deficiency resulted in a pre-eclampsia–like condition and abortion that was reversed by soluble HLA-G (42). In a meta-analysis study elevated circulating levels of PMN-MDSCs were associated with and predictive of a more favorable outcome for women undergoing in vitro fertilization (43, 45).

PMN-MDSCs are also present in preterm and normal term infants but decrease rapidly after the 28-d neonatal period. Preterm delivery is a major cause of perinatal morbidity and mortality, and neonates are typically highly susceptible to infection. The presence of MDSCs may contribute to these conditions (46, 47). However, mouse studies indicate that MDSCs may also play a protective role in that their transitory presence downregulates potentially lethal inflammatory conditions that can occur in newborns (48, 49).

Acute and chronic graft-versus-host disease (GVHD) is the dominant complicating factor for allogeneic hematopoietic stem cell transplantation (alloHCT), while host-versus-graft responses are the major deterrent for successful solid organ transplantation. MDSCs naturally occur in alloHCT; however, the levels are insufficient to prevent GVHD. AlloHCT into irradiated recipients produces an inflammatory environment conducive to MDSC development. M-MDSCs and PMN-MDSCs are associated with reduced incidence of acute GVHD in humans (50, 51), and adoptive transfer of M-MDSCs protects against acute GVHD in mice (52). Likewise, increased M-MDSCs in posttransplant renal patients may protect indirectly against host-versus-graft rejection by induction of Tregs (53). Therefore, MDSCs have the potential to play an important role in reducing unwanted immune responses in transplant settings.

Exploiting MDSCs for improving transplantation efficacy

The immunosuppressive activity of MDSCs led to studies aimed at using in vivo induction or adoptive transfer of in vitro–generated MDSCs for combating GVHD in alloHCT and for minimizing host-versus-graft responses in solid organ transplantation (173, 174). Early mouse studies used GM-CSF in combination with G-CSF to expand MDSCs from bone marrow, which were then adoptively transferred into allogeneic mice providing proof of principle of this strategy (56). Dexamethasone in combination with GM-CSF extends survival of allogeneic cardiac transplants in mice via MDSCs that increase Tregs and use NOS2 to suppress T cell function (175). A comparison of naturally occurring MDSCs, GM-CSF–induced MDSCs, and G-CSF–induced MDSCs in a mouse cardiac transplant model identified G-CSF as most effective in extending graft survival (176). GM-CSF also expands MDSCs from human cord blood CD34+ cells, and inclusion of stem cell factor generates more robust MDSCs. In a xenogeneic NSG mouse system adoptive transfer of these human MDSCs decreased GVHD and upregulated Tregs (177). Thus, multiple strategies have been used to generate MDSCs in mouse and human settings.

Clinical studies

Clinical data support a role for MDSCs in limiting GVHD in transplant settings. Posttransplant treatment with cyclophosphamide improved PMN-MDSC generation, maintenance, and suppressive activity and reduced grade II to IV acute GVHD in children undergoing alloHCT (MCC protocol 19295) (54). AlloHCT patients adoptively transferred with cells from pegylated GCF-mobilized MDSCs had reduced rates of grade III–IV acute GVHD compared with patients given nonpegylated GCF-mobilized MDSCs, suggesting that pegylation increased MDSC half-life. Overall survival rates did not differ between the pegylated and nonpegylated groups; however, pegylation may reduce the number of MDSC transfers and enhance MDSC function when using growth factor–induced MDSCs (178).

Studies have also focused on location of both solid organ and alloHCT grafts to determine whether MDSCs suppress at the graft site or systemically. CCR2-mediated migration of MDSCs to the graft site is critical in a mouse allogeneic islet transplant system and resulted in site-specific inhibition of CD8+ T cell function and enhancement of Tregs (179). Adoptive transfer of γδ Th17 cells in a mouse alloHCT model increased MDSC suppressive potency and drove infiltration of MDSCs into the inflamed intestine, thereby reducing local GVHD (180). In a mouse allogeneic cardiac transplant model, MDSC activation required host NKT cells and their production of IL-4 (181), and umbilical cord mesenchymal stromal cells secreting HLA-G enhanced MDSC generation, suppressive activity, and the ability to reduce acute GVHD in a mouse alloHCT system (55). Thus, other cells influence MDSC activation and function, and graft location may dictate MDSC efficacy.

MDSCs in graft-versus leukemia

In patients with leukemias it is essential that GVHD be reduced but the graft-versus-leukemia (GVL) effect be maintained. This outcome requires that MDSCs suppress GVHD without impacting the GVL response, a requirement that is supported by several studies (55, 56). This observation is perplexing because MDSCs are neither Ag specific nor MHC restricted. A study using mouse models of allogeneic T cell transfer into lethally irradiated recipients followed by engrafting with tumor cells suggested that NKG2D+CD8+ T cells with a memory phenotype are essential for elimination of leukemic cells and are resistant to MDSC-mediated suppression (57), providing a potential explanation for how GVHD can be reduced by MDSC therapy without diminishing the GVL response.

Autoimmune diseases are typically associated with local chronic inflammation and the activation of autoreactive CD8+ T cells coupled with excessive differentiation of CD4+ Th1 and Th17 cells and inhibition of protective Tregs. MDSCs are often present in animal models of autoimmunity and in patients with autoimmune diseases but do not completely prevent pathology. In several autoimmune conditions MDSCs arise spontaneously prior to remission, and some mouse models indicate that MDSCs may downregulate pathogenic Th1 and Th17 cells, suggesting that MDSCs may contribute to resolution of disease. However, there are also studies suggesting that MDSCs may exacerbate autoimmunity. Therefore, whether MDSCs can be exploited as therapeutic agents is controversial.

Beneficial effects of MDSCs in autoimmune diseases

Most studies use adoptive transfer of MDSCs into mice with an established autoimmune disease. Experimental autoimmune encephalomyelitis (EAE) is an accepted mouse model for multiple sclerosis (MS). Mice are immunized with myelin oligodendrocyte glycoprotein (MOG)35–55 and develop Ag-specific Th1 and Th17 cells. EAE mimics human MS in that there are cycles of disease and remission. Plasmacytoid DCs suppress MDSC accumulation in EAE (182); however, glycolipid-activated invariant NKT cells favor the accumulation of MDSCs (183). Adoptive transfer of syngeneic MOG-induced MDSCs into EAE mice reduces EAE severity by downregulating myelin-reactive Th1 and Th17 cells. PD-L1 expression by the transferred MDSCs is essential for disease remission in mice and probably in SLE patients (58). B cell production of GM-CSF increases EAE severity. PMN-MDSCs are beneficial in that they downregulate B cell synthesis of GM-CSF. MS patient data support a role for PMN-MDSC B cell interactions because cerebral spinal fluid frequencies of CD138+ B cells negatively correlate with PMN-MDSC and cytokine levels (184).

B regulatory cells (Bregs) play an important role in some autoimmune diseases by their production of IL-10, which downregulates proinflammatory cytokines and drives Treg differentiation (185). C57BL/6 female Roquinsan/san mice spontaneously develop systemic lupus erythematosus (SLE). IL-10–secreting Bregs expand when cocultured with MDSCs, and their adoptive transfer into SLE mice decreases renal pathology by reducing anti-DNA Abs, effector B cells, germinal center B cells, and follicular Th1 and Th17 cells (64). As with EAE, expression of PD-L1 is important for MDSC-mediated protection against SLE (186).

Similar MDSC protective effects are seen in mice with dextran sodium sulfate and/or 2,4,6-trinitrobenzene sulfonic acid–induced autoimmune colitis (66). Adoptively transferred MDSCs suppressed T cell activation in vitro and decreased intestinal inflammation, IFN-γ, IL-17, and TNF-α (60). Autoimmune uveitis is induced in mice by immunization with the retina-specific Ag interphotoreceptor retinal-binding protein in CFA followed by an injection of pertussis toxin. M-MDSCs increase in these mice during and before spontaneous resolution of disease, and adoptive transfer of these M-MDSCs accelerates remission and reduces Th1 and Th17 cells. Similar increases in M-MDSCs were associated with disease remission in patients with autoimmune uveitis (61). Adoptive transfer of mesenchymal stem/stromal cells to mice with uveitis also increased MDSC levels, resulting in decreased Th1 and Th17 cells (67).

Collagen-induced arthritis (CIA) in mice is a model for human rheumatoid arthritis and is initiated by injection of type 2 xenogeneic collagen in IFA. MDSCs develop spontaneously in CIA mice by day 35 postinitiation of CIA but do not prevent disease. However, when splenic PMN-MDSCs, M-MDSCs, or total MDSCs from CIA mice are adoptively transferred into CIA mice at day 21 after CIA induction, then arthritis symptoms are reduced. MDSC production of IL-10 is essential for the therapeutic effect and acts by increasing Tregs and downregulating Th17 cells, TNF-α, IL-6, and IFN-γ (59, 63). Adoptive transfer of MDSCs in a mouse model of psoriasis diminished imiquimod-induced skin inflammation by reducing TNF-α and IFN-γ and by increasing Tregs (62). Likewise, adoptive transfer of MDSCs reduced or prevented the onset of diabetes by 60 and 70%, respectively, in a type 1 transgenic NOD mouse diabetes model (65).

Detrimental effects of MDSCs in autoimmune diseases

Although many mouse models suggest that MDSCs protect against autoimmune-induced inflammation, contrasting studies indicate that MDSCs may exacerbate autoimmunity by enhancing Th17 differentiation. In a MOG EAE study, the MDSCs suppressed T cells in vitro but lost potency when adoptively transferred and instead enhanced the differentiation of Th17 cells, increasing disease severity. Depletion of these MDSCs reduced disease severity (68, 69). In SLE patients, M-MDSCs and PMN-MDSCs correlate with disease severity and increase Th17 cell differentiation in vitro. Studies with humanized mice confirmed that SLE-induced MDSCs drive the differentiation of Th17 cells through an Arg1-dependent process (70). Other studies in MRL/lpr mice with SLE have confirmed that adoptive transfer of PMN-MDSCs impairs Treg development whereas M-MDSCs promote Th17 polarization via IL-1β (71). In a mouse CIA model MDSCs promoted Th17 differentiation through an IL-1β–dependent process (187). The MDSCs in this study were osteoclast progenitors that facilitate arthritis bone resorption. MDSCs in NOD mice decreased Th2 responses, and depletion of MDSCs improved Sjögren-like autoimmune symptoms (188).

Chronic stress can diminish cell-mediated immunity. The decrease in immunocompetence is commonly attributed to glucocorticoids and catecholamines via T cells that have receptors for these hormones, thus polarizing the T cells toward type 2 immunity. However, stress also induces IL-6, IL-1, and VEGF (189), raising the possibility that MDSCs may contribute to stress-induced immune effects.

Physical and psychological stress

Stress studies in patients are challenging because of the difficulty in evaluating the severity of stress and the multiple conditions that lead to it. In cancer, stress can accompany the period immediately following removal of primary tumor and is associated with decreased T cell function, which may contribute to metastasis. Mouse breast cancer studies indicate that removal of primary tumor increases MDSCs and enhances metastasis (72), supporting the hypothesis that metastasis increases postsurgery due to the accumulation of MDSCs. However, a clinical trial (NCT03578627) of 16 patients following breast cancer surgery indicated that circulating MDSC levels do not increase in the period immediately after surgery but are elevated in postsurgery patients experiencing additional life-induced stress factors (190).

Physical conditions such as crowding, food and/or water deprivation, and improper day/night lighting have been used to induce physical and psychological stress in animal models. In a mouse restraint model stress-induced IL-6 led to an increase in PMN-MDSCs via STAT3 signaling, an effect that was reversed by administration of β-adrenergic inhibitors (191–193).

Trauma

Clinical studies suggest that MDSC differentiation is induced following trauma-induced inflammation but indicate that the MDSCs may be beneficial in the immediate aftermath of the incident. Patients with mild to severe trauma develop high intracellular ROS and elevated levels of activated PMN-MDSCs. Because MDSCs produce the anti-inflammatory cytokine IL-10, they downregulate trauma-associated inflammation by decreasing Th17 and Th1 cells (75). Immune deficiency can occur following a severe traumatic event, and patients can succumb to infection (194) possibly via MDSC-mediated T cell suppression. In a mouse model of abdominal trauma, splenic MDSCs were elevated 6–72 h following the initiation of trauma, and T cell dysfunction ensued via an Arg1-dependent mechanism (73), supporting this hypothesis. In rat femur fracture and polytrauma models, MDSC levels peaked in the spleen 2 h after initiation of trauma, returning to normal within 6–18 h. MDSCs also increased in a mouse pseudofracture trauma model where neutralization of the DAMP HMGB1 reversed the MDSC increase and restored T cell function (74).

As laboratory mice age, hematopoiesis becomes skewed toward myelopoiesis and this is associated with increased differentiation of functional MDSCs (195). Myeloid skewing also occurs in humans (196), but whether functional MDSCs are increased with age in the absence of pathology remains unclear. The wealth of mouse data are matched by knowledge of human MDSCs in cancer, but not in healthy aging, although sparse data suggest there may be parallels with the mouse. Despite the many reviews on this topic, the marked differences between humans and mice (197) are not usually made clear. Hence, the limited data solely on human MDSCs and aging are considered here.

A much-cited seminal paper (198) reported that the mean absolute numbers of peripheral blood cells with the PMN-MDSC phenotype CD11b+CD15+CD33+HLA-DR were on average slightly but significantly higher in healthy older-versus-younger Canadians (61–76 versus 19–59 y of age) but notably higher in frail seniors (67–99 y of age), associated with higher levels of proinflammatory cytokines that may facilitate MDSC production. Hence, higher levels of cells with the phenotype of MDSCs may primarily be associated with the degree of individual frailty and “inflammaging” (199). Other investigations on MDSCs in human aging have suggested that very old Brazilians (80–100 y of age) possess a higher percentage of circulating PMN-MDSCs than do 20- to 30-y-old controls, but absolute numbers per milliliter of blood were not different in this case (200). However, suppressive capacity was not tested in either of these studies, so the cells could not be confirmed as MDSCs. As far as we are aware, there is a single paper addressing functionality, showing that bone marrow MDSCs from older donors retain their suppressive functionality (76). The papers (196, 198, 200) with quite sparse data are constantly cited in support of the contention that MDSCs increase with age in healthy humans, but the actual published evidence for this is not overwhelming. It is nonetheless highly likely that the commonly slightly enhanced systemic levels of inflammatory mediators seen in older humans, especially in frailty, coupled with the likely myeloid skewing of hematopoiesis contribute to higher levels of immunosuppression by MDSCs in older adults as a further negative impact of inflammaging, also associated with poorer responses to vaccination. As we have argued before (201), there are few data on the numbers, types, and functions of MDSCs in older humans. However, the much larger datasets in the clinical context are consistent with their important role in healthy aging.

Perhaps surprisingly, there are few studies on the impact of MDSCs on either prophylactic or therapeutic vaccination, but most of the available data suggest that MDSCs reduce vaccine efficacy. Vaccination itself induces a local inflammatory reaction and MDSCs. Surprisingly, multiple doses of SARS-CoV-2 vaccines do not induce MDSCs (202), perhaps due to the unique lipid composition of the vaccines. In mice, bacillus Calmette–Guérin recruits immune-suppressive M-MDSCs that impair T cell priming in the draining lymph node (77). In rhesus macaques immunized with an mRNA influenza vaccine, M-MDSCs and PMN-MDSCs invade the injection site but not the draining lymph nodes (78), suggesting that vaccine immune responsiveness may not always be affected. This possibility is supported by a study of South African infants up to 1 y of age that showed no correlation between MDSC levels and responsiveness to common childhood vaccines; however, the levels of MDSCs were only <1% at 6 wk of age and decreased thereafter (203). Elevated levels of MDSCs that occur in diet-induced obese mice inhibit Ab production and T cell proliferation to a hepatitis B vaccine (79). MDSCs play a unique role in HIV vaccines as shown in a mouse model using EcoHIV infection. Initial infection produces HIV-1 GAG-specific CD8+ T cells that reduce virally infected cells. However, within 7 d CD8+ T cells are rapidly reduced and PD-L1–expressing HIV-infected MDSCs appear and persist (80), potentially reducing vaccine efficacy and duration of protection.

In disease-prevalent low- and middle-income countries, individuals in endemic areas of parasite infection may have infection-induced MDSCs, and vaccine design for use in such countries needs to consider MDSC immune suppressive activity regardless of vaccine target (81). Therefore, vaccine design against parasitic infection in these areas needs to consider MDSC immune-suppressive activity. Indeed, in a mouse model of Chagas disease, depletion of MDSCs enhanced the efficacy of an anti–T. cruzi vaccine (82).

Along similar lines, therapeutic vaccines for cancer (e.g., tumor peptides, recombinant viral vector vaccines, DC-based vaccines) must consider the presence of existing MDSCs, given that MDSCs impair DC function (204). In an ovarian cancer mouse model, a prime/boost vaccine strategy with an Ag-armed oncolytic virus combined with depletion of MDSCs and PD-1 checkpoint blockade was most efficacious in restoring antitumor CD8+ T cell activity (83). Immunization with a viral vector–based cancer vaccine targeting a mouse human papillomavirus (HPV) tumor similarly resulted in optimal activation of HPV E7-specific CD8+ T cells and maximal mouse survival when combined with sunitinib to deplete MDSCs (84). In a clinical trial of small-cell lung cancer patients (NCT00617409), 41.7% of patients given DC-transduced with wild-type p53 in combination with all-trans retinoic acid to deplete MDSCs developed p53-reactive CD8+ T cells compared with 20% of patients receiving only the vaccine (85).

The presence of MDSCs in many diseases and nonpathological physiological conditions has made them an obvious target for intervention. In diseases where they are detrimental, strategies are being developed to eliminate them. In conditions where they are beneficial, MDSCs are being exploited as therapeutic agents. For example, in pregnancy, MDSCs facilitate maternal–fetal tolerance and could be induced or adoptively transferred to enhance in vitro fertilization and successful pregnancies.

Where MDSCs exhibit a dual protective/detrimental role it is unclear whether they should be targeted. Their protective effects in reducing metabolic dysfunction in obesity are countered by their association with increased cancer risk. Likewise, MDSCs provide protection against excessive inflammation during acute sepsis, but they contribute to postsepsis immune suppression. MDSCs may also play a dual role in immunity to infections in that their anti-inflammatory activity prevents excessive inflammation while impairing cell-mediated immune mechanisms for clearing pathogens. The same situation accompanies normal aging where MDSCs could temper inflammaging but at the cost of depressing immunity to infection. Similarly, the immune-suppressive potency of MDSCs makes them a prime cell for controlling GVHD in transplant patients, and multiple mouse studies have shown that adoptive transfer of large quantities of MDSCs can produce disease remission in autoimmune conditions. However, this strategy has the potential to increase the risk of infection in the recipients.

In pathologic situations such as cancer where MDSCs are exclusively detrimental, therapeutic strategies to eliminate them are already benefiting patients in clinical trials. However, for situations in which MDSCs play a dual beneficial and detrimental role, a comprehensive understanding of the quantity, timing, location, and specific phenotype of MDSCs is needed to intervene specifically and effectively. Given the variation in pathologies and individuals, efficacious intervention in the latter conditions is likely to require customization.

The authors have no financial conflicts of interest

This work was supported by the National Cancer Institute Grant R01CA115880 and General Medical Sciences Grant R01GM021248.

alloHCT

allogeneic hematopoietic stem cell transplantation

Arg1

arginase 1

Breg

B regulatory cell

CIA

collagen-induced arthritis

DC

dendritic cell

EAE

experimental autoimmune encephalomyelitis

FAO

fatty acid oxidation

FATP

fatty acid transport protein

GVHD

graft-versus-host disease

GVL

graft-versus-leukemia

HFD

high-fat diet

HPV

human papillomavirus

MDSC

myeloid-derived suppressor cell

M-MDSC

monocytic MDSC

MOG

myelin oligodendrocyte glycoprotein

MS

multiple sclerosis

NOS2

NO synthase 2

PMN-MDSC

polymorphonuclear or granulocytic MDSC

ROS

reactive oxygen species

SLE

systemic lupus erythematosus

TME

tumor microenvironment

Treg

T regulatory cell

1
Grover
,
A.
,
E.
Sanseviero
,
E.
Timosenko
,
D. I.
Gabrilovich
.
2021
.
Myeloid-derived suppressor cells: a propitious road to clinic
.
Cancer Discov.
11
:
2693
2706
.
2
Ostrand-Rosenberg
,
S.
2021
.
Myeloid-derived suppressor cells: facilitators of cancer and obesity-induced cancer
.
Annu. Rev. Cancer Biol.
5
:
17
38
.
3
Ostrand-Rosenberg
,
S.
,
C.
Fenselau
.
2018
.
Myeloid-derived suppressor cells: immune-suppressive cells that impair antitumor immunity and are sculpted by their environment
.
J. Immunol.
200
:
422
431
.
4
Tcyganov
,
E.
,
J.
Mastio
,
E.
Chen
,
D. I.
Gabrilovich
.
2018
.
Plasticity of myeloid-derived suppressor cells in cancer
.
Curr. Opin. Immunol.
51
:
76
82
.
5
Veglia
,
F.
,
E.
Sanseviero
,
D. I.
Gabrilovich
.
2021
.
Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity
.
Nat. Rev. Immunol.
21
:
485
498
.
6
Dietrich
,
O.
,
A.
Heinz
,
O.
Goldmann
,
R.
Geffers
,
A.
Beineke
,
K.
Hiller
,
A. E.
Saliba
,
E.
Medina
.
2022
.
Dysregulated immunometabolism is associated with the generation of myeloid-derived suppressor cells in Staphylococcus aureus chronic infection
.
J. Innate Immun.
14
:
257
274
.
7
Heim
,
C. E.
,
D.
Vidlak
,
T.
Kielian
.
2015
.
Interleukin-10 production by myeloid-derived suppressor cells contributes to bacterial persistence during Staphylococcus aureus orthopedic biofilm infection
.
J. Leukoc. Biol.
98
:
1003
1013
.
8
du Plessis
,
N.
,
L.
Loebenberg
,
M.
Kriel
,
F.
von Groote-Bidlingmaier
,
E.
Ribechini
,
A. G.
Loxton
,
P. D.
van Helden
,
M. B.
Lutz
,
G.
Walzl
.
2013
.
Increased frequency of myeloid-derived suppressor cells during active tuberculosis and after recent Mycobacterium tuberculosis infection suppresses T-cell function
.
Am. J. Respir. Crit. Care Med.
188
:
724
732
.
9
Singh
,
V. K.
,
A.
Khan
,
Y.
Xu
,
S.
Mai
,
L.
Zhang
,
A.
Mishra
,
B. I.
Restrepo
,
P. Y.
Pan
,
S. H.
Chen
,
C.
Jagannath
.
2022
.
Antibody-mediated LILRB2-receptor antagonism induces human myeloid-derived suppressor cells to kill Mycobacterium tuberculosis
.
Front. Immunol.
13
:
865503
.
10
Peñaloza
,
H. F.
,
L. P.
Noguera
,
D.
Ahn
,
O. P.
Vallejos
,
R. M.
Castellanos
,
Y.
Vazquez
,
F. J.
Salazar-Echegarai
,
L.
González
,
I.
Suazo
,
C.
Pardo-Roa
, et al
.
2019
.
Interleukin-10 produced by myeloid-derived suppressor cells provides protection to carbapenem-resistant Klebsiella pneumoniae sequence type 258 by enhancing its clearance in the airways
.
Infect. Immun.
87
:
e00665-18
https://doi.org/10.1128/IAI.00665-18.
11
Schrijver
,
I. T.
,
E.
Karakike
,
C.
Théroude
,
P.
Baumgartner
,
A.
Harari
,
E. J.
Giamarellos-Bourboulis
,
T.
Calandra
,
T.
Roger
.
2022
.
High levels of monocytic myeloid-derived suppressor cells are associated with favorable outcome in patients with pneumonia and sepsis with multi-organ failure
.
Intensive Care Med. Exp.
10
:
5
.
12
Huang
,
A.
,
B.
Zhang
,
W.
Yan
,
B.
Wang
,
H.
Wei
,
F.
Zhang
,
L.
Wu
,
K.
Fan
,
Y.
Guo
.
2014
.
Myeloid-derived suppressor cells regulate immune response in patients with chronic hepatitis B virus infection through PD-1-induced IL-10
.
J. Immunol.
193
:
5461
5469
.
13
Pal
,
S.
,
M.
Nandi
,
D.
Dey
,
B. C.
Chakraborty
,
A.
Shil
,
S.
Ghosh
,
S.
Banerjee
,
A.
Santra
,
S. K. M.
Ahammed
,
A.
Chowdhury
,
S.
Datta
.
2019
.
Myeloid-derived suppressor cells induce regulatory T cells in chronically HBV infected patients with high levels of hepatitis B surface antigen and persist after antiviral therapy
.
Aliment. Pharmacol. Ther.
49
:
1346
1359
.
14
Ren
,
J. P.
,
J.
Zhao
,
J.
Dai
,
J. W.
Griffin
,
L.
Wang
,
X. Y.
Wu
,
Z. D.
Morrison
,
G. Y.
Li
,
M.
El Gazzar
,
S. B.
Ning
, et al
.
2016
.
Hepatitis C virus-induced myeloid-derived suppressor cells regulate T-cell differentiation and function via the signal transducer and activator of transcription 3 pathway
.
Immunology
148
:
377
386
.
15
Tacke
,
R. S.
,
H. C.
Lee
,
C.
Goh
,
J.
Courtney
,
S. J.
Polyak
,
H. R.
Rosen
,
Y. S.
Hahn
.
2012
.
Myeloid suppressor cells induced by hepatitis C virus suppress T-cell responses through the production of reactive oxygen species
.
Hepatology
55
:
343
353
.
16
Garg
,
A.
,
S. A.
Spector
.
2014
.
HIV type 1 gp120-induced expansion of myeloid derived suppressor cells is dependent on interleukin 6 and suppresses immunity
.
J. Infect. Dis.
209
:
441
451
.
17
Singh
,
R.
,
M.
Chakraborty
,
A.
Gautam
,
S. K.
Roy
,
I.
Halder
,
J.
Barber
,
A.
Garg
.
2021
.
Residual immune activation in HIV-Infected individuals expands monocytic-myeloid derived suppressor cells
.
Cell. Immunol.
362
:
104304
.
18
Vollbrecht
,
T.
,
R.
Stirner
,
A.
Tufman
,
J.
Roider
,
R. M.
Huber
,
J. R.
Bogner
,
A.
Lechner
,
C.
Bourquin
,
R.
Draenert
.
2012
.
Chronic progressive HIV-1 infection is associated with elevated levels of myeloid-derived suppressor cells
.
AIDS
26
:
F31
F37
.
19
Sacchi
,
A.
,
G.
Grassi
,
V.
Bordoni
,
P.
Lorenzini
,
E.
Cimini
,
R.
Casetti
,
E.
Tartaglia
,
L.
Marchioni
,
N.
Petrosillo
,
F.
Palmieri
, et al
.
2020
.
Early expansion of myeloid-derived suppressor cells inhibits SARS-CoV-2 specific T-cell response and may predict fatal COVID-19 outcome
.
Cell Death Dis.
11
:
921
.
20
Reizine
,
F.
,
M.
Lesouhaitier
,
M.
Gregoire
,
K.
Pinceaux
,
A.
Gacouin
,
A.
Maamar
,
B.
Painvin
,
C.
Camus
,
Y.
Le Tulzo
,
P.
Tattevin
, et al
.
2021
.
SARS-CoV-2-induced ARDS associates with MDSC expansion, lymphocyte dysfunction, and arginine shortage
.
J. Clin. Immunol.
41
:
515
525
.
21
Arocena
,
A. R.
,
L. I.
Onofrio
,
A. V.
Pellegrini
,
A. E.
Carrera Silva
,
A.
Paroli
,
R. C.
Cano
,
M. P.
Aoki
,
S.
Gea
.
2014
.
Myeloid-derived suppressor cells are key players in the resolution of inflammation during a model of acute infection
.
Eur. J. Immunol.
44
:
184
194
.
22
Fresno
,
M.
,
N.
Gironès
.
2021
.
Myeloid-derived suppressor cells in Trypanosoma cruzi infection
.
Front. Cell. Infect. Microbiol.
11
:
737364
.
23
Bandyopadhyay
,
S.
,
A.
Bhattacharjee
,
S.
Banerjee
,
K.
Halder
,
S.
Das
,
B.
Paul Chowdhury
,
S.
Majumdar
.
2015
.
Glycyrrhizic acid-mediated subdual of myeloid-derived suppressor cells induces antileishmanial immune responses in a susceptible host
.
Infect. Immun.
83
:
4476
4486
.
24
Pereira
,
W. F.
,
F. L.
Ribeiro-Gomes
,
L. V.
Guillermo
,
N. S.
Vellozo
,
F.
Montalvão
,
G. A.
Dosreis
,
M. F.
Lopes
.
2011
.
Myeloid-derived suppressor cells help protective immunity to Leishmania major infection despite suppressed T cell responses
.
J. Leukoc. Biol.
90
:
1191
1197
.
25
Flora
,
R.
,
S.
Aghazadeh-Dibavar
,
M.
Bandyopadhyay
,
S.
Dasgupta
.
2014
.
Immunosuppression during Leishmania donovani infection: a potential target for the development of therapy
.
Ann. Parasitol.
60
:
239
245
.
26
Lamsfus Calle
,
C.
,
R.
Fendel
,
A.
Singh
,
T. L.
Richie
,
S. L.
Hoffman
,
P. G.
Kremsner
,
B.
Mordmüller
.
2021
.
Expansion of functional myeloid-derived suppressor cells in controlled human malaria infection
.
Front. Immunol.
12
:
625712
.
27
Mukherjee
,
S.
,
S.
Ghosh
,
A.
Sengupta
,
S.
Sarkar
,
T.
Keswani
,
R.
Chatterjee
,
A.
Bhattacharyya
.
2022
.
IL-6 dependent expansion of inflammatory MDSCs (CD11b+ Gr-1+) promote Th-17 mediated immune response during experimental cerebral malaria
.
Cytokine
155
:
155910
.
28
Zhang
,
Y.
,
Y.
Wu
,
H.
Liu
,
W.
Gong
,
Y.
Hu
,
Y.
Shen
,
J.
Cao
.
2021
.
Granulocytic myeloid-derived suppressor cells inhibit T follicular helper cells during experimental Schistosoma japonicum infection
.
Parasit. Vectors
14
:
497
.
29
Zhou
,
X.
,
W.
Wang
,
F.
Cui
,
C.
Shi
,
X.
Gao
,
J.
Ouyang
,
Y.
Wang
,
A.
Nagai
,
W.
Zhao
,
M.
Yin
,
J.
Zhao
.
2020
.
Myeloid-derived suppressor cells exert immunosuppressive function on the T helper 2 in mice infected with Echinococcus granulosus
.
Exp. Parasitol.
215
:
107917
.
30
Hollen
,
M. K.
,
J. A.
Stortz
,
D.
Darden
,
M. L.
Dirain
,
D. C.
Nacionales
,
R. B.
Hawkins
,
M. C.
Cox
,
M. C.
Lopez
,
J. C.
Rincon
,
R.
Ungaro
, et al
.
2019
.
Myeloid-derived suppressor cell function and epigenetic expression evolves over time after surgical sepsis
.
Crit. Care
23
:
355
.
31
Waeckel
,
L.
,
F.
Venet
,
M.
Gossez
,
C.
Monard
,
T.
Rimmelé
,
G.
Monneret
.
2020
.
Delayed persistence of elevated monocytic MDSC associates with deleterious outcomes in septic shock: a retrospective cohort study
.
Crit. Care
24
:
132
.
32
Malavika
,
M.
,
S.
Sanju
,
M. R.
Poorna
,
V.
Vishnu Priya
,
N.
Sidharthan
,
P.
Varma
,
U.
Mony
.
2022
.
Role of myeloid derived suppressor cells in sepsis
.
Int. Immunopharmacol.
104
:
108452
.
33
Clements
,
V. K.
,
T.
Long
,
R.
Long
,
C.
Figley
,
D. M. C.
Smith
,
S.
Ostrand-Rosenberg
.
2018
.
Frontline science: high fat diet and leptin promote tumor progression by inducing myeloid-derived suppressor cells
.
J. Leukoc. Biol.
103
:
395
407
.
34
Xia
,
S.
,
H.
Sha
,
L.
Yang
,
Y.
Ji
,
S.
Ostrand-Rosenberg
,
L.
Qi
.
2011
.
Gr-1+ CD11b+ myeloid-derived suppressor cells suppress inflammation and promote insulin sensitivity in obesity
.
J. Biol. Chem.
286
:
23591
23599
.
35
Yang
,
Q.
,
B.
Yu
,
J.
Kang
,
A.
Li
,
J.
Sun
.
2021
.
Obesity promotes tumor immune evasion in ovarian cancer through increased production of myeloid-derived suppressor cells via IL-6
.
Cancer Manag. Res.
13
:
7355
7363
.
36
Ostrand-Rosenberg
,
S.
2018
.
Myeloid derived-suppressor cells: their role in cancer and obesity
.
Curr. Opin. Immunol.
51
:
68
75
.
37
Hale
,
M.
,
F.
Itani
,
C. M.
Buchta
,
G.
Wald
,
M.
Bing
,
L. A.
Norian
.
2015
.
Obesity triggers enhanced MDSC accumulation in murine renal tumors via elevated local production of CCL2
.
PLoS One
10
:
e0118784
.
38
Köstlin-Gille
,
N.
,
S.
Dietz
,
J.
Schwarz
,
B.
Spring
,
J.
Pauluschke-Fröhlich
,
C. F.
Poets
,
C.
Gille
.
2019
.
HIF-1α-deficiency in myeloid cells leads to a disturbed accumulation of myeloid derived suppressor cells (MDSC) during pregnancy and to an increased abortion rate in mice
.
Front. Immunol.
10
:
161
.
39
Ostrand-Rosenberg
,
S.
,
P.
Sinha
,
C.
Figley
,
R.
Long
,
D.
Park
,
D.
Carter
,
V. K.
Clements
.
2017
.
Frontline science: myeloid-derived suppressor cells (MDSCs) facilitate maternal-fetal tolerance in mice
.
J. Leukoc. Biol.
101
:
1091
1101
.
40
Pan
,
T.
,
Y.
Liu
,
L. M.
Zhong
,
M. H.
Shi
,
X. B.
Duan
,
K.
Wu
,
Q.
Yang
,
C.
Liu
,
J. Y.
Wei
,
X. R.
Ma
, et al
.
2016
.
Myeloid-derived suppressor cells are essential for maintaining feto-maternal immunotolerance via STAT3 signaling in mice
.
J. Leukoc. Biol.
100
:
499
511
.
41
Zhao
,
H.
,
F.
Kalish
,
S.
Schulz
,
Y.
Yang
,
R. J.
Wong
,
D. K.
Stevenson
.
2015
.
Unique roles of infiltrating myeloid cells in the murine uterus during early to midpregnancy
.
J. Immunol.
194
:
3713
3722
.
42
Dietz
,
S.
,
J.
Schwarz
,
A.
Velic
,
I.
González-Menéndez
,
L.
Quintanilla-Martinez
,
N.
Casadei
,
A.
Marmé
,
C. F.
Poets
,
C.
Gille
,
N.
Köstlin-Gille
.
2022
.
Human leucocyte antigen G and murine Qa-2 are critical for myeloid derived suppressor cell expansion and activation and for successful pregnancy outcome
.
Front. Immunol.
12
:
787468
.
43
Hu
,
C.
,
Y.
Zhen
,
B.
Pang
,
X.
Lin
,
H.
Yi
.
2019
.
Myeloid-derived suppressor cells are regulated by estradiol and are a predictive marker for IVF outcome
.
Front. Endocrinol. (Lausanne)
10
:
521
.
44
Köstlin
,
N.
,
A. L.
Ostermeir
,
B.
Spring
,
J.
Schwarz
,
A.
Marmé
,
C. B.
Walter
,
C. F.
Poets
,
C.
Gille
.
2017
.
HLA-G promotes myeloid-derived suppressor cell accumulation and suppressive activity during human pregnancy through engagement of the receptor ILT4
.
Eur. J. Immunol.
47
:
374
384
.
45
Zhu
,
M.
,
X.
Huang
,
S.
Yi
,
H.
Sun
,
J.
Zhou
.
2019
.
High granulocytic myeloid-derived suppressor cell levels in the peripheral blood predict a better IVF treatment outcome
.
J. Matern. Fetal Neonatal Med.
32
:
1092
1097
.
46
Gervassi
,
A.
,
N.
Lejarcegui
,
S.
Dross
,
A.
Jacobson
,
G.
Itaya
,
E.
Kidzeru
,
S.
Gantt
,
H.
Jaspan
,
H.
Horton
.
2014
.
Myeloid derived suppressor cells are present at high frequency in neonates and suppress in vitro T cell responses
.
PLoS One
9
:
e107816
.
47
Schwarz
,
J.
,
V.
Scheckenbach
,
H.
Kugel
,
B.
Spring
,
J.
Pagel
,
C.
Härtel
,
J.
Pauluschke-Fröhlich
,
A.
Peter
,
C. F.
Poets
,
C.
Gille
,
N.
Köstlin
.
2018
.
Granulocytic myeloid-derived suppressor cells (GR-MDSC) accumulate in cord blood of preterm infants and remain elevated during the neonatal period
.
Clin. Exp. Immunol.
191
:
328
337
.
48
He
,
Y. M.
,
X.
Li
,
M.
Perego
,
Y.
Nefedova
,
A. V.
Kossenkov
,
E. A.
Jensen
,
V.
Kagan
,
Y. F.
Liu
,
S. Y.
Fu
,
Q. J.
Ye
, et al
.
2018
.
Transitory presence of myeloid-derived suppressor cells in neonates is critical for control of inflammation
.
Nat. Med.
24
:
224
231
.
49
Liu
,
Y.
,
M.
Perego
,
Q.
Xiao
,
Y.
He
,
S.
Fu
,
J.
He
,
W.
Liu
,
X.
Li
,
Y.
Tang
,
X.
Li
, et al
.
2019
.
Lactoferrin-induced myeloid-derived suppressor cell therapy attenuates pathologic inflammatory conditions in newborn mice
.
J. Clin. Invest.
129
:
4261
4275
.
50
Fan
,
Q.
,
H.
Liu
,
X.
Liang
,
T.
Yang
,
Z.
Fan
,
F.
Huang
,
Y.
Ling
,
X.
Liao
,
L.
Xuan
,
N.
Xu
, et al
.
2017
.
Superior GVHD-free, relapse-free survival for G-BM to G-PBSC grafts is associated with higher MDSCs content in allografting for patients with acute leukemia
.
J. Hematol. Oncol.
10
:
135
.
51
Lv
,
M.
,
X. S.
Zhao
,
Y.
Hu
,
Y. J.
Chang
,
X. Y.
Zhao
,
Y.
Kong
,
X. H.
Zhang
,
L. P.
Xu
,
K. Y.
Liu
,
X. J.
Huang
.
2015
.
Monocytic and promyelocytic myeloid-derived suppressor cells may contribute to G-CSF-induced immune tolerance in haplo-identical allogeneic hematopoietic stem cell transplantation
.
Am. J. Hematol.
90
:
E9
E16
.
52
Vendramin
,
A.
,
S.
Gimondi
,
A.
Bermema
,
P.
Longoni
,
S.
Rizzitano
,
P.
Corradini
,
C.
Carniti
.
2014
.
Graft monocytic myeloid-derived suppressor cell content predicts the risk of acute graft-versus-host disease after allogeneic transplantation of granulocyte colony-stimulating factor-mobilized peripheral blood stem cells
.
Biol. Blood Marrow Transplant.
20
:
2049
2055
.
53
Luan
,
Y.
,
E.
Mosheir
,
M. C.
Menon
,
D.
Wilson
,
C.
Woytovich
,
J.
Ochando
,
B.
Murphy
.
2013
.
Monocytic myeloid-derived suppressor cells accumulate in renal transplant patients and mediate CD4+ Foxp3+ Treg expansion
.
Am. J. Transplant.
13
:
3123
3131
.
54
Oshrine
,
B.
,
P.
Innamarato
,
H.
Branthoover
,
L.
Nagle
,
P.
Verdugo
,
S.
Pilon-Thomas
,
M.
Beatty
.
2022
.
Early recovery of myeloid-derived suppressor cells after allogeneic hematopoietic transplant: comparison of post-transplantation cyclophosphamide to standard graft-versus-host disease prophylaxis
.
Transplant. Cell. Ther.
28
:
203.e1
203.e7
.
55
Yang
,
S.
,
Y.
Wei
,
R.
Sun
,
W.
Lu
,
H.
Lv
,
X.
Xiao
,
Y.
Cao
,
X.
Jin
,
M.
Zhao
.
2020
.
Umbilical cord blood-derived mesenchymal stromal cells promote myeloid-derived suppressor cell proliferation by secreting HLA-G to reduce acute graft-versus-host disease after hematopoietic stem cell transplantation
.
Cytotherapy
22
:
718
733
.
56
Highfill
,
S. L.
,
P. C.
Rodriguez
,
Q.
Zhou
,
C. A.
Goetz
,
B. H.
Koehn
,
R.
Veenstra
,
P. A.
Taylor
,
A.
Panoskaltsis-Mortari
,
J. S.
Serody
,
D. H.
Munn
, et al
.
2010
.
Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13
.
Blood
116
:
5738
5747
.
57
Zhang
,
J.
,
H. M.
Chen
,
G.
Ma
,
Z.
Zhou
,
D.
Raulet
,
A. L.
Rivera
,
S. H.
Chen
,
P. Y.
Pan
.
2019
.
The mechanistic study behind suppression of GVHD while retaining GVL activities by myeloid-derived suppressor cells
.
Leukemia
33
:
2078
2089
.
58
Ioannou
,
M.
,
T.
Alissafi
,
I.
Lazaridis
,
G.
Deraos
,
J.
Matsoukas
,
A.
Gravanis
,
V.
Mastorodemos
,
A.
Plaitakis
,
A.
Sharpe
,
D.
Boumpas
,
P.
Verginis
.
2012
.
Crucial role of granulocytic myeloid-derived suppressor cells in the regulation of central nervous system autoimmune disease
.
J. Immunol.
188
:
1136
1146
.
59
Fujii
,
W.
,
E.
Ashihara
,
H.
Hirai
,
H.
Nagahara
,
N.
Kajitani
,
K.
Fujioka
,
K.
Murakami
,
T.
Seno
,
A.
Yamamoto
,
H.
Ishino
, et al
.
2013
.
Myeloid-derived suppressor cells play crucial roles in the regulation of mouse collagen-induced arthritis
.
J. Immunol.
191
:
1073
1081
.
60
Guan
,
Q.
,
S.
Moreno
,
G.
Qing
,
C. R.
Weiss
,
L.
Lu
,
C. N.
Bernstein
,
R. J.
Warrington
,
Y.
Ma
,
Z.
Peng
.
2013
.
The role and potential therapeutic application of myeloid-derived suppressor cells in TNBS-induced colitis
.
J. Leukoc. Biol.
94
:
803
811
.
61
Jeong
,
H. J.
,
H. J.
Lee
,
J. H.
Ko
,
B. J.
Cho
,
S. Y.
Park
,
J. W.
Park
,
S. R.
Choi
,
J. W.
Heo
,
S. O.
Yoon
,
J. Y.
Oh
.
2018
.
Myeloid-derived suppressor cells mediate inflammation resolution in humans and mice with autoimmune uveoretinitis
.
J. Immunol.
200
:
1306
1315
.
62
Kim
,
C. H.
,
J. K.
Yoo
,
S. H.
Jeon
,
C. Y.
Lim
,
J. H.
Lee
,
D. B.
Koo
,
M. Y.
Park
.
2019
.
Anti-psoriatic effect of myeloid-derived suppressor cells on imiquimod-induced skin inflammation in mice
.
Scand. J. Immunol.
89
:
e12742
.
63
Park
,
M. J.
,
S. H.
Lee
,
E. K.
Kim
,
E. J.
Lee
,
J. A.
Baek
,
S. H.
Park
,
S. K.
Kwok
,
M. L.
Cho
.
2018
.
Interleukin-10 produced by myeloid-derived suppressor cells is critical for the induction of Tregs and attenuation of rheumatoid inflammation in mice
.
Sci. Rep.
8
:
3753
.
64
Park
,
M. J.
,
S. H.
Lee
,
E. K.
Kim
,
E. J.
Lee
,
S. H.
Park
,
S. K.
Kwok
,
M. L.
Cho
.
2016
.
Myeloid-derived suppressor cells induce the expansion of regulatory b cells and ameliorate autoimmunity in the sanroque mouse model of systemic lupus erythematosus
.
Arthritis Rheumatol.
68
:
2717
2727
.
65
Yin
,
B.
,
G.
Ma
,
C. Y.
Yen
,
Z.
Zhou
,
G. X.
Wang
,
C. M.
Divino
,
S.
Casares
,
S. H.
Chen
,
W. C.
Yang
,
P. Y.
Pan
.
2010
.
Myeloid-derived suppressor cells prevent type 1 diabetes in murine models
.
J. Immunol.
185
:
5828
5834
.
66
Zhang
,
R.
,
S.
Ito
,
N.
Nishio
,
Z.
Cheng
,
H.
Suzuki
,
K. I.
Isobe
.
2011
.
Dextran sulphate sodium increases splenic Gr1+CD11b+ cells which accelerate recovery from colitis following intravenous transplantation
.
Clin. Exp. Immunol.
164
:
417
427
.
67
Lee
,
H. J.
,
J. H.
Ko
,
H. J.
Jeong
,
A. Y.
Ko
,
M. K.
Kim
,
W. R.
Wee
,
S. O.
Yoon
,
J. Y.
Oh
.
2015
.
Mesenchymal stem/stromal cells protect against autoimmunity via CCL2-dependent recruitment of myeloid-derived suppressor cells
.
J. Immunol.
194
:
3634
3645
.
68
Yi
,
H.
,
C.
Guo
,
X.
Yu
,
D.
Zuo
,
X. Y.
Wang
.
2012
.
Mouse CD11b+Gr-1+ myeloid cells can promote Th17 cell differentiation and experimental autoimmune encephalomyelitis
.
J. Immunol.
189
:
4295
4304
.
69
Glenn
,
J. D.
,
C.
Liu
,
K. A.
Whartenby
.
2019
.
Frontline science: induction of experimental autoimmune encephalomyelitis mobilizes Th17-promoting myeloid derived suppressor cells to the lung
.
J. Leukoc. Biol.
105
:
829
841
.
70
Wu
,
H.
,
Y.
Zhen
,
Z.
Ma
,
H.
Li
,
J.
Yu
,
Z. G.
Xu
,
X. Y.
Wang
,
H.
Yi
,
Y. G.
Yang
.
2016
.
Arginase-1-dependent promotion of TH17 differentiation and disease progression by MDSCs in systemic lupus erythematosus
.
Sci. Transl. Med.
8
:
331ra40
.
71
Ji
,
J.
,
J.
Xu
,
S.
Zhao
,
F.
Liu
,
J.
Qi
,
Y.
Song
,
J.
Ren
,
T.
Wang
,
H.
Dou
,
Y.
Hou
.
2016
.
Myeloid-derived suppressor cells contribute to systemic lupus erythaematosus by regulating differentiation of Th17 cells and Tregs
.
Clin. Sci. (Lond.)
130
:
1453
1467
.
72
Ma
,
X.
,
M.
Wang
,
T.
Yin
,
Y.
Zhao
,
X.
Wei
.
2019
.
Myeloid-derived suppressor cells promote metastasis in breast cancer after the stress of operative removal of the primary cancer
.
Front. Oncol.
9
:
855
.
73
Makarenkova
,
V. P.
,
V.
Bansal
,
B. M.
Matta
,
L. A.
Perez
,
J. B.
Ochoa
.
2006
.
CD11b+/Gr-1+ myeloid suppressor cells cause T cell dysfunction after traumatic stress
.
J. Immunol.
176
:
2085
2094
.
74
Ruan
,
X.
,
S. S.
Darwiche
,
C.
Cai
,
M. J.
Scott
,
H. C.
Pape
,
T. R.
Billiar
.
2015
.
Anti-HMGB1 monoclonal antibody ameliorates immunosuppression after peripheral tissue trauma: attenuated T-lymphocyte response and increased splenic CD11b+Gr-1+ myeloid-derived suppressor cells require HMGB1
.
Mediators Inflamm.
2015
:
458626
.
75
Li
,
X.
,
J.
Liu
,
Z.
Xing
,
J.
Tang
,
H.
Sun
,
X.
Zhang
,
S.
Lv
,
Z.
Chen
,
M.
Shi
,
M.
Chen
, et al
.
2021
.
Polymorphonuclear myeloid-derived suppressor cells link inflammation and damage response after trauma
.
J. Leukoc. Biol.
110
:
1143
1161
.
76
Magri
,
S.
,
E.
Masetto
,
S.
Solito
,
S.
Francescato
,
E.
Belluzzi
,
A.
Pozzuoli
,
A.
Berizzi
,
P.
Ruggieri
,
S.
Mandruzzato
.
2020
.
Human MDSCs derived from the bone marrow maintain their functional ability but have a reduced frequency of induction in the elderly compared to pediatric donors. [Published erratum appears in 2020 Immun. Ageing 17: 39.]
Immun. Ageing
17
:
27
.
77
Martino
,
A.
,
E.
Badell
,
V.
Abadie
,
V.
Balloy
,
M.
Chignard
,
M. Y.
Mistou
,
B.
Combadière
,
C.
Combadière
,
N.
Winter
.
2010
.
Mycobacterium bovis bacillus Calmette-Guérin vaccination mobilizes innate myeloid-derived suppressor cells restraining in vivo T cell priming via IL-1R-dependent nitric oxide production
.
J. Immunol.
184
:
2038
2047
.
78
Lin
,
A.
,
F.
Liang
,
E. A.
Thompson
,
M.
Vono
,
S.
Ols
,
G.
Lindgren
,
K.
Hassett
,
H.
Salter
,
G.
Ciaramella
,
K.
Loré
.
2018
.
Rhesus macaque myeloid-derived suppressor cells demonstrate T cell inhibitory functions and are transiently increased after vaccination
.
J. Immunol.
200
:
286
294
.
79
Chen
,
S.
,
S. M.
Akbar
,
T.
Miyake
,
M.
Abe
,
M.
Al-Mahtab
,
S.
Furukawa
,
M.
Bunzo
,
Y.
Hiasa
,
M.
Onji
.
2015
.
Diminished immune response to vaccinations in obesity: role of myeloid-derived suppressor and other myeloid cells
.
Obes. Res. Clin. Pract.
9
:
35
44
.
80
Liu
,
L.
,
Q.
Lin
,
J.
Peng
,
J.
Fang
,
Z.
Tan
,
H.
Tang
,
K.
Kwan
,
K.
Nishiura
,
J.
Liang
,
H.
Kwok
, et al
.
2020
.
Persistent lentivirus infection induces early myeloid suppressor cells expansion to subvert protective memory CD8 T cell response
.
EBioMedicine
60
:
103008
.
81
Cabrera
,
G.
,
I.
Marcipar
.
2019
.
Vaccines and the regulatory arm of the immune system. An overview from the Trypanosoma cruzi infection model
.
Vaccine
37
:
3628
3637
.
82
Gamba
,
J. C.
,
C.
Roldán
,
E.
Prochetto
,
G.
Lupi
,
I.
Bontempi
,
C. V.
Poncini
,
M.
Vermeulen
,
A. R.
Pérez
,
I.
Marcipar
,
G.
Cabrera
.
2021
.
Targeting myeloid-derived suppressor cells to enhance a trans-sialidase-based vaccine against Trypanosoma cruzi
.
Front. Cell. Infect. Microbiol.
11
:
671104
.
83
McGray
,
A. J. R.
,
C.
Eppolito
,
A.
Miliotto
,
K. L.
Singel
,
K.
Stephenson
,
A.
Lugade
,
B. H.
Segal
,
T.
Keler
,
G.
Webster
,
B.
Lichty
, et al
.
2021
.
A prime/boost vaccine platform efficiently identifies CD27 agonism and depletion of myeloid-derived suppressor cells as therapies that rationally combine with checkpoint blockade in ovarian cancer
.
Cancer Immunol. Immunother.
70
:
3451
3460
.
84
Draghiciu
,
O.
,
H. W.
Nijman
,
B. N.
Hoogeboom
,
T.
Meijerhof
,
T.
Daemen
.
2015
.
Sunitinib depletes myeloid-derived suppressor cells and synergizes with a cancer vaccine to enhance antigen-specific immune responses and tumor eradication
.
OncoImmunology
4
:
e989764
.
85
Iclozan
,
C.
,
S.
Antonia
,
A.
Chiappori
,
D. T.
Chen
,
D.
Gabrilovich
.
2013
.
Therapeutic regulation of myeloid-derived suppressor cells and immune response to cancer vaccine in patients with extensive stage small cell lung cancer
.
Cancer Immunol. Immunother.
62
:
909
918
.
86
Bronte
,
V.
,
S.
Brandau
,
S. H.
Chen
,
M. P.
Colombo
,
A. B.
Frey
,
T. F.
Greten
,
S.
Mandruzzato
,
P. J.
Murray
,
A.
Ochoa
,
S.
Ostrand-Rosenberg
, et al
.
2016
.
Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards
.
Nat. Commun.
7
:
12150
.
87
Iannone
,
R.
,
L.
Miele
,
P.
Maiolino
,
A.
Pinto
,
S.
Morello
.
2013
.
Blockade of A2b adenosine receptor reduces tumor growth and immune suppression mediated by myeloid-derived suppressor cells in a mouse model of melanoma
.
Neoplasia
15
:
1400
1409
.
88
Yan
,
D.
,
Q.
Yang
,
M.
Shi
,
L.
Zhong
,
C.
Wu
,
T.
Meng
,
H.
Yin
,
J.
Zhou
.
2013
.
Polyunsaturated fatty acids promote the expansion of myeloid-derived suppressor cells by activating the JAK/STAT3 pathway
.
Eur. J. Immunol.
43
:
2943
2955
.
89
Hossain
,
F.
,
A. A.
Al-Khami
,
D.
Wyczechowska
,
C.
Hernandez
,
L.
Zheng
,
K.
Reiss
,
L. D.
Valle
,
J.
Trillo-Tinoco
,
T.
Maj
,
W.
Zou
, et al
.
2015
.
Inhibition of fatty acid oxidation modulates immunosuppressive functions of myeloid-derived suppressor cells and enhances cancer therapies
.
Cancer Immunol. Res.
3
:
1236
1247
.
90
Adeshakin
,
A. O.
,
W.
Liu
,
F. O.
Adeshakin
,
L. O.
Afolabi
,
M.
Zhang
,
G.
Zhang
,
L.
Wang
,
Z.
Li
,
L.
Lin
,
Q.
Cao
, et al
.
2021
.
Regulation of ROS in myeloid-derived suppressor cells through targeting fatty acid transport protein 2 enhanced anti-PD-L1 tumor immunotherapy
.
Cell. Immunol.
362
:
104286
.
91
Al-Khami
,
A. A.
,
L.
Zheng
,
L.
Del Valle
,
F.
Hossain
,
D.
Wyczechowska
,
J.
Zabaleta
,
M. D.
Sanchez
,
M. J.
Dean
,
P. C.
Rodriguez
,
A. C.
Ochoa
.
2017
.
Exogenous lipid uptake induces metabolic and functional reprogramming of tumor-associated myeloid-derived suppressor cells
.
OncoImmunology
6
:
e1344804
.
92
Veglia
,
F.
,
V. A.
Tyurin
,
M.
Blasi
,
A.
De Leo
,
A. V.
Kossenkov
,
L.
Donthireddy
,
T. K. J.
To
,
Z.
Schug
,
S.
Basu
,
F.
Wang
, et al
.
2019
.
Fatty acid transport protein 2 reprograms neutrophils in cancer
.
Nature
569
:
73
78
.
93
Sinha
,
P.
,
V. K.
Clements
,
A. M.
Fulton
,
S.
Ostrand-Rosenberg
.
2007
.
Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells
.
Cancer Res.
67
:
4507
4513
.
94
Rodriguez
,
P. C.
,
C. P.
Hernandez
,
D.
Quiceno
,
S. M.
Dubinett
,
J.
Zabaleta
,
J. B.
Ochoa
,
J.
Gilbert
,
A. C.
Ochoa
.
2005
.
Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma
.
J. Exp. Med.
202
:
931
939
.
95
Danelli
,
L.
,
B.
Frossi
,
G.
Gri
,
F.
Mion
,
C.
Guarnotta
,
L.
Bongiovanni
,
C.
Tripodo
,
L.
Mariuzzi
,
S.
Marzinotto
,
A.
Rigoni
, et al
.
2015
.
Mast cells boost myeloid-derived suppressor cell activity and contribute to the development of tumor-favoring microenvironment
.
Cancer Immunol. Res.
3
:
85
95
.
96
Baumann
,
T.
,
A.
Dunkel
,
C.
Schmid
,
S.
Schmitt
,
M.
Hiltensperger
,
K.
Lohr
,
V.
Laketa
,
S.
Donakonda
,
U.
Ahting
,
B.
Lorenz-Depiereux
, et al
.
2020
.
Regulatory myeloid cells paralyze T cells through cell-cell transfer of the metabolite methylglyoxal
.
Nat. Immunol.
21
:
555
566
.
97
Husain
,
Z.
,
Y.
Huang
,
P.
Seth
,
V. P.
Sukhatme
.
2013
.
Tumor-derived lactate modifies antitumor immune response: effect on myeloid-derived suppressor cells and NK cells
.
J. Immunol.
191
:
1486
1495
.
98
Bronte
,
V.
,
P.
Zanovello
.
2005
.
Regulation of immune responses by l-arginine metabolism
.
Nat. Rev. Immunol.
5
:
641
654
.
99
Srivastava
,
M. K.
,
P.
Sinha
,
V. K.
Clements
,
P.
Rodriguez
,
S.
Ostrand-Rosenberg
.
2010
.
Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine
.
Cancer Res.
70
:
68
77
.
100
Yu
,
J.
,
W.
Du
,
F.
Yan
,
Y.
Wang
,
H.
Li
,
S.
Cao
,
W.
Yu
,
C.
Shen
,
J.
Liu
,
X.
Ren
.
2013
.
Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer
.
J. Immunol.
190
:
3783
3797
.
101
Molon
,
B.
,
S.
Ugel
,
F.
Del Pozzo
,
C.
Soldani
,
S.
Zilio
,
D.
Avella
,
A.
De Palma
,
P.
Mauri
,
A.
Monegal
,
M.
Rescigno
, et al
.
2011
.
Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells
.
J. Exp. Med.
208
:
1949
1962
.
102
Nagaraj
,
S.
,
K.
Gupta
,
V.
Pisarev
,
L.
Kinarsky
,
S.
Sherman
,
L.
Kang
,
D. L.
Herber
,
J.
Schneck
,
D. I.
Gabrilovich
.
2007
.
Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer
.
Nat. Med.
13
:
828
835
.
103
Beury
,
D. W.
,
K. A.
Carter
,
C.
Nelson
,
P.
Sinha
,
E.
Hanson
,
M.
Nyandjo
,
P. J.
Fitzgerald
,
A.
Majeed
,
N.
Wali
,
S.
Ostrand-Rosenberg
.
2016
.
Myeloid-derived suppressor cell survival and function are regulated by the transcription factor Nrf2
.
J. Immunol.
196
:
3470
3478
.
104
Hanson
,
E. M.
,
V. K.
Clements
,
P.
Sinha
,
D.
Ilkovitch
,
S.
Ostrand-Rosenberg
.
2009
.
Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells
.
J. Immunol.
183
:
937
944
.
105
Ku
,
A. W.
,
J. B.
Muhitch
,
C. A.
Powers
,
M.
Diehl
,
M.
Kim
,
D. T.
Fisher
,
A. P.
Sharda
,
V. K.
Clements
,
K.
O’Loughlin
,
H.
Minderman
, et al
.
2016
.
Tumor-induced MDSC act via remote control to inhibit L-selectin-dependent adaptive immunity in lymph nodes
.
eLife
5
:
e17375
.
106
Huang
,
B.
,
P. Y.
Pan
,
Q.
Li
,
A. I.
Sato
,
D. E.
Levy
,
J.
Bromberg
,
C. M.
Divino
,
S. H.
Chen
.
2006
.
Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host
.
Cancer Res.
66
:
1123
1131
.
107
Lu
,
C.
,
P. S.
Redd
,
J. R.
Lee
,
N.
Savage
,
K.
Liu
.
2016
.
The expression profiles and regulation of PD-L1 in tumor-induced myeloid-derived suppressor cells
.
OncoImmunology
5
:
e1247135
.
108
Dardalhon
,
V.
,
A. C.
Anderson
,
J.
Karman
,
L.
Apetoh
,
R.
Chandwaskar
,
D. H.
Lee
,
M.
Cornejo
,
N.
Nishi
,
A.
Yamauchi
,
F. J.
Quintana
, et al
.
2010
.
Tim-3/galectin-9 pathway: regulation of Th1 immunity through promotion of CD11b+Ly-6G+ myeloid cells
.
J. Immunol.
185
:
1383
1392
.
109
Sinha
,
P.
,
O.
Chornoguz
,
V. K.
Clements
,
K. A.
Artemenko
,
R. A.
Zubarev
,
S.
Ostrand-Rosenberg
.
2011
.
Myeloid-derived suppressor cells express the death receptor Fas and apoptose in response to T cell-expressed FasL
.
Blood
117
:
5381
5390
.
110
Cheng
,
P.
,
C. A.
Corzo
,
N.
Luetteke
,
B.
Yu
,
S.
Nagaraj
,
M. M.
Bui
,
M.
Ortiz
,
W.
Nacken
,
C.
Sorg
,
T.
Vogl
, et al
.
2008
.
Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein
.
J. Exp. Med.
205
:
2235
2249
.
111
Lelis
,
F. J. N.
,
J.
Jaufmann
,
A.
Singh
,
K.
Fromm
,
A. C.
Teschner
,
S.
Pöschel
,
I.
Schäfer
,
S.
Beer-Hammer
,
N.
Rieber
,
D.
Hartl
.
2017
.
Myeloid-derived suppressor cells modulate B-cell responses
.
Immunol. Lett.
188
:
108
115
.
112
Wang
,
Y.
,
C. C.
Schafer
,
K. P.
Hough
,
S.
Tousif
,
S. R.
Duncan
,
J. F.
Kearney
,
S.
Ponnazhagan
,
H. C.
Hsu
,
J. S.
Deshane
.
2018
.
Myeloid-derived suppressor cells impair B cell responses in lung cancer through IL-7 and STAT5
.
J. Immunol.
201
:
278
295
.
113
Li
,
H.
,
Y.
Han
,
Q.
Guo
,
M.
Zhang
,
X.
Cao
.
2009
.
Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-β1
.
J. Immunol.
182
:
240
249
.
114
Sinha
,
P.
,
V. K.
Clements
,
S. K.
Bunt
,
S. M.
Albelda
,
S.
Ostrand-Rosenberg
.
2007
.
Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response
.
J. Immunol.
179
:
977
983
.
115
Burke
,
M.
,
W.
Choksawangkarn
,
N.
Edwards
,
S.
Ostrand-Rosenberg
,
C.
Fenselau
.
2014
.
Exosomes from myeloid-derived suppressor cells carry biologically active proteins
.
J. Proteome Res.
13
:
836
843
.
116
Fenselau
,
C.
,
S.
Ostrand-Rosenberg
.
2021
.
Molecular cargo in myeloid-derived suppressor cells and their exosomes
.
Cell. Immunol.
359
:
104258
.
117
Gabrilovich
,
D. I.
,
V.
Bronte
,
S. H.
Chen
,
M. P.
Colombo
,
A.
Ochoa
,
S.
Ostrand-Rosenberg
,
H.
Schreiber
.
2007
.
The terminology issue for myeloid-derived suppressor cells
.
Cancer Res.
67
:
425
.
118
Condamine
,
T.
,
G. A.
Dominguez
,
J. I.
Youn
,
A. V.
Kossenkov
,
S.
Mony
,
K.
Alicea-Torres
,
E.
Tcyganov
,
A.
Hashimoto
,
Y.
Nefedova
,
C.
Lin
, et al
.
2016
.
Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients
.
Sci. Immunol.
1
:
aaf8943
.
119
Kwak
,
T.
,
F.
Wang
,
H.
Deng
,
T.
Condamine
,
V.
Kumar
,
M.
Perego
,
A.
Kossenkov
,
L. J.
Montaner
,
X.
Xu
,
W.
Xu
, et al
.
2020
.
Distinct populations of immune-suppressive macrophages differentiate from monocytic myeloid-derived suppressor cells in cancer
.
Cell Rep.
33
:
108571
.
120
Niedermeier
,
M.
,
B.
Reich
,
M.
Rodriguez Gomez
,
A.
Denzel
,
K.
Schmidbauer
,
N.
Göbel
,
Y.
Talke
,
F.
Schweda
,
M.
Mack
.
2009
.
CD4+ T cells control the differentiation of Gr1+ monocytes into fibrocytes
.
Proc. Natl. Acad. Sci. USA
106
:
17892
17897
.
121
Alshetaiwi
,
H.
,
N.
Pervolarakis
,
L. L.
McIntyre
,
D.
Ma
,
Q.
Nguyen
,
J. A.
Rath
,
K.
Nee
,
G.
Hernandez
,
K.
Evans
,
L.
Torosian
, et al
.
2020
.
Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics
.
Sci. Immunol.
5
:
eaay6017
.
122
Agrati
,
C.
,
A.
Sacchi
,
V.
Bordoni
,
E.
Cimini
,
S.
Notari
,
G.
Grassi
,
R.
Casetti
,
E.
Tartaglia
,
E.
Lalle
,
A.
D’Abramo
, et al
.
2020
.
Expansion of myeloid-derived suppressor cells in patients with severe coronavirus disease (COVID-19)
.
Cell Death Differ.
27
:
3196
3207
.
123
Falck-Jones
,
S.
,
S.
Vangeti
,
M.
Yu
,
R.
Falck-Jones
,
A.
Cagigi
,
I.
Badolati
,
B.
Österberg
,
M. J.
Lautenbach
,
E.
Åhlberg
,
A.
Lin
, et al
.
2021
.
Functional monocytic myeloid-derived suppressor cells increase in blood but not airways and predict COVID-19 severity
.
J. Clin. Invest.
131
:
e144734
.
124
Grassi
,
G.
,
S.
Notari
,
S.
Gili
,
V.
Bordoni
,
R.
Casetti
,
E.
Cimini
,
E.
Tartaglia
,
D.
Mariotti
,
C.
Agrati
,
A.
Sacchi
.
2022
.
Myeloid-derived suppressor cells in COVID-19: the paradox of good
.
Front. Immunol.
13
:
842949
.
125
Dean
,
M. J.
,
J. B.
Ochoa
,
M. D.
Sanchez-Pino
,
J.
Zabaleta
,
J.
Garai
,
L.
Del Valle
,
D.
Wyczechowska
,
L. B.
Baiamonte
,
P.
Philbrook
,
R.
Majumder
, et al
.
2021
.
Severe COVID-19 is characterized by an impaired type I interferon response and elevated levels of arginase producing granulocytic myeloid derived suppressor cells
.
Front. Immunol.
12
:
695972
.
126
Siemińska
,
I.
,
K.
Węglarczyk
,
M.
Surmiak
,
D.
Kurowska-Baran
,
M.
Sanak
,
M.
Siedlar
,
J.
Baran
.
2021
.
Mild and asymptomatic COVID-19 convalescents present long-term endotype of immunosuppression associated with neutrophil subsets possessing regulatory functions
.
Front. Immunol.
12
:
748097
.
127
Van Ginderachter
,
J. A.
,
A.
Beschin
,
P.
De Baetselier
,
G.
Raes
.
2010
.
Myeloid-derived suppressor cells in parasitic infections
.
Eur. J. Immunol.
40
:
2976
2985
.
128
Rieber
,
N.
,
A.
Singh
,
H.
Öz
,
M.
Carevic
,
M.
Bouzani
,
J.
Amich
,
M.
Ost
,
Z.
Ye
,
M.
Ballbach
,
I.
Schäfer
, et al
.
2015
.
Pathogenic fungi regulate immunity by inducing neutrophilic myeloid-derived suppressor cells
.
Cell Host Microbe
17
:
507
514
.
129
Lilly
,
E. A.
,
B. E.
Bender
,
S.
Esher Righi
,
P. L.
Fidel
Jr.
,
M. C.
Noverr
.
2021
.
Trained innate immunity induced by vaccination with low-virulence Candida species mediates protection against several forms of fungal sepsis via Ly6G+ Gr-1+ leukocytes
.
MBio
12
:
e0254821
.
130
Singh
,
A.
,
F.
Lelis
,
S.
Braig
,
I.
Schäfer
,
D.
Hartl
,
N.
Rieber
.
2016
.
Differential regulation of myeloid-derived suppressor cells by Candida species
.
Front. Microbiol.
7
:
1624
.
131
Li
,
Y. N.
,
Z. W.
Wang
,
F.
Li
,
L. H.
Zhou
,
Y. S.
Jiang
,
Y.
Yu
,
H. H.
Ma
,
L. P.
Zhu
,
J. M.
Qu
,
X. M.
Jia
.
2022
.
Inhibition of myeloid-derived suppressor cell arginase-1 production enhances T-cell-based immunotherapy against Cryptococcus neoformans infection
.
Nat. Commun.
13
:
4074
.
132
Cuervo
,
H.
,
N. A.
Guerrero
,
S.
Carbajosa
,
A.
Beschin
,
P.
De Baetselier
,
N.
Gironès
,
M.
Fresno
.
2011
.
Myeloid-derived suppressor cells infiltrate the heart in acute Trypanosoma cruzi infection
.
J. Immunol.
187
:
2656
2665
.
133
Silva
,
D. N.
,
B. S. F.
Souza
,
J. F.
Vasconcelos
,
C. M.
Azevedo
,
C. X. R.
Valim
,
B. D.
Paredes
,
V. P. C.
Rocha
,
G. B.
Carvalho
,
P. S.
Daltro
,
S. G.
Macambira
, et al
.
2018
.
Granulocyte-colony stimulating factor-overexpressing mesenchymal stem cells exhibit enhanced immunomodulatory actions through the recruitment of suppressor cells in experimental Chagas disease cardiomyopathy
.
Front. Immunol.
9
:
1449
.
134
Sanmarco
,
L. M.
,
L. M.
Visconti
,
N.
Eberhardt
,
M. C.
Ramello
,
N. E.
Ponce
,
N. B.
Spitale
,
M. L.
Vozza
,
G. A.
Bernardi
,
S.
Gea
,
A. R.
Minguez
,
M. P.
Aoki
.
2016
.
IL-6 improves the nitric oxide-induced cytotoxic CD8+ T cell dysfunction in human Chagas disease
.
Front. Immunol.
7
:
626
.
135
Leonardo
,
L.
,
E.
Kenangalem
,
J. R.
Poespoprodjo
,
R.
Noviyanti
,
R. N.
Price
,
N. M.
Anstey
,
G.
Minigo
,
S.
Kho
.
2022
.
Increased circulating myeloid-derived suppressor cells in vivax malaria and severe falciparum malaria
.
Malar. J.
21
:
255
.
136
Ishida
,
H.
,
C.
Matsuzaki-Moriya
,
T.
Imai
,
K.
Yanagisawa
,
Y.
Nojima
,
K.
Suzue
,
M.
Hirai
,
Y.
Iwakura
,
A.
Yoshimura
,
S.
Hamano
, et al
.
2010
.
Development of experimental cerebral malaria is independent of IL-23 and IL-17
.
Biochem. Biophys. Res. Commun.
402
:
790
795
.
137
Yang
,
Q.
,
H.
Qiu
,
H.
Xie
,
Y.
Qi
,
H.
Cha
,
J.
Qu
,
M.
Wang
,
Y.
Feng
,
X.
Ye
,
J.
Mu
,
J.
Huang
.
2017
.
A Schistosoma japonicum infection promotes the expansion of myeloid-derived suppressor cells by activating the JAK/STAT3 pathway
.
J. Immunol.
198
:
4716
4727
.
138
Pan
,
W.
,
H. J.
Zhou
,
Y. J.
Shen
,
Y.
Wang
,
Y. X.
Xu
,
Y.
Hu
,
Y. Y.
Jiang
,
Z. Y.
Yuan
,
C. E.
Ugwu
,
J. P.
Cao
.
2013
.
Surveillance on the status of immune cells after Echinnococcus granulosus protoscoleces infection in Balb/c mice
.
PLoS One
8
:
e59746
.
139
Yin
,
J. H.
,
C. S.
Liu
,
A. P.
Yu
,
J. Q.
Yao
,
Y. J.
Shen
,
J. P.
Cao
.
2018
.
Pro-angiogenic activity of monocytic-type myeloid-derived suppressor cells from Balb/C mice infected with Echinococcus granulosus and the regulatory role of miRNAs
.
Cell. Physiol. Biochem.
51
:
1207
1220
.
140
Yu
,
A.
,
Y.
Wang
,
J.
Yin
,
J.
Zhang
,
S.
Cao
,
J.
Cao
,
Y.
Shen
.
2018
.
Microarray analysis of long non-coding RNA expression profiles in monocytic myeloid-derived suppressor cells in Echinococcus granulosus-infected mice
.
Parasit. Vectors
11
:
327
.
141
Cheon
,
E. C.
,
K.
Khazaie
,
M. W.
Khan
,
M. J.
Strouch
,
S. B.
Krantz
,
J.
Phillips
,
N. R.
Blatner
,
L. M.
Hix
,
M.
Zhang
,
K. L.
Dennis
, et al
.
2011
.
Mast cell 5-lipoxygenase activity promotes intestinal polyposis in APCΔ468 mice
.
Cancer Res.
71
:
1627
1636
.
142
Yang
,
Z.
,
B.
Zhang
,
D.
Li
,
M.
Lv
,
C.
Huang
,
G. X.
Shen
,
B.
Huang
.
2010
.
Mast cells mobilize myeloid-derived suppressor cells and Treg cells in tumor microenvironment via IL-17 pathway in murine hepatocarcinoma model
.
PLoS One
5
:
e8922
.
143
Martin
,
R. K.
,
S. J.
Saleem
,
L.
Folgosa
,
H. B.
Zellner
,
S. R.
Damle
,
G. K.
Nguyen
,
J. J.
Ryan
,
H. D.
Bear
,
A. M.
Irani
,
D. H.
Conrad
.
2014
.
Mast cell histamine promotes the immunoregulatory activity of myeloid-derived suppressor cells
.
J. Leukoc. Biol.
96
:
151
159
.
144
Saleem
,
S. J.
,
R. K.
Martin
,
J. K.
Morales
,
J. L.
Sturgill
,
D. R.
Gibb
,
L.
Graham
,
H. D.
Bear
,
M. H.
Manjili
,
J. J.
Ryan
,
D. H.
Conrad
.
2012
.
Cutting edge: mast cells critically augment myeloid-derived suppressor cell activity
.
J. Immunol.
189
:
511
515
.
145
Morales
,
J. K.
,
S. J.
Saleem
,
R. K.
Martin
,
B. L.
Saunders
,
B. O.
Barnstein
,
T. W.
Faber
,
N. A.
Pullen
,
E. M.
Kolawole
,
K. B.
Brooks
,
S. K.
Norton
, et al
.
2014
.
Myeloid-derived suppressor cells enhance IgE-mediated mast cell responses
.
J. Leukoc. Biol.
95
:
643
650
.
146
Bouchery
,
T.
,
M.
Moyat
,
J.
Sotillo
,
S.
Silverstein
,
B.
Volpe
,
G.
Coakley
,
T. D.
Tsourouktsoglou
,
L.
Becker
,
K.
Shah
,
M.
Kulagin
, et al
.
2020
.
Hookworms evade host immunity by secreting a deoxyribonuclease to degrade neutrophil extracellular traps
.
Cell Host Microbe
27
:
277
289.e6
.
147
Pejler
,
G.
,
S.
Alanazi
,
M.
Grujic
,
J.
Adler
,
A. K.
Olsson
,
C. P.
Sommerhoff
,
F.
Rabelo Melo
.
2022
.
Mast cell tryptase potentiates neutrophil extracellular trap formation
.
J. Innate Immun.
14
:
433
446
.
148
Huang
,
B.
,
S.
Huang
,
X.
Chen
,
X. B.
Liu
,
Q.
Wu
,
Y.
Wang
,
X.
Li
,
K.
Li
,
H.
Gao
,
S.
Cen
, et al
.
2019
.
Activation of mast cells promote Plasmodium berghei ANKA infection in murine model
.
Front. Cell. Infect. Microbiol.
9
:
322
.
149
Furuta
,
T.
,
T.
Kikuchi
,
Y.
Iwakura
,
N.
Watanabe
.
2006
.
Protective roles of mast cells and mast cell-derived TNF in murine malaria
.
J. Immunol.
177
:
3294
3302
.
150
Huang
,
M.
,
S.
Cai
,
J.
Su
.
2019
.
The pathogenesis of sepsis and potential therapeutic targets
.
Int. J. Mol. Sci.
20
:
5376
.
151
Mathias
,
B.
,
A. L.
Delmas
,
T.
Ozrazgat-Baslanti
,
E. L.
Vanzant
,
B. E.
Szpila
,
A. M.
Mohr
,
F. A.
Moore
,
S. C.
Brakenridge
,
B. A.
Brumback
,
L. L.
Moldawer
,
P. A.
Efron
;
The Sepsis, Critical Illness Research Center Investigators
.
2017
.
Human myeloid-derived suppressor cells are associated with chronic immune suppression after severe sepsis/septic shock
.
Ann. Surg.
265
:
827
834
.
152
Rincon
,
J. C.
,
P. A.
Efron
,
L. L.
Moldawer
.
2022
.
Immunopathology of chronic critical illness in sepsis survivors: role of abnormal myelopoiesis
.
J. Leukoc. Biol.
112
:
1525
1534
.
153
Schrijver
,
I. T.
,
C.
Théroude
,
T.
Roger
.
2019
.
Myeloid-derived suppressor cells in sepsis
.
Front. Immunol.
10
:
327
.
154
Sebastian
,
A.
,
S.
Sanju
,
P.
Jain
,
V. V.
Priya
,
P. K.
Varma
,
U.
Mony
.
2021
.
Non-classical monocytes and its potential in diagnosing sepsis post cardiac surgery
.
Int. Immunopharmacol.
99
:
108037
.
155
Darden
,
D. B.
,
R.
Bacher
,
M. A.
Brusko
,
P.
Knight
,
R. B.
Hawkins
,
M. C.
Cox
,
M. L.
Dirain
,
R.
Ungaro
,
D. C.
Nacionales
,
J. C.
Rincon
, et al
.
2021
.
Single-cell RNA-seq of human myeloid-derived suppressor cells in late sepsis reveals multiple subsets with unique transcriptional responses: a pilot study
.
Shock
55
:
587
595
.
156
Baudesson de Chanville
,
C.
,
B. G.
Chousterman
,
P.
Hamon
,
M.
Laviron
,
N.
Guillou
,
P. L.
Loyher
,
A.
Meghraoui-Kheddar
,
S.
Barthelemy
,
P.
Deterre
,
A.
Boissonnas
,
C.
Combadière
.
2020
.
Sepsis triggers a late expansion of functionally impaired tissue-vascular inflammatory monocytes during clinical recovery
.
Front. Immunol.
11
:
675
.
157
Chen
,
J.
,
S.
Cai
,
R.
Li
,
J.
Xie
,
F.
Yang
,
T.
Liu
.
2022
.
Blockade of cycloxygenase-2 ameliorates sepsis induced immune-suppression by regulating myeloid-derived suppressor cells
.
Int. Immunopharmacol.
104
:
108506
.
158
Dai
,
J.
,
A.
Kumbhare
,
D.
Youssef
,
C. E.
McCall
,
M.
El Gazzar
.
2017
.
Intracellular S100A9 promotes myeloid-derived suppressor cells during late sepsis
.
Front. Immunol.
8
:
1565
.
159
Ohl
,
K.
,
A.
Fragoulis
,
P.
Klemm
,
J.
Baumeister
,
W.
Klock
,
E.
Verjans
,
S.
Böll
,
J.
Möllmann
,
M.
Lehrke
,
I.
Costa
, et al
.
2018
.
Nrf2 is a central regulator of metabolic reprogramming of myeloid-derived suppressor cells in steady state and sepsis
.
Front. Immunol.
9
:
1552
.
160
Vance
,
J. K.
,
T. W.
Rawson
,
J. M.
Povroznik
,
K. M.
Brundage
,
C. M.
Robinson
.
2021
.
Myeloid-derived suppressor cells gain suppressive function during neonatal bacterial sepsis
.
Int. J. Mol. Sci.
22
:
7047
.
161
Bao
,
Y.
,
J.
Mo
,
L.
Ruan
,
G.
Li
.
2015
.
Increased monocytic CD14+HLADRlow/− myeloid-derived suppressor cells in obesity
.
Mol. Med. Rep.
11
:
2322
2328
.
162
Friedrich
,
K.
,
M.
Sommer
,
S.
Strobel
,
S.
Thrum
,
M.
Blüher
,
U.
Wagner
,
M.
Rossol
.
2019
.
Perturbation of the monocyte compartment in human obesity
.
Front. Immunol.
10
:
1874
.
163
Turbitt
,
W. J.
,
S. D.
Collins
,
H.
Meng
,
C. J.
Rogers
.
2019
.
Increased adiposity enhances the accumulation of MDSCs in the tumor microenvironment and adipose tissue of pancreatic tumor-bearing mice and in immune organs of tumor-free hosts
.
Nutrients
11
:
3012
.
164
Sanchez-Pino
,
M. D.
,
W. S.
Richardson
,
J.
Zabaleta
,
R. T.
Puttalingaiah
,
A. G.
Chapple
,
J.
Liu
,
Y.
Kim
,
M.
Ponder
,
R.
DeArmitt
,
L. B.
Baiamonte
, et al
.
2022
.
Increased inflammatory low-density neutrophils in severe obesity and effect of bariatric surgery: results from case-control and prospective cohort studies
.
EBioMedicine
77
:
103910
.
165
Calle
,
E. E.
,
C.
Rodriguez
,
K.
Walker-Thurmond
,
M. J.
Thun
.
2003
.
Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults
.
N. Engl. J. Med.
348
:
1625
1638
.
166
Donohoe
,
C. L.
,
J.
Lysaght
,
J.
O’Sullivan
,
J. V.
Reynolds
.
2017
.
Emerging concepts linking obesity with the hallmarks of cancer
.
Trends Endocrinol. Metab.
28
:
46
62
.
167
World Cancer Research Fund and American Institute for Cancer Research
.
2018
.
Body fatness and weight gain and the risk of cancer
. .
168
Nair
,
R. R.
,
P.
Sinha
,
A.
Khanna
,
K.
Singh
.
2015
.
Reduced myeloid-derived suppressor cells in the blood and endometrium is associated with early miscarriage
.
Am. J. Reprod. Immunol.
73
:
479
486
.
169
Rieber
,
N.
,
C.
Gille
,
N.
Köstlin
,
I.
Schäfer
,
B.
Spring
,
M.
Ost
,
H.
Spieles
,
H. A.
Kugel
,
M.
Pfeiffer
,
V.
Heininger
, et al
.
2013
.
Neutrophilic myeloid-derived suppressor cells in cord blood modulate innate and adaptive immune responses
.
Clin. Exp. Immunol.
174
:
45
52
.
170
Köstlin
,
N.
,
M.
Vogelmann
,
B.
Spring
,
J.
Schwarz
,
J.
Feucht
,
C.
Härtel
,
T. W.
Orlikowsky
,
C. F.
Poets
,
C.
Gille
.
2017
.
Granulocytic myeloid-derived suppressor cells from human cord blood modulate T-helper cell response towards an anti-inflammatory phenotype
.
Immunology
152
:
89
101
.
171
Köstlin
,
N.
,
H.
Kugel
,
B.
Spring
,
A.
Leiber
,
A.
Marmé
,
M.
Henes
,
N.
Rieber
,
D.
Hartl
,
C. F.
Poets
,
C.
Gille
.
2014
.
Granulocytic myeloid derived suppressor cells expand in human pregnancy and modulate T-cell responses
.
Eur. J. Immunol.
44
:
2582
2591
.
172
Verma
,
P.
,
R.
Verma
,
R. R.
Nair
,
S.
Budhwar
,
A.
Khanna
,
N. R.
Agrawal
,
R.
Sinha
,
R.
Birendra
,
S.
Rajender
,
K.
Singh
.
2019
.
Altered crosstalk of estradiol and progesterone with myeloid-derived suppressor cells and Th1/Th2 cytokines in early miscarriage is associated with early breakdown of maternal-fetal tolerance
.
Am. J. Reprod. Immunol.
81
:
e13081
.
173
Zhang
,
J.
,
A.
Hodges
,
S. H.
Chen
,
P. Y.
Pan
.
2021
.
Myeloid-derived suppressor cells as cellular immunotherapy in transplantation and autoimmune diseases
.
Cell. Immunol.
362
:
104300
.
174
Scalea
,
J. R.
,
Y. S.
Lee
,
E.
Davila
,
J. S.
Bromberg
.
2018
.
Myeloid-derived suppressor cells and their potential application in transplantation
.
Transplantation
102
:
359
367
.
175
Zhao
,
Y.
,
X. F.
Shen
,
K.
Cao
,
J.
Ding
,
X.
Kang
,
W. X.
Guan
,
Y. T.
Ding
,
B. R.
Liu
,
J. F.
Du
.
2018
.
Dexamethasone-induced myeloid-derived suppressor cells prolong allo cardiac graft survival through iNOS- and glucocorticoid receptor-dependent mechanism
.
Front. Immunol.
9
:
282
.
176
Lee
,
Y. S.
,
T.
Zhang
,
V.
Saxena
,
L.
Li
,
W.
Piao
,
J. S.
Bromberg
,
J. R.
Scalea
.
2020
.
Myeloid-derived suppressor cells expand after transplantation and their augmentation increases graft survival
.
Am. J. Transplant.
20
:
2343
2355
.
177
Park
,
M. Y.
,
B. G.
Lim
,
S. Y.
Kim
,
H. J.
Sohn
,
S.
Kim
,
T. G.
Kim
.
2019
.
GM-CSF promotes the expansion and differentiation of cord blood myeloid-derived suppressor cells, which attenuate xenogeneic graft-vs.-host disease
.
Front. Immunol.
10
:
183
.
178
Li
,
L.
,
J.
Yin
,
Y.
Li
,
C.
Wang
,
X.
Mao
,
J.
Wei
,
Y.
Cao
,
N.
Wang
,
L.
Lin
,
J.
Xu
,
Y.
Zhang
.
2021
.
Allogeneic hematopoietic stem cell transplantation mobilized with pegylated granulocyte colony-stimulating factor ameliorates severe acute graft-versus-host disease through enrichment of monocytic myeloid-derived suppressor cells in the graft: a real world experience
.
Front. Immunol.
12
:
621935
.
179
Qin
,
J.
,
Y.
Arakawa
,
M.
Morita
,
J. J.
Fung
,
S.
Qian
,
L.
Lu
.
2017
.
C-C chemokine receptor type 2-dependent migration of myeloid-derived suppressor cells in protection of islet transplants
.
Transplantation
101
:
1793
1800
.
180
Xu
,
J.
,
Y.
Peng
,
M.
Yang
,
N.
Guo
,
H.
Liu
,
H.
Gao
,
F.
Niu
,
R.
Wang
,
C.
Wang
,
K.
Yu
.
2020
.
Increased levels of myeloid-derived suppressor cells in esophageal cancer patients is associated with the complication of sepsis
.
Biomed. Pharmacother.
125
:
109864
.
181
Hongo
,
D.
,
X.
Tang
,
J.
Baker
,
E. G.
Engleman
,
S.
Strober
.
2014
.
Requirement for interactions of natural killer T cells and myeloid-derived suppressor cells for transplantation tolerance
.
Am. J. Transplant.
14
:
2467
2477
.
182
Ioannou
,
M.
,
T.
Alissafi
,
L.
Boon
,
D.
Boumpas
,
P.
Verginis
.
2013
.
In vivo ablation of plasmacytoid dendritic cells inhibits autoimmunity through expansion of myeloid-derived suppressor cells
.
J. Immunol.
190
:
2631
2640
.
183
Parekh
,
V. V.
,
L.
Wu
,
D.
Olivares-Villagómez
,
K. T.
Wilson
,
L.
Van Kaer
.
2013
.
Activated invariant NKT cells control central nervous system autoimmunity in a mechanism that involves myeloid-derived suppressor cells
.
J. Immunol.
190
:
1948
1960
.
184
Knier
,
B.
,
M.
Hiltensperger
,
C.
Sie
,
L.
Aly
,
G.
Lepennetier
,
T.
Engleitner
,
G.
Garg
,
A.
Muschaweckh
,
M.
Mitsdörffer
,
U.
Koedel
, et al
.
2018
.
Myeloid-derived suppressor cells control B cell accumulation in the central nervous system during autoimmunity
.
Nat. Immunol.
19
:
1341
1351
.
185
Carter
,
N. A.
,
R.
Vasconcellos
,
E. C.
Rosser
,
C.
Tulone
,
A.
Muñoz-Suano
,
M.
Kamanaka
,
M. R.
Ehrenstein
,
R. A.
Flavell
,
C.
Mauri
.
2011
.
Mice lacking endogenous IL-10-producing regulatory B cells develop exacerbated disease and present with an increased frequency of Th1/Th17 but a decrease in regulatory T cells
.
J. Immunol.
186
:
5569
5579
.
186
Park
,
M. J.
,
J. A.
Baek
,
J. W.
Choi
,
S. G.
Jang
,
D. S.
Kim
,
S. H.
Park
,
M. L.
Cho
,
S. K.
Kwok
.
2021
.
Programmed death-ligand 1 expression potentiates the immune modulatory function of myeloid-derived suppressor cells in systemic lupus erythematosus
.
Front. Immunol.
12
:
606024
.
187
Zhang
,
H.
,
S.
Wang
,
Y.
Huang
,
H.
Wang
,
J.
Zhao
,
F.
Gaskin
,
N.
Yang
,
S. M.
Fu
.
2015
.
Myeloid-derived suppressor cells are proinflammatory and regulate collagen-induced arthritis through manipulating Th17 cell differentiation
.
Clin. Immunol.
157
:
175
186
.
188
Qi
,
J.
,
D.
Li
,
G.
Shi
,
X.
Zhang
,
Y.
Pan
,
H.
Dou
,
G.
Yao
,
Y.
Hou
.
2018
.
Myeloid-derived suppressor cells exacerbate Sjögren’s syndrome by inhibiting Th2 immune responses
.
Mol. Immunol.
101
:
251
258
.
189
Moreno-Smith
,
M.
,
S. K.
Lutgendorf
,
A. K.
Sood
.
2010
.
Impact of stress on cancer metastasis
.
Future Oncol.
6
:
1863
1881
.
190
Mundy-Bosse
,
B. L.
,
L. M.
Thornton
,
H. C.
Yang
,
B. L.
Andersen
,
W. E.
Carson
.
2011
.
Psychological stress is associated with altered levels of myeloid-derived suppressor cells in breast cancer patients
.
Cell. Immunol.
270
:
80
87
.
191
An
,
J.
,
L.
Feng
,
J.
Ren
,
Y.
Li
,
G.
Li
,
C.
Liu
,
Y.
Yao
,
Y.
Yao
,
Z.
Jiang
,
Y.
Gao
, et al
.
2021
.
Chronic stress promotes breast carcinoma metastasis by accumulating myeloid-derived suppressor cells through activating β-adrenergic signaling
.
OncoImmunology
10
:
2004659
.
192
Jin
,
J.
,
X.
Wang
,
Q.
Wang
,
X.
Guo
,
J.
Cao
,
X.
Zhang
,
T.
Zhu
,
D.
Zhang
,
W.
Wang
,
J.
Wang
, et al
.
2013
.
Chronic psychological stress induces the accumulation of myeloid-derived suppressor cells in mice
.
PLoS One
8
:
e74497
.
193
Cao
,
M.
,
W.
Huang
,
Y.
Chen
,
G.
Li
,
N.
Liu
,
Y.
Wu
,
G.
Wang
,
Q.
Li
,
D.
Kong
,
T.
Xue
, et al
.
2021
.
Chronic restraint stress promotes the mobilization and recruitment of myeloid-derived suppressor cells through β-adrenergic-activated CXCL5-CXCR2-Erk signaling cascades
.
Int. J. Cancer
149
:
460
472
.
194
Spruijt
,
N. E.
,
T.
Visser
,
L. P.
Leenen
.
2010
.
A systematic review of randomized controlled trials exploring the effect of immunomodulative interventions on infection, organ failure, and mortality in trauma patients
.
Crit. Care
14
:
R150
.
195
Flores
,
R. R.
,
C. L.
Clauson
,
J.
Cho
,
B. C.
Lee
,
S. J.
McGowan
,
D. J.
Baker
,
L. J.
Niedernhofer
,
P. D.
Robbins
.
2017
.
Expansion of myeloid-derived suppressor cells with aging in the bone marrow of mice through a NF-κB-dependent mechanism
.
Aging Cell
16
:
480
487
.
196
Pang
,
W. W.
,
E. A.
Price
,
D.
Sahoo
,
I.
Beerman
,
W. J.
Maloney
,
D. J.
Rossi
,
S. L.
Schrier
,
I. L.
Weissman
.
2011
.
Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age
.
Proc. Natl. Acad. Sci. USA
108
:
20012
20017
.
197
Shipp
,
C.
,
L.
Speigl
,
N.
Janssen
,
A.
Martens
,
G.
Pawelec
.
2016
.
A clinical and biological perspective of human myeloid-derived suppressor cells in cancer
.
Cell. Mol. Life Sci.
73
:
4043
4061
.
198
Verschoor
,
C. P.
,
J.
Johnstone
,
J.
Millar
,
M. G.
Dorrington
,
M.
Habibagahi
,
A.
Lelic
,
M.
Loeb
,
J. L.
Bramson
,
D. M.
Bowdish
.
2013
.
Blood CD33+HLA-DR myeloid-derived suppressor cells are increased with age and a history of cancer
.
J. Leukoc. Biol.
93
:
633
637
.
199
Fulop
,
T.
,
A.
Larbi
,
G.
Pawelec
,
A.
Khalil
,
A. A.
Cohen
,
K.
Hirokawa
,
J. M.
Witkowski
,
C.
Franceschi
.
2021
.
Immunology of aging: the birth of inflammaging
.
Clin. Rev. Allergy Immunol.
DOI: 10.1007/s12016-021-08899-6.
200
Alves
,
A. S.
,
M. E.
Ishimura
,
Y. A. O.
Duarte
,
V.
Bueno
.
2018
.
Parameters of the immune system and vitamin D levels in old individuals
.
Front. Immunol.
9
:
1122
.
201
Pawelec
,
G.
,
E.
Picard
,
V.
Bueno
,
C. P.
Verschoor
,
S.
Ostrand-Rosenberg
.
2021
.
MDSCs, ageing and inflammageing
.
Cell. Immunol.
362
:
104297
.
202
Özbay Kurt
,
F. G.
,
A.
Lepper
,
C.
Gerhards
,
M.
Roemer
,
S.
Lasser
,
I.
Arkhypov
,
R.
Bitsch
,
P.
Bugert
,
P.
Altevogt
,
C.
Gouttefangeas
, et al
.
2022
.
Booster dose of mRNA vaccine augments waning T cell and antibody responses against SARS-CoV-2
.
Front. Immunol.
13
:
1012526
.
203
Kidzeru
,
E.
,
M. A.
Gasper
,
D.
Shao
,
P. T.
Edlefsen
,
N.
Lejarcegui
,
E.
Havyarimana
,
K.
Urdahl
,
S.
Gantt
,
H.
Horton
,
H.
Jaspan
,
A.
Gervassi
.
2021
.
Myeloid-derived suppressor cells and their association with vaccine immunogenicity in South African infants
.
J. Leukoc. Biol.
110
:
939
950
.
204
Poschke
,
I.
,
Y.
Mao
,
L.
Adamson
,
F.
Salazar-Onfray
,
G.
Masucci
,
R.
Kiessling
.
2012
.
Myeloid-derived suppressor cells impair the quality of dendritic cell vaccines
.
Cancer Immunol. Immunother.
61
:
827
838
.