Minireviews Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Immunol. Mar 27, 2014; 4(1): 26-33
Published online Mar 27, 2014. doi: 10.5411/wji.v4.i1.26
Role of myeloid-derived suppressor cells in autoimmune disease
Kristen R Crook, Peng Liu, Department of Medicine, Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
Author contributions: Crook KR and Liu P designed the review, wrote the paper, and approved for publication.
Correspondence to: Peng Liu, MD, PhD, Associate Professor, Department of Medicine, Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, 3300 Thurston Building, CB# 7280, 104 Manning Drive, Chapel Hill, NC 27599, United States. liupz@med.unc.edu
Telephone: +1-919-9660570 Fax: +1-919-9669269
Received: October 25, 2013
Revised: December 12, 2013
Accepted: January 17, 2014
Published online: March 27, 2014

Abstract

Myeloid-derived suppressor cells (MDSCs) represent an important class of immunoregulatory cells that can be activated to suppress T cell functions. These MDSCs can inhibit T cell functions through cell surface interactions and the release of soluble mediators. MDSCs accumulate in the inflamed tissues and lymphoid organs of patients with autoimmune diseases. Much of our knowledge of MDSC function has come from studies involving cancer models, however many recent studies have helped to characterize MDSC involvement in autoimmune diseases. MDSCs are a heterogeneous group of immature myeloid cells with a number of different functions for the suppression of T cell responses. However, we have yet to fully understand their contributions to the development and regulation of autoimmune diseases. A number of studies have described beneficial functions of MDSCs during autoimmune diseases, and thus there appears to be a potential role for MDSCs in the treatment of these diseases. Nevertheless, many questions remain as to the activation, differentiation, and inhibitory functions of MDSCs. This review aims to summarize our current knowledge of MDSC subsets and suppressive functions in tissue-specific autoimmune disorders. We also describe the potential of MDSC-based cell therapy for the treatment of autoimmune diseases and note some of hurdles facing the implementation of this therapy.

Key Words: Myeloid-derived suppressor cells, Autoimmune disease, Autoimmunity, T cells, Chronic inflammation, Immune regulation

Core tip: Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of cells with immunosuppressive abilities. MDSCs inhibit T cell function and regulate immune responses in cancer and autoimmune diseases. Therapeutic administration of MDSCs in the mouse models of multiple sclerosis, rheumatoid arthritis, and diabetes has shown promising results. Thus, MDSCs have potential in cell-based treatments of autoimmune disorders. However, the role of MDSCs in autoimmunity is complex and not fully understood. Further studies are needed before new therapies can be implemented.



INTRODUCTION

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of immature myeloid cells that have the ability to suppress T cell functions[1]. MDSCs are derived from the bone marrow and arise from a delay in maturation during pathologic conditions, such as cancer, chronic inflammation, infection, and traumatic stress[2]. Most studies focus on the pathogenic nature of MDSCs in cancer, where suppression of T cell-mediated immune responses prevents immune surveillance and clearance of developing tumors[3-5]. Recently, MDSCs have been reported to regulate autoimmunity and control the generation and perpetuation of autoimmune diseases[6]. In this review, we will summarize the current knowledge of MDSC subsets and suppressive functions in tissue-specific autoimmune disorders. We also describe the potential of MDSC-based cell therapy for the treatment of these autoimmune diseases, while noting some of the obstacles that may hinder the implementation of this therapy.

MDSC INVOLVEMENT IN AUTOIMMUNE DISEASE

Our knowledge of the origination and functions of MDSCs has come mainly from studies in tumor models and from cancer patients[1,5,7]. The role of MDSCs in autoimmune diseases is only starting to be elucidated. We now know that MDSCs are involved in a number of different autoimmune disorders, including multiple sclerosis (MS), type 1 diabetes, rheumatoid arthritis (RA), inflammatory bowel disease (IBD) and autoimmune hepatitis. In steady state conditions, MDSCs reside primarily in the bone marrow. Under pathological conditions, MDSC populations expand and can be detected in the spleen, lymph nodes, cancerous tumors, and bloodstream. An early study using a mouse model of autoimmune uveoretinitis showed that the accumulation of nitric oxide-producing monocytes in the choroid and retina of the eye correlated with the severity of disease[8]. A later study showed similar results and confirmed the identity of these cells to be MDSCs[9]. Studies using the mouse model of MS, experimental autoimmune encephalomyelitis (EAE), showed that MDSCs were present in the demyelinated areas of the spinal cord tissue of mice. Another EAE model showed that MDSC accumulation in the spleen correlated with disease progression[10]. Here, they showed that the start of MDSC accumulation occurred during the asymptomatic phase and increased throughout the onset phase. At the peak of the disease, MDSC accumulation reached its highest level, and then began to decrease during the recovery phase and returned to steady state levels by disease resolution. Similar results were found using collagen-induced arthritis (CIA), a mouse model of RA, where MDSC accumulation in the spleen correlated with the course of disease[11]. In humans, MDSCs were found to be enriched in the bloodstream of patients with active MS, but were only slightly elevated in the blood of patients in recovery[12].

MDSCs require certain signals for their expansion and activation. The factors responsible for driving the expansion of MDSCs include cyclooxygenase-2, prostaglandins, interleukin 6 (IL-6), macrophage colony-stimulating factor (M-CSF), and granulocyte-macrophage colony-stimulating factor (GM-CSF)[9,13-18]. Most of these factors trigger signaling pathways that stimulate the proliferation of myeloid cells in the bone marrow and inhibit their differentiation into mature cells[3]. MDSCs can be activated to suppress T cell functions via interferon gamma (IFNγ) and transforming growth factor beta (TGF-β)[13]. Blocking IFNγ production by activated T cells abolishes MDSC-mediated T cell suppression[1,11,19]. Cancer models have identified IL-6, IL-1β, prostaglandin E2, and the calcium binding proteins S100A8 and S100A9, as factors important for the accumulation of MDSCs at sites of inflammation[17,20,21]. Tumor necrosis factor (TNF) signaling drives MDSC accumulation in the periphery by promoting MDSC survival and inhibiting apoptosis[22]. Treatment with a TNF-α antagonist showed decreased MDSC accumulation in the spleen in response to chronic inflammation[23].

MDSC SUBSETS IN AUTOIMMUNITY

Early classification of MDSCs was based on cell surface expression of CD11b and Gr-1. The CD11b+Gr-1+ subgroup is now divided into two separate groups, exhibiting either a monocytic morphology or a granulocytic morphology[24]. Granulocytic MDSCs (G-MDSCs) display a CD11b+Ly6ClowLy6G+ phenotype, whereas monocytic MDSCs (M-MDSCs) are CD11b+Ly6C+Ly6G-[18,24-26]. The two groups also differ in functionality[18,25,27]. MDSCs suppress T cell functions via a number of different mechanisms involving the production of soluble mediators or through cell-cell contact[28-31]. G-MDSCs frequently inhibit T cell function through arginase-1 enzyme activity. M-MDSCs more commonly inhibit T cell functions via nitric oxide production. IFNγ-mediated activation of MDSCs results in the upregulation of arginase-1 and nitric oxide production. In the CIA model, MDSCs were found to inhibit both T cell proliferation and CD4+ T cell differentiation into Th17 cells[11]. Here, the researchers used the total CD11b+-Gr-1+ population from the spleen and found both arginase-1 and nitric oxide to be mechanisms of inhibition. The Gr-1 antibody recognizes both Ly6G and Ly6C surface antigens, therefore the population of cells used for their studies contained both G-MDSCs and M-MDSCs. In a mouse model of diabetes, CD11b+Gr-1+ cells were found to inhibit CD8+ and CD4+ T cell responses via nitric oxide- and IL-10-dependent mechanisms[32]. In the EAE model, G-MDSCs from myelin oligodendrocyte glycoprotein-immunized mice were found to express high levels of programmed cell death 1 ligand 1 (PD-L1), a costimulatory molecule that negatively regulates T cell proliferation. G-MDSCs were found to inhibit autoantigen-priming of Th1 and Th17 cells in a PD-L1-dependent manner[12]. Interestingly, one report showed that CD11b+Gr-1+ cells isolated from mice with EAE inhibited T cell proliferation in co-culture but promoted Th17 cell differentiation under Th17-polarizing conditions[33].

M-MDSCs also display immunosuppressive effects during autoimmune diseases. Recent data showed that M-MDSCs induced during the priming phase of EAE were potent suppressors of activated T cells and mediated T cell inhibition through the production of nitric oxide[18]. Nitric oxide production by MDSCs results in the nitrosylation of cysteine residues, leading to a significant decrease in mRNA stability, and thereby preventing the production of cytokines required for T cell proliferation[28]. Another study demonstrated that activation of M-MDSC suppressive function occurred at the peak of EAE disease[34]. This study determined that the suppression of T cell responses was due to M-MDSC-mediated nitric oxide production. Furthermore, transfer of activated M-MDSCs led to apoptosis of T cells in the central nervous system and decreased EAE severity. In autoimmune arthritis, clinical trials against C-C chemokine receptor 2 (CCR2), the major chemokine receptor mediating monocyte recruitment, were surprisingly unsuccessful as monocytes/macrophages were thought to be pathogenic in RA[35-37]. Interestingly, CCR2-deficient mice are now known to develop exacerbated CIA[38,39]. The underlying mechanisms contributing to the aggravated disease are not clear. However, our data showed that M-MDSCs were absent from the periphery of collagen-immunized CCR2-deficient mice, as CCR2 is required for the emigration of M-MDSCs from the bone marrow[38,40]. Further, M-MDSCs isolated from the bone marrow of CCR2-deficient mice with CIA inhibited CD4+ T cell proliferation and mitigated CIA severity, suggesting M-MDSCs are required for the regulation of autoimmune arthritis[41].

Human MDSCs are identified as CD14+CD16+ and CD14+CD16- cells. These CD14+ cells were found to be abundant in the blood and synovial fluid of RA patients[42,43]. Recently, MDSCs were shown to mediate enhancement of regulatory T cell (Treg) suppressive functions[43]. Here, Tregs were isolated from healthy subjects and their suppressive activity and cytokine expression were analyzed after co-culture with CD14+ cells. Results showed an increase in the expression of IFNγ, TNF-α, IL-17, and IL-10 by Tregs, a sustained Treg phenotype, and an enhanced capacity to suppress T cell-mediated proinflammatory cytokine production and T cell proliferation.

Taken together, these studies demonstrate that MDSCs can use various functions to suppress T cell responses and suggest that MDSC differentiation and function may be influenced by the distinct environment associated with each type of disease. Although both G-MDSCs and M-MDSCs can suppress T cell functions, further research is needed to confirm whether the two subsets have different outcomes in different diseases (Table 1).

Table 1 Myeloid-derived suppressor cells in autoimmune disease models.
Human diseaseMouse modelPhenotypeT cell suppressionSuppressive mechanismSuppressive role in vivo
Multiple sclerosisEAECD11b+Ly6Chigh (M-MDSCs)CD4+ T cellsNO-apoptosisNot determined[18]
EAECD11b+Ly6G- (M-MDSCs)CD4+, CD8+, Ag-specific CD4+ T cellsNOSNo effect by naïve MDSCs[77]
EAECD11b+Ly6Chigh (M-MDSCs)Not determinedNot determinedIncrease severity[73]
EAEArg-1+CD11b+Gr-1low (M-MDSCs)CD3+ T cellsApoptosisNot determined[10]
EAECD11b+Ly6C+ (M-MDSCs)CD4+ T cellsNOReduce severity by late phase MDSCs[34]
EAECD11bhighLy6G+Ly6C- (G-MDSCs)Th1 and Th17 cellsPD-L1Reduce severity[12]
EAECD11b+Gr-1+Promote Th17 cellsIL-1βIncrease severity[33]
EAECD11b+Gr-1+Ag-specific Th17 cellsiNOS, arginase-1 and IL-10Ablated iNKT-induced disease mitigation[78]
Rheumatoid arthritisCIACD11b+Ly6C+Ly6G- (M-MDSC)CD4+ T cellsNOReduce severity[41]
CIACD11b+Gr-1+Th17 cellsArginase and iNOSReduce severity[11]
Proteoglycan- induced arthritisCD11b+Gr-1+Ag-specific T cellsNO and ROSNot determined[79]
Systemic lupus erythematosusMRL-faslprCD11b+Gr-1low (M-MDSCs)CD4+ T cellsArginase-1Not determined[80]
Inflammatory bowel diseaseHA-transgenic miceCD11b+Gr-1+Ag-specific CD8+ T cellsNO-apoptosisReduce severity[14]
DDS-induced colitisCD11b+Gr-1+Not determinedNot determinedReduce severity[75]
IL-10-/-CD11b+Gr-1+MLN T cellsNot determinedNot determined[81]
TNBS-induced colitisCD11b+Gr-1+SplenocytesNot determinedReduce severity[74]
T1DINS-HA/ RAG-/-Gr-1+CD115+ (M-MDSCs)Induce Tregs and inhibit Teff cellsTGF-β and IL-10Reduce severity[54]
h-CD20/NODCD11b+Gr-1+CD4+ and CD8+ T cells induce TregsNO and IL-10Not determined[32]
Autoimmune hepatitisTgfb-/-CD11b+Ly6Chigh Ly6G- (M-MDSCs)CD4+ T cellsNONot determined[82]
Inflammatory eye diseaseEAUCD11b+Gr-1+ Ly6G- (M-MDSCs)CD4+ T cellsTNFR-dependent, ArginaseNot Determined[9]
EAUREP-induced CD11b+Gr-1+CD4+ T cellsNot determinedReduce severity[76]
Alopecia areataAlopecia areata-eczemaCD11b+Gr-1+CD4+ and CD8+ T cellsCD3-zeta down-regulationLocal MDSC administration reduces severity[83]
MDSC-MEDIATED SUPPRESSION OF ANTIGEN-SPECIFIC IMMUNE RESPONSES

Loss of immunological tolerance is the basis for the development of autoimmune diseases. Recognition of self-antigens leads to autoimmune-driven tissue inflammation. However, regulation of the responses to self-antigens must be highly specific in order for the host immune recognition of pathogens to remain intact. MDSCs may play a crucial role in maintaining this balance as they are capable of suppressing antigen-specific immune responses. It is believed that MDSCs internalize antigens and present them to T cells, bringing the two cells into close contact. Peroxynitrite, a derivative of nitric oxide, causes nitration of tyrosine residues on the T cell receptor (TCR), thereby preventing binding between the major histocompatibility complex (MHC) and peptide[44]. Increased levels of nitrotyrosine have been documented for patients suffering from MS, RA, autoimmune myocarditis, and diabetes[45-48]. In a cancer model, increased production of peroxynitrite and hydrogen peroxide resulted from the interaction between immature myeloid cells and antigen-specific CD8+ T cells in the presence of the specific antigen, but not in the presence of the control antigen[29]. In some cancer models, arginase-1 production is the mechanism of MDSC-mediated suppression[31,49]. The arginase-1 enzyme hydrolyzes arginine, depleting the pool of arginine available to the cell[50-52]. A deficiency in arginine prevents the formation of CD3 molecules[53]. The absence of CD3 prevents signaling through the TCR upon recognition of a specific antigen-MHC complex.

In one study of autoimmune diabetes, MDSCs induced the antigen-specific expansion of Tregs, which resulted in the suppression of T cell proliferation and prevented the onset of disease[54]. The authors described that MDSC-mediated expansion of Tregs was dependent on antigen presentation by MHC class II molecules. For these experiments, hemagglutinin (HA)-specific CD4+ T cells were adoptively transferred to mice, followed by the administration of MDSCs and HA antigen. The results showed a significant reduction in disease upon administration of MDSCs and HA, but no decrease in disease when MDSCs were administered with the ovalbumin peptide, confirming that the MDSC-mediated suppression was antigen-specific.

MDSCs also mediate suppression of non-specific T cell responses, i.e., mitogen-activated T cell responses, suggesting MDSCs may be involved in the late phase of tissue inflammation during autoimmune diseases. Others have hypothesized that MDSCs function in both antigen-specific and non-specific manners depending on the signals they are exposed to in a particular microenvironment[55]. Indeed, comparison of MDSCs isolated from the spleen to those isolated from a tumor showed that splenic MDSCs were able to inhibit antigen-specific T cell responses via the production of reactive oxygen species, whereas MDSCs isolated from the tumor inhibited T cells nonspecifically and more potently than those from the spleen[56]. T cells isolated from the peripheral lymphoid organs of human cancer patients, or from a mouse tumor model, are still responsive to non-cancer related stimuli, including viruses, IL-2, and anti-CD3/CD28 antibodies[1,57]. This suggests that the expansion of MDSCs does not induce systemic immune suppression. Taken together, these data suggest that MDSCs from the site of inflammation may be more potent and far-reaching in their suppressive effects than those MDSCs in the peripheral organs. The MDSCs in circulation may function to prevent the spread of inflammation to other areas of the body, without compromising immune recognition of pathogens.

THERAPEUTIC POTENTIAL OF MDSC-BASED TREATMENTS

Therapeutic approaches involving MDSCs require their purification and/or proliferation in vitro. MDSCs migrate to peripheral lymphoid organs where they differentiate into granulocytes, monocytes/macrophages, and dendritic cells (DCs). GM-CSF has been shown to drive MDSC accumulation at sites of inflammation[58,59] and has been used to generate MDSCs from bone marrow cells in vitro[60]. However, the concentration of GM-CSF in the media must be tightly regulated as different concentrations of GM-CSF may lead to the generation of neutrophils or DCs[60,61]. Vascular endothelial growth factor (VEGF) is important in the differentiation of hematopoietic progenitor cells[62], and studies have shown that blocking VEGF binding leads to increased differentiation of MDSCs into DCs[63]. Similar results were shown for stem cell factor, where blocking its function led to reduced MDSC expansion[64]. Factors such as granulocyte colony-stimulating factor (G-CSF) and M-CSF are also known to induce MDSC expansion. G-CSF induces the proliferation of G-MDSCs via the Janus kinase/signal transducers and activators of transcription pathway (Jak/STAT)[65]. In the presence of IL-6, M-CSF was shown to inhibit DC generation from hematopoietic stem cells (HSCs), thereby redirecting HSC differentiation towards MDSCs[66]. The calcium binding proteins, S100A8 and S100A9, are upregulated in some autoimmune conditions, including RA, MS, and IBD[66-68]. These proteins are secreted by MDSCs[69] and may work in an autocrine fashion to promote the accumulation of MDSCs while simultaneously preventing their differentiation into DCs[70]. MDSC generation, expansion, and gain of specific suppressive abilities occur primarily under inflammatory conditions such as infection, cancer, trauma, and autoimmune diseases. It is important to note that MDSCs are not terminally differentiated, and thus may mature into antigen-presenting cells, such as macrophages or DCs, highlighting a potential complication for therapeutic attempts. Therefore, in order to develop effective MDSC-based therapies, we must first understand how different cell types respond to different inflammatory mediators and determine how these inflammatory mediators affect the potency and/or suppressive mechanisms of MDSCs.

A number of studies have provided insight into the use of MDSCs for treatment of autoimmune diseases. In a murine model of diabetes, MDSCs were generated in vitro by culturing hepatic stellate cells with DCs[71]. This method of MDSC generation was previously shown to produce highly suppressive cells in an IFNγ-dependent manner[72]. In the diabetes study, these in vitro-generated MDSCs were mixed with pancreatic islet cells and transplanted into diabetic mice. The MDSCs induced Treg expansion in the allograft site, resulting in the inhibition of CD8+ T cell responses[71]. In a mouse model of IBD, MDSCs were found to be upregulated in the spleen and intestine of IBD mice[14]. Further data showed that these MDSCs effectively prevented T cell proliferation and induced T cell apoptosis after transfer of CD8+ T cells[14]. One report showed that the in vivo transfer of G-MDSCs in the EAE model resulted in the delayed onset of disease and a significant reduction in demyelination[12], however other studies were not as successful[33,73]. Adoptive transfer of MDSCs also led to reduced disease severity in models of RA[11,41], IBD[74,75], and inflammatory eye disease[76].

CONCLUSION

MDSCs represent an important class of immunoregulatory cells. MDSCs display particular heterogeneity and plasticity, and for these reasons they have become an attractive candidate for the treatment of autoimmune diseases. On the other hand, MDSCs are very difficult to work with because of their diverse nature. MDSCs have multiple phenotypes which inhibit T cell responses by multiple mechanisms, and their environment dictates the development of suppressive properties and activation pathways. Additionally, the maturation/differentiation of these cells may depend on the particular inflammatory signals received from their microenvironment. Though MDSCs hold promise in the treatment of autoimmune diseases, their full utilization is stalled by our limited understanding of their phenotype, differentiation, cellular functions, and influence on the microenvironment.

Footnotes

P- Reviewers: Sotelo J, Yves R S- Editor: Song XX L- Editor: A E- Editor: Liu SQ

References
1.  Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162-174.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Nagaraj S, Collazo M, Corzo CA, Youn JI, Ortiz M, Quiceno D, Gabrilovich DI. Regulatory myeloid suppressor cells in health and disease. Cancer Res. 2009;69:7503-7506.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32:19-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 567]  [Cited by in F6Publishing: 634]  [Article Influence: 45.3]  [Reference Citation Analysis (0)]
4.  Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12:253-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2369]  [Cited by in F6Publishing: 2651]  [Article Influence: 220.9]  [Reference Citation Analysis (0)]
5.  Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor cells in cancer: recent progress and prospects. Immunol Cell Biol. 2013;91:493-502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 168]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
6.  Cripps JG, Gorham JD. MDSC in autoimmunity. Int Immunopharmacol. 2011;11:789-793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 109]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
7.  Greten TF, Manns MP, Korangy F. Myeloid derived suppressor cells in human diseases. Int Immunopharmacol. 2011;11:802-807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 322]  [Cited by in F6Publishing: 319]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
8.  Liversidge J, Dick A, Gordon S. Nitric oxide mediates apoptosis through formation of peroxynitrite and Fas/Fas-ligand interactions in experimental autoimmune uveitis. Am J Pathol. 2002;160:905-916.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 68]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
9.  Kerr EC, Raveney BJ, Copland DA, Dick AD, Nicholson LB. Analysis of retinal cellular infiltrate in experimental autoimmune uveoretinitis reveals multiple regulatory cell populations. J Autoimmun. 2008;31:354-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 124]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
10.  Moliné-Velázquez V, Cuervo H, Vila-Del Sol V, Ortega MC, Clemente D, de Castro F. Myeloid-derived suppressor cells limit the inflammation by promoting T lymphocyte apoptosis in the spinal cord of a murine model of multiple sclerosis. Brain Pathol. 2011;21:678-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 83]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
11.  Fujii W, Ashihara E, Hirai H, Nagahara H, Kajitani N, Fujioka K, Murakami K, Seno T, Yamamoto A, Ishino H. Myeloid-derived suppressor cells play crucial roles in the regulation of mouse collagen-induced arthritis. J Immunol. 2013;191:1073-1081.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 122]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
12.  Ioannou M, Alissafi T, Lazaridis I, Deraos G, Matsoukas J, Gravanis A, Mastorodemos V, Plaitakis A, Sharpe A, Boumpas D. Crucial role of granulocytic myeloid-derived suppressor cells in the regulation of central nervous system autoimmune disease. J Immunol. 2012;188:1136-1146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 172]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
13.  Fujio K, Okamura T, Sumitomo S, Yamamoto K. Regulatory T cell-mediated control of autoantibody-induced inflammation. Front Immunol. 2012;3:28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
14.  Haile LA, von Wasielewski R, Gamrekelashvili J, Krüger C, Bachmann O, Westendorf AM, Buer J, Liblau R, Manns MP, Korangy F. Myeloid-derived suppressor cells in inflammatory bowel disease: a new immunoregulatory pathway. Gastroenterology. 2008;135:871-881, 881.e1-5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 233]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
15.  Makarenkova VP, Bansal V, Matta BM, Perez LA, Ochoa JB. CD11b+/Gr-1+ myeloid suppressor cells cause T cell dysfunction after traumatic stress. J Immunol. 2006;176:2085-2094.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Serafini P, Carbley R, Noonan KA, Tan G, Bronte V, Borrello I. High-dose granulocyte-macrophage colony-stimulating factor-producing vaccines impair the immune response through the recruitment of myeloid suppressor cells. Cancer Res. 2004;64:6337-6343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 389]  [Cited by in F6Publishing: 379]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
17.  Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007;67:4507-4513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 544]  [Cited by in F6Publishing: 559]  [Article Influence: 32.9]  [Reference Citation Analysis (0)]
18.  Zhu B, Bando Y, Xiao S, Yang K, Anderson AC, Kuchroo VK, Khoury SJ. CD11b+Ly-6C(hi) suppressive monocytes in experimental autoimmune encephalomyelitis. J Immunol. 2007;179:5228-5237.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Corzo CA, Cotter MJ, Cheng P, Cheng F, Kusmartsev S, Sotomayor E, Padhya T, McCaffrey TV, McCaffrey JC, Gabrilovich DI. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol. 2009;182:5693-5701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 519]  [Cited by in F6Publishing: 564]  [Article Influence: 37.6]  [Reference Citation Analysis (0)]
20.  Bunt SK, Sinha P, Clements VK, Leips J, Ostrand-Rosenberg S. Inflammation induces myeloid-derived suppressor cells that facilitate tumor progression. J Immunol. 2006;176:284-290.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Sinha P, Okoro C, Foell D, Freeze HH, Ostrand-Rosenberg S, Srikrishna G. Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells. J Immunol. 2008;181:4666-4675.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Zhao X, Rong L, Zhao X, Li X, Liu X, Deng J, Wu H, Xu X, Erben U, Wu P. TNF signaling drives myeloid-derived suppressor cell accumulation. J Clin Invest. 2012;122:4094-4104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 245]  [Cited by in F6Publishing: 266]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
23.  Sade-Feldman M, Kanterman J, Ish-Shalom E, Elnekave M, Horwitz E, Baniyash M. Tumor necrosis factor-α blocks differentiation and enhances suppressive activity of immature myeloid cells during chronic inflammation. Immunity. 2013;38:541-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 201]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
24.  Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol. 2008;181:5791-5802.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Dietlin TA, Hofman FM, Lund BT, Gilmore W, Stohlman SA, van der Veen RC. Mycobacteria-induced Gr-1+ subsets from distinct myeloid lineages have opposite effects on T cell expansion. J Leukoc Biol. 2007;81:1205-1212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 84]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
26.  Hettinger J, Richards DM, Hansson J, Barra MM, Joschko AC, Krijgsveld J, Feuerer M. Origin of monocytes and macrophages in a committed progenitor. Nat Immunol. 2013;14:821-830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 429]  [Cited by in F6Publishing: 455]  [Article Influence: 41.4]  [Reference Citation Analysis (0)]
27.  Movahedi K, Guilliams M, Van den Bossche J, Van den Bergh R, Gysemans C, Beschin A, De Baetselier P, Van Ginderachter JA. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood. 2008;111:4233-4244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 913]  [Cited by in F6Publishing: 941]  [Article Influence: 58.8]  [Reference Citation Analysis (0)]
28.  Bronte V, Zanovello P. Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol. 2005;5:641-654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1219]  [Cited by in F6Publishing: 1276]  [Article Influence: 67.2]  [Reference Citation Analysis (0)]
29.  Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol. 2004;172:989-999.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L, Herber DL, Schneck J, Gabrilovich DI. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med. 2007;13:828-835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 779]  [Cited by in F6Publishing: 828]  [Article Influence: 48.7]  [Reference Citation Analysis (0)]
31.  Rodríguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev. 2008;222:180-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 469]  [Cited by in F6Publishing: 512]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
32.  Hu C, Du W, Zhang X, Wong FS, Wen L. The role of Gr1+ cells after anti-CD20 treatment in type 1 diabetes in nonobese diabetic mice. J Immunol. 2012;188:294-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 28]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
33.  Yi H, Guo C, Yu X, Zuo D, Wang XY. Mouse CD11b+Gr-1+ myeloid cells can promote Th17 cell differentiation and experimental autoimmune encephalomyelitis. J Immunol. 2012;189:4295-4304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 125]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
34.  Zhu B, Kennedy JK, Wang Y, Sandoval-Garcia C, Cao L, Xiao S, Wu C, Elyaman W, Khoury SJ. Plasticity of Ly-6C(hi) myeloid cells in T cell regulation. J Immunol. 2011;187:2418-2432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
35.  Horuk R. Chemokine receptor antagonists: overcoming developmental hurdles. Nat Rev Drug Discov. 2009;8:23-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 235]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
36.  Vergunst CE, Gerlag DM, Lopatinskaya L, Klareskog L, Smith MD, van den Bosch F, Dinant HJ, Lee Y, Wyant T, Jacobson EW. Modulation of CCR2 in rheumatoid arthritis: a double-blind, randomized, placebo-controlled clinical trial. Arthritis Rheum. 2008;58:1931-1939.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Kalinowska A, Losy J. Investigational C-C chemokine receptor 2 antagonists for the treatment of autoimmune diseases. Expert Opin Investig Drugs. 2008;17:1267-1279.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Rampersad RR, Tarrant TK, Vallanat CT, Quintero-Matthews T, Weeks MF, Esserman DA, Clark J, Di Padova F, Patel DD, Fong AM. Enhanced Th17-cell responses render CCR2-deficient mice more susceptible for autoimmune arthritis. PLoS One. 2011;6:e25833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
39.  Quinones MP, Estrada CA, Kalkonde Y, Ahuja SK, Kuziel WA, Mack M, Ahuja SS. The complex role of the chemokine receptor CCR2 in collagen-induced arthritis: implications for therapeutic targeting of CCR2 in rheumatoid arthritis. J Mol Med (Berl). 2005;83:672-681.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Serbina NV, Pamer EG. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat Immunol. 2006;7:311-317.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Crook K, Weeks M, Rampersad R, Baldi R, Liu P. Myeloid-derived suppressor cells ameliorate the pathogenesis of autoimmune arthritis (P1058). J Immunol. 2013;190:185.3.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Evans HG, Gullick NJ, Kelly S, Pitzalis C, Lord GM, Kirkham BW, Taams LS. In vivo activated monocytes from the site of inflammation in humans specifically promote Th17 responses. Proc Natl Acad Sci USA. 2009;106:6232-6237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 157]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
43.  Walter GJ, Evans HG, Menon B, Gullick NJ, Kirkham BW, Cope AP, Geissmann F, Taams LS. Interaction with activated monocytes enhances cytokine expression and suppressive activity of human CD4+CD45ro+CD25+CD127(low) regulatory T cells. Arthritis Rheum. 2013;65:627-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 65]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
44.  Birnboim HC, Lemay AM, Lam DK, Goldstein R, Webb JR. Cutting edge: MHC class II-restricted peptides containing the inflammation-associated marker 3-nitrotyrosine evade central tolerance and elicit a robust cell-mediated immune response. J Immunol. 2003;171:528-532.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Bachmaier K, Neu N, Pummerer C, Duncan GS, Mak TW, Matsuyama T, Penninger JM. iNOS expression and nitrotyrosine formation in the myocardium in response to inflammation is controlled by the interferon regulatory transcription factor 1. Circulation. 1997;96:585-591.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Bagasra O, Michaels FH, Zheng YM, Bobroski LE, Spitsin SV, Fu ZF, Tawadros R, Koprowski H. Activation of the inducible form of nitric oxide synthase in the brains of patients with multiple sclerosis. Proc Natl Acad Sci USA. 1995;92:12041-12045.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Ceriello A, Mercuri F, Quagliaro L, Assaloni R, Motz E, Tonutti L, Taboga C. Detection of nitrotyrosine in the diabetic plasma: evidence of oxidative stress. Diabetologia. 2001;44:834-838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 305]  [Cited by in F6Publishing: 317]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
48.  Kaur H, Halliwell B. Evidence for nitric oxide-mediated oxidative damage in chronic inflammation. Nitrotyrosine in serum and synovial fluid from rheumatoid patients. FEBS Lett. 1994;350:9-12.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Bronte V, Serafini P, De Santo C, Marigo I, Tosello V, Mazzoni A, Segal DM, Staib C, Lowel M, Sutter G. IL-4-induced arginase 1 suppresses alloreactive T cells in tumor-bearing mice. J Immunol. 2003;170:270-278.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Cederbaum SD, Yu H, Grody WW, Kern RM, Yoo P, Iyer RK. Arginases I and II: do their functions overlap? Mol Genet Metab. 2004;81 Suppl 1:S38-S44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 178]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
51.  Hesse M, Modolell M, La Flamme AC, Schito M, Fuentes JM, Cheever AW, Pearce EJ, Wynn TA. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of L-arginine metabolism. J Immunol. 2001;167:6533-6544.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Jenkinson CP, Grody WW, Cederbaum SD. Comparative properties of arginases. Comp Biochem Physiol B Biochem Mol Biol. 1996;114:107-132.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Rodriguez PC, Zea AH, Culotta KS, Zabaleta J, Ochoa JB, Ochoa AC. Regulation of T cell receptor CD3zeta chain expression by L-arginine. J Biol Chem. 2002;277:21123-21129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 362]  [Cited by in F6Publishing: 350]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
54.  Yin B, Ma G, Yen CY, Zhou Z, Wang GX, Divino CM, Casares S, Chen SH, Yang WC, Pan PY. Myeloid-derived suppressor cells prevent type 1 diabetes in murine models. J Immunol. 2010;185:5828-5834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 136]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
55.  Nagaraj S, Gabrilovich DI. Tumor escape mechanism governed by myeloid-derived suppressor cells. Cancer Res. 2008;68:2561-2563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 236]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
56.  Corzo CA, Condamine T, Lu L, Cotter MJ, Youn JI, Cheng P, Cho HI, Celis E, Quiceno DG, Padhya T. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207:2439-2453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 757]  [Cited by in F6Publishing: 871]  [Article Influence: 62.2]  [Reference Citation Analysis (0)]
57.  Solito S, Bronte V, Mandruzzato S. Antigen specificity of immune suppression by myeloid-derived suppressor cells. J Leukoc Biol. 2011;90:31-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 73]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
58.  Fu YX, Watson G, Jimenez JJ, Wang Y, Lopez DM. Expansion of immunoregulatory macrophages by granulocyte-macrophage colony-stimulating factor derived from a murine mammary tumor. Cancer Res. 1990;50:227-234.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Ilkovitch D, Lopez DM. Urokinase-mediated recruitment of myeloid-derived suppressor cells and their suppressive mechanisms are blocked by MUC1/sec. Blood. 2009;113:4729-4739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
60.  Rössner S, Voigtländer C, Wiethe C, Hänig J, Seifarth C, Lutz MB. Myeloid dendritic cell precursors generated from bone marrow suppress T cell responses via cell contact and nitric oxide production in vitro. Eur J Immunol. 2005;35:3533-3544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 113]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
61.  Lutz MB, Suri RM, Niimi M, Ogilvie AL, Kukutsch NA, Rössner S, Schuler G, Austyn JM. Immature dendritic cells generated with low doses of GM-CSF in the absence of IL-4 are maturation resistant and prolong allograft survival in vivo. Eur J Immunol. 2000;30:1813-1822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
62.  Gabrilovich D, Ishida T, Oyama T, Ran S, Kravtsov V, Nadaf S, Carbone DP. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood. 1998;92:4150-4166.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Roland CL, Lynn KD, Toombs JE, Dineen SP, Udugamasooriya DG, Brekken RA. Cytokine levels correlate with immune cell infiltration after anti-VEGF therapy in preclinical mouse models of breast cancer. PLoS One. 2009;4:e7669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 155]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
64.  Pan PY, Wang GX, Yin B, Ozao J, Ku T, Divino CM, Chen SH. Reversion of immune tolerance in advanced malignancy: modulation of myeloid-derived suppressor cell development by blockade of stem-cell factor function. Blood. 2008;111:219-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 259]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
65.  Nefedova Y, Nagaraj S, Rosenbauer A, Muro-Cacho C, Sebti SM, Gabrilovich DI. Regulation of dendritic cell differentiation and antitumor immune response in cancer by pharmacologic-selective inhibition of the janus-activated kinase 2/signal transducers and activators of transcription 3 pathway. Cancer Res. 2005;65:9525-9535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 233]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
66.  Leach ST, Yang Z, Messina I, Song C, Geczy CL, Cunningham AM, Day AS. Serum and mucosal S100 proteins, calprotectin (S100A8/S100A9) and S100A12, are elevated at diagnosis in children with inflammatory bowel disease. Scand J Gastroenterol. 2007;42:1321-1331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 138]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
67.  Nukui T, Ehama R, Sakaguchi M, Sonegawa H, Katagiri C, Hibino T, Huh NH. S100A8/A9, a key mediator for positive feedback growth stimulation of normal human keratinocytes. J Cell Biochem. 2008;104:453-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 91]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
68.  Roth J, Goebeler M, Sorg C. S100A8 and S100A9 in inflammatory diseases. Lancet. 2001;357:1041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
69.  Zhao F, Hoechst B, Duffy A, Gamrekelashvili J, Fioravanti S, Manns MP, Greten TF, Korangy F. S100A9 a new marker for monocytic human myeloid-derived suppressor cells. Immunology. 2012;136:176-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 153]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
70.  Cheng P, Corzo CA, Luetteke N, Yu B, Nagaraj S, Bui MM, Ortiz M, Nacken W, Sorg C, Vogl T. Inhibition of dendritic cell differentiation and accumulation of myeloid-derived suppressor cells in cancer is regulated by S100A9 protein. J Exp Med. 2008;205:2235-2249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 640]  [Cited by in F6Publishing: 682]  [Article Influence: 42.6]  [Reference Citation Analysis (0)]
71.  Chou HS, Hsieh CC, Charles R, Wang L, Wagner T, Fung JJ, Qian S, Lu LL. Myeloid-derived suppressor cells protect islet transplants by B7-H1 mediated enhancement of T regulatory cells. Transplantation. 2012;93:272-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
72.  Chou HS, Hsieh CC, Yang HR, Wang L, Arakawa Y, Brown K, Wu Q, Lin F, Peters M, Fung JJ. Hepatic stellate cells regulate immune response by way of induction of myeloid suppressor cells in mice. Hepatology. 2011;53:1007-1019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 98]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
73.  King IL, Dickendesher TL, Segal BM. Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease. Blood. 2009;113:3190-3197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 329]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
74.  Guan Q, Moreno S, Qing G, Weiss CR, Lu L, Bernstein CN, Warrington RJ, Ma Y, Peng Z. The role and potential therapeutic application of myeloid-derived suppressor cells in TNBS-induced colitis. J Leukoc Biol. 2013;94:803-811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
75.  Zhang R, Ito S, Nishio N, Cheng Z, Suzuki H, Isobe KI. Dextran sulphate sodium increases splenic Gr1(+)CD11b(+) cells which accelerate recovery from colitis following intravenous transplantation. Clin Exp Immunol. 2011;164:417-427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
76.  Tu Z, Li Y, Smith D, Doller C, Sugita S, Chan CC, Qian S, Fung J, Caspi RR, Lu L. Myeloid suppressor cells induced by retinal pigment epithelial cells inhibit autoreactive T-cell responses that lead to experimental autoimmune uveitis. Invest Ophthalmol Vis Sci. 2012;53:959-966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
77.  Slaney CY, Toker A, La Flamme A, Bäckström BT, Harper JL. Naïve blood monocytes suppress T-cell function. A possible mechanism for protection from autoimmunity. Immunol Cell Biol. 2011;89:7-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
78.  Parekh VV, Wu L, Olivares-Villagómez D, Wilson KT, Van Kaer L. Activated invariant NKT cells control central nervous system autoimmunity in a mechanism that involves myeloid-derived suppressor cells. J Immunol. 2013;190:1948-1960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
79.  Egelston C, Kurkó J, Besenyei T, Tryniszewska B, Rauch TA, Glant TT, Mikecz K. Suppression of dendritic cell maturation and T cell proliferation by synovial fluid myeloid cells from mice with autoimmune arthritis. Arthritis Rheum. 2012;64:3179-3188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
80.  Iwata Y, Furuichi K, Kitagawa K, Hara A, Okumura T, Kokubo S, Shimizu K, Sakai N, Sagara A, Kurokawa Y. Involvement of CD11b+ GR-1 low cells in autoimmune disorder in MRL-Fas lpr mouse. Clin Exp Nephrol. 2010;14:411-417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
81.  Singh UP, Singh NP, Singh B, Hofseth LJ, Taub DD, Price RL, Nagarkatti M, Nagarkatti PS. Role of resveratrol-induced CD11b(+) Gr-1(+) myeloid derived suppressor cells (MDSCs) in the reduction of CXCR3(+) T cells and amelioration of chronic colitis in IL-10(-/-) mice. Brain Behav Immun. 2012;26:72-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 70]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
82.  Cripps JG, Wang J, Maria A, Blumenthal I, Gorham JD. Type 1 T helper cells induce the accumulation of myeloid-derived suppressor cells in the inflamed Tgfb1 knockout mouse liver. Hepatology. 2010;52:1350-1359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 73]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
83.  Marhaba R, Vitacolonna M, Hildebrand D, Baniyash M, Freyschmidt-Paul P, Zöller M. The importance of myeloid-derived suppressor cells in the regulation of autoimmune effector cells by a chronic contact eczema. J Immunol. 2007;179:5071-5081.  [PubMed]  [DOI]  [Cited in This Article: ]