Skip to main content
Erschienen in: Journal of Clinical Immunology 5/2022

Open Access 29.04.2022 | Original Article

Intravenous Immunoglobulins Promote an Expansion of Monocytic Myeloid-Derived Suppressor Cells (MDSC) in CVID Patients

verfasst von: Miriam Simón-Fuentes, Silvia Sánchez-Ramón, Lidia Fernández-Paredes, Bárbara Alonso, Kissy Guevara-Hoyer, Miguel A. Vega, Angel L. Corbí, Ángeles Domínguez-Soto

Erschienen in: Journal of Clinical Immunology | Ausgabe 5/2022

Abstract

Common variable immunodeficiency disorders (CVID), the most common primary immune deficiency, includes heterogeneous syndromes characterized by hypogammaglobulinemia and impaired antibody responses. CVID patients frequently suffer from recurrent infections and inflammatory conditions. Currently, immunoglobulin replacement therapy (IgRT) is the first-line treatment to prevent infections and aminorate immune alterations in CVID patients. Intravenous Immunoglobulin (IVIg), a preparation of highly purified poly-specific IgG, is used for treatment of immunodeficiencies as well as for autoimmune and inflammatory disorders, as IVIg exerts immunoregulatory and anti-inflammatory actions on innate and adaptive immune cells. To determine the mechanism of action of IVIg in CVID in vivo, we determined the effect of IVIg infusion on the transcriptome of peripheral blood mononuclear cells from CVID patients, and found that peripheral blood monocytes are primary targets of IVIg in vivo, and that IVIg triggers the acquisition of an anti-inflammatory gene profile in human monocytes. Moreover, IVIg altered the relative proportions of peripheral blood monocyte subsets and enhanced the proportion of CD14+ cells with a transcriptional, phenotypic, and functional profile that resembles that of monocytic myeloid-derived suppressor cells (MDSC). Therefore, our results indicate that CD14 + MDSC-like cells might contribute to the immunoregulatory effects of IVIg in CVID and other inflammatory disorders.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10875-022-01277-7.
Miriam Simón-Fuentes and Silvia Sánchez-Ramón contributed equally to this work.
Miguel A. Vega, Angel L. Corbí and Ángeles Domínguez-Soto contributed equally to this work.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Common variable immunodeficiency disorders (CVID) are one of the most common clinically important primary immune deficiencies. CVID encompasses a group of heterogeneous primary antibody failure syndromes characterized by hypogammaglobulinemia associated with reduced or absent specific antibody production [1, 2]. CVID patients commonly suffer from recurrent infections of the gastrointestinal and upper respiratory tracts [3, 4], and inflammatory conditions and autoimmunity are also frequent in CVID patients [46]. Immune dysregulation in CVID might be secondary to defects of B-cell differentiation and function, chronic and reduced T cell function, and altered NK cells and dendritic cells function, with chronic microbial translocation possibly contributing to the systemic immune activation and altered homeostasis of lymphocytic and myeloid lineages [79]. Currently, immunoglobulin replacement therapy (IgRT), either intravenous (IVIg) or subcutaneous (SCIg), is a first-line therapy to prevent infections and aminorate all these immune alteration in CVID patients [10, 11].
IVIg is a preparation of highly purified polyclonal poly-specific IgG isolated from plasma of thousands of healthy donors and widely used for the treatment of primary and secondary immunodeficiencies, as well as autoimmune and inflammatory disorders (reviewed in [12]). Besides their antibody replacement effect, IVIg exerts immunoregulatory and anti-inflammatory actions on innate and adaptive immune cells [1317], and various non-mutually exclusive mechanisms have been proposed [1721] to explain its clinical effectiveness [1824]. The high number of donors used to prepare IVIg is particularly relevant to its anti-inflammatory effects, given the extensive pool of unique IgG antibody repertoire and anti-carbohydrate repertoire [25]. The ample anti-inflammatory activity of IVIg has recently attracted attention to its potential therapeutic use for COVID-19 [2629], COVID-19-related Kawasaki disease [3032], and for the thrombotic thrombocytopenia observed after ChAdOx1 nCov-19 vaccination [3335]. We have previously demonstrated that IVIg skews macrophage polarization through FcγR-dependent mechanisms [36] and that IVIg promotes tolerance towards inflammatory stimuli [37]. However, extrapolation of the prophylactically administration in animal models of disease falls short in providing definitive answers about its mode of action in humans in vivo, where IVIg is commonly used in numerous other therapeutic strategies [21].
To address the mechanisms of action of IVIg in vivo, we have now determined the phenotypic, transcriptomic, and functional profile of peripheral blood mononuclear cells (PBMC) from CVID patients prior and after IVIg infusion. Our results indicate that IVIg triggers the acquisition of an anti-inflammatory profile in PBMCs and monocytes, reduces the number of inflammatory circulating monocytes, and enhances the proportion of CD14+ monocytes whose phenotype and suppressor activity is compatible with that of myeloid-derived suppressor cells (MDSC). Our results indicate that monocytes are primary targets for the anti-inflammatory and inmmunosupressive effects of IVIg in CVID patients in vivo.

Materials and Methods

Patients and Clinical Samples

We studied a cohort of 11 CVID patients (age range of 20–70 years; mean age: 48.3 ± 13.8 years) followed at the Department of Clinical Immunology at the Hospital Clínico San Carlos (Madrid, Spain). CVID patients were diagnosed according to the classification of European Society of Immune Deficiencies (ESID) and the Pan-American Group for Immune Deficiency (PAGID) [38, 39]. Given the inherent heterogeneity in CVID manifestations, we took into account the classification of clinical phenotypes proposed by Chapel et al. [40], and our cohort included patients with “no-disease-related” complications (“infections only” phenotype) (n = 5) and patients with inflammatory/autoimmune/lymphoproliferative complications (“inflammatory” phenotype) (n = 6) (Supplementary Table 1). The study protocol was approved by the Ethics Committee of Hospital Clínico San Carlos (Madrid, Spain), and all subjects provided signed informed consent (Project 19/284-E).
All CVID patients were in a stable state with no apparent acute infection, and received IgRT as part of their routine treatment. The mean cumulative monthly dosage of IgRT was 400 mg/kg, with an infusion time from 4 to 6 h established according to the individual patient´s tolerability. The mean IVIg dose administered at the time of blood sampling was 28.7 ± 3.9 g. None of the 11 patients were taking steroids or other immunosuppressive or immunomodulatory drugs at the time of the study or the previous 6 months. Blood was obtained from CVID patients both before and after (6 h) receiving IgRT infusion, and PBMC were isolated over a Lymphoprep (Nycomed Pharma, Oslo, Norway) gradient according to standard procedures. Monocytes, T lymphocytes, and B lymphocytes were purified from PBMC by magnetic cell sorting using CD14, CD3, and CD20 immunomagnetic beads (Miltenyi Biotech, Bergisch Gladbach, Germany), respectively.

Microarray Analysis

Global gene expression analysis was performed on RNA obtained from PBMC, monocytes, T lymphocytes, and B lymphocytes isolated immediately before or after IVIg therapy of four independent patients. RNA isolation, microarray analysis (whole human genome microarray, Agilent Technologies, Palo Alto, CA), and statistical treatment of microarray data were performed following previously described procedures [4143]. Microarray data were deposited in the Gene Expression Omnibus (http://​www.​ncbi.​nlm.​nih.​gov/​geo/​) under accession nos. GSE133835 (PBMCs), GSE133907 (CD14 cells), GSE158576 (CD20 cells), and GSE158573 (CD3 cells). For Gene Set Enrichment Analysis (GSEA) (http://​software.​broadinstitute.​org/​gsea/​index.​jsp) [44], the gene sets available at the website, as well as previously defined gene sets, were used.

Phenotypic Analysis of Monocyte

Whole blood samples were collected before and after IVIg administration. Immediately after collection, blood sample was incubated at room temperature for 20 min with the indicated fluorescently tagged monoclonal antibodies. Following RBC lysis (RT, 15 min) using FACS Lysing solution (Becton Dickinson), cells were washed twice and analyzed on a FACS Canto II (Becton Dickinson) flow cytometer. For simultaneous surface and intracellular staining, cell surface antigen staining was performed first, and cells were later resuspended in Buffer Perm/Wash 1 × solution, treated with fixation and permeabilization solution (4 °C, 30 min in the dark) and subjected to intracellular staining. Monocyte subpopulations were phenotypically identified by a 8-color flow cytometry single platform assay using anti-CD14-APC Cy7, CD16-FITC, CX3CR1-PerCP Cy5, HLA-DR-BV510, CD86-PE, CCR5-BV421, CCR2-APC, and TNF-PE mAbs (BD, Becton- Dickinson Biosciences, Franklin Lakes, NJ).

T Cell Suppression Assay and Cytokine Secretion

Human peripheral blood CD4+ lymphocytes were isolated from CVID patients using magnetic cell sorting with anti-CD4 microbeads (Miltenyi Biotec), resuspended in RMPI 5% human AB serum (Sigma-Aldrich), and added into flatbottom 96-well plates (105 cells/well) that had been coated overnight with anti-human CD3 (10 μg/ml, BD Biosciences) and anti-human CD28 (1 μg/ml, BD Biosciences). Then, CD14+ cells isolated from CVID patients (both before and after IVIg infusion) were resuspended in RMPI 5% human AB serum, and co-cultured with CD4+ lymphocytes at the indicated ratios. After 48 h, [3H]thymidine was added (1 uCi/well, Perkin Elmer) during the last 20 h of coculture and thymidine incorporation was determined using a MicroBeta2 2450 Microplate Counter. Cell culture supernatants from the suppression assay were collected after 48 h and IFN-γ levels determined by ELISA (PBL Assay Science) following the protocol supplied by the manufacturers.

Statistical Analysis

Unless otherwise indicated and for comparisons of means, statistical analysis was performed using the Student t test, and a p value < 0.05 was considered significant (*p < 0.05; **p < 0.01; ***p < 0.001).

Results

In Vivo IVIg Infusion Prompts the Acquisition of an Anti-inflammatory and Immunosuppressive Transcriptional Profile in Peripheral Blood Mononuclear Cells from CVID Patients

To identify the cell types that mediate the anti-inflammatory and immunosuppressive effects of IVIg in vivo, we initially compared the transcriptional profiles of PBMCs, CD14+ monocytes, CD3+ T cells and CD20+ B cells isolated from CVID patients before and 6 h after IVIg infusion (Fig. 1A). IVIg triggered substantial changes in the gene signature of all analyzed cell subsets (Fig. 1B) and, in fact, modulated the expression of a common set of genes in CD14+, CD3+, and CD20+ cells (Fig. 1C). Gene ontology analysis using GSEA further supported the similarity among the transcriptional effects of IVIg on PBMC and the three cell types, with a shared negative enrichment of genes associated to the term “TNF signaling via NFkB” (Fig. 1D). Moreover, GSEA showed a marked resemblance of the gene ontology terms negatively enriched in IVIg-treated CD14+ monocytes and IVIg-treated PBMCs, including the terms “Coagulation” and “Inflammatory Response” (Fig. 1D). Indeed, the genes significantly upregulated or downregulated by IVIg in CD14+ monocytes were found to be positively or negatively enriched in the transcriptome of PBMC post-IVIg (Fig. 1E). As a whole, these analysis revealed the overlapping transcriptional effects of IVIg on PBMCs and CD14+ monocytes in vivo, and that the IVIg-induced transcriptional changes in PBMCs and CD14+ monocytes are compatible with the anti-inflammatory and immunosuppressive effects of IVIg [1721].
Based on the above findings, we next focused our subsequent analysis on the transcriptome of CD14+ monocytes after IVIg treatment. As shown in Fig. 1F, IVIg infusion provoked a very significant negative enrichment of the terms “TNF signaling via NFKB,” ´”inflammatory response,” "PD-1 signaling," and "CD28 co-stimulation" in IVIg-treated CD14+ cells (Fig. 1F). Furthermore, Enrichr and GSEA gene ontology analysis revealed that the transcriptional changes induced by IVIg in CD14+ monocytes greatly resemble the transcriptional changes that take place in monocytes in breast cancer patients (GSE65517) [45], and in PBMCs from heart failure (GSE9128) [46] and hepatocellular carcinoma [47] patients (GSE49515) (Fig. 1G). Specifically, the genes specifically upregulated (or downregulated) in IVIg-treated CD14 + monocytes were similarly upregulated (or downregulated) in monocytes or PMBCs from the indicated conditions (Fig. 1G), where immunosuppression predominates and profound phenotypic and functional changes take place in peripheral leukocytes [4549]. In addition, a similar result was obtained when the expression of genes modulated by IVIg in PBMCs was analyzed (Supplementary Fig. 1A). Altogether, these results illustrate the ability of IVIg to impair the acquisition of an inflammatory and immuno-stimulatory gene signature in human monocytes in vivo, and that IVIg infusion prompts the acquisition of a transcriptome that resembles the gene profiles of peripheral blood myeloid cells found in disorders associated with strong immunosuppression.

Phenotypic Consequences of IVIg Infusion on Peripheral Blood Monocytes from CVID Patients

To functionally assess the effects of IVIg on CVID monocytes, and since the expression of the CD16-encoding FCGR3 gene is significantly downregulated by IVIg (Fig. 2A), we initially checked the consequences of IVIg infusion on the phenotype of peripheral blood monocytes. Three major human peripheral blood monocyte subsets exist, whose defining phenotypes are CD14++CD16 (classical monocytes), CD14++CD16+ (intermediate monocytes), and CD14+/−CD16+ (non-classical monocytes) [50]. Flow cytometry showed a drastic and significant reduction of CD16+ (intermediate and non-classical) monocytes (Fig. 2B, left panel), as well as a reduction of 22% in the CD16 cell surface expression (Fig. 2B, right panel), in peripheral blood from IVIg-treated CVID patients. Besides, the percentage of monocytes expressing CX3CR1, another specific marker for CD16+ monoytes [51], was also significantly reduced in the peripheral blood of IVIg-treated CVID patients (Supplementary Fig. 1B), suggesting that the decrease in CD16+ monocytes in IVIg-treated CVID patients reflects changes in monocyte subsets proportions and is not merely due to IVIg-mediated occupancy or internalization of CD16. As a whole, flow cytometry revealed that the percentage of intermediate and non-classical monocyte subsets decrease upon IVIg treatment in CVID patients, while the percentage of the classical monocyte subset is elevated after IVIg infusion in vivo (Fig. 2C). Furthermore, the effects of IVIg on the relative proportion of monocyte subsets in CVID patients were substantiated through the analysis of the genesets that define the three major monocyte subsets [52, 53]. Indeed, the most specific marker genes for CD16+ monocytes (GSE16836) [52] were found to be preferentially expressed by pre-IVIg CVID monocytes, whereas post-IVIg monocytes displayed enhanced expression of genes that characterize CD16 monocytes (Fig. 2D). Likewise, classical monocyte-specific genes [53] were expressed at higher levels in post-IVIg CVID monocytes, while intermediate- and non-classical-specific genes [53] were preferentially expressed by pre-IVIg CVID monocytes (Fig. 2E), and similar results were seen upon evaluation of the expression of additional monocyte subset-specific gene sets in pre-IVIg and post-IVIg CVID monocytes [53] (Fig. 2F). Therefore, the combination of cytofluorimetric and transcriptomic experiments provided solid evidence for a reduction in the intermediate (CD14++CD16+) and non-classical (CD14+CD16++) peripheral blood monocyte subsets in IVIg-treated CVID patients, a result that fits with the transcriptional data and is in agreement with previous reports [54]. Noteworthy, similar results were obtained in all CVID patients regardless of their clinical CVID phenotype.
Finally, since CD16+ monocytes are increased in several inflammatory and autoimmune diseases [55] and possess a more mature phenotype [52], we explored whether IVIg also affected the in vivo expression of TNF in the distinct monocyte subsets. Cytometry analysis after intracellular staining revealed that IVIg infusion significantly reduced TNF expression in both CD16+ and CD16 monocyte subsets, albeit the inhibitory action was more profound in the intermediate CD14+CD16+ monocyte population (Supplementary Fig. 1C and Fig. 2G). Thus, all monocyte populations from CVID patients appear to exhibit a reduced inflammatory profile after IVIg treatment, what might contribute to the immunomodulatory actions of IVIg in vivo.

IVIg Infusion in CVID Patients Increases the Level of Immunosuppressive M-MDSC in Peripheral Blood

The above results indicated that IVIg limits the pro-inflammatory capacity of monocytes in CVID patients, but did not fully justify the net anti-inflammatory and immunosuppressive action of IVIg [21]. An initial hint of the anti-inflammatory/immunosuppressor effect of IVIg was suggested by the resemblance of the transcriptome of post-IVIg monocytes to the gene signature of monocytes and PBMCs from diseases where elevated levels of myeloid-derived suppressor cells (M-DSC) are found (cancer, cardiomyophathy) [4549] (Fig. 1G). MDSC are a heterogeneous population of immature myeloid cells that expands in chronic and acute inflammation and in various cancer types [56, 57], and are functionally defined by their potent ability to suppress T cell activation [5759] through various molecular mechanisms [56, 58, 60]. MDSC include two phenotypically distinct subsets: monocytic MDSC (M-MDSC), characterized by a Lin − CD11b + CD14 + CD15 − HLA-DR − /low phenotype, and polymorphonuclear MDSC (PMN-MDSC), with a Lin − CD11b + CD14 − CD15 + HLA-DR − or Lin − CD11b + CD14 − CD66b + phenotypic profile [61, 62].
To assess whether IVIg affects the level of MDSC, we initially compared HLA-DR expression, a hallmark of M-MDSCs [61], in CD14 + cells from CVID patients before and after IVIg infusion. HLA-DR expression was found to be significantly lower in post-IVIg CD14 + cells from CVID patients (Fig. 3A), a pattern that was observed in all monocyte subsets (Fig. 3B). Next, and following the consensus accepted for the identification of M-MDSCs [61], we determined the percentage of CD14 + HLA-DRlow cells before and after IVIg treatment and using the gating strategy shown in Supplementary Fig. 1D. Analysis of nine CVID patients revealed that IVIg treatment significantly increases the percentage of HLA-DRlow CD14 + cells (Fig. 3C), an increase that coincided with reduced cell surface expression of HLA-DR (Fig. 3D) and of the M-MDSCs markers CD16, CD86, CX3CR1 and CCR5 (Fig. 3E). This increase in the percentage of HLA-DRlow CD14 + cells was transitory, since a week after the infusion with IVIg only a few patients maintained certain levels of HLA-DRlow CD14 + cells increased compared to before IVIg treatment (data not shown). Therefore, IVIg infusion in CVID patients results in elevated proportion of CD14 + cells whose phenotype is compatible with that of M-MDSC. Further support for such an increase was obtained through GSEA on the transcriptomes of pre- and post-IVIg CD14+ cells. Specifically, the transcriptome of post-IVIg CD14+ cells was positively enriched in genes upregulated during in vitro MDSC induction (GSE73333) [63, 64], whereas pre-IVIg CD14+ showed a higher expression of genes whose expression is reduced in along monocyte-to-MDSC differentiation [63, 64] (Fig. 3F). Moreover, genes directly associated with MDSC proliferation and immunosuppressive function (GSE65517) [45, 65] were preferentially expressed by post-IVIg CD14+ cells (Fig. 3G). Therefore, these results indicate that IVIg treatment of CVID patients results in enhanced levels of CD14+ cells phenotypically and transcriptionally similar to M-MDSCs. Again, results were similar in all CVID patients regardless of their clinical CVID phenotype.
Finally, to assess the inmunosuppressive ability of IVIg-induced M-MDSC-like CD14+ cells in CVID patients, they were co-cultured with autologous CD4+ T cells in the presence of T cell-activating anti-CD3 and anti-CD28 antibodies (Fig. 4A). As shown in Fig. 4B, activated CD4+ T cells cultured in the presence of post-IVIg CD14+ cells exhibited a weaker proliferation level than in the presence of pre-IVIg CD14+ cells, a feature consistent with the inmunosuppressive phenotype of M-MDSCs. As expected, a stronger inmunosuppressive effect was observed at lower CD4+ T cell/CD14+ ratios (Fig. 4C). More importantly, the presence of post-IVIg CD14+ cells also resulted in diminished production of IFNγ production (Fig. 4D), a feature that was also dependent on the CD4+ T cell/CD14+ ratio (Fig. 4E). Taken together, the transcriptional, phenotypic, and functional data indicate that IVIg infusion leads to higher levels of immunosuppressive M-MDSCs in CVID patients.

Discussion

CVID is the most frequently diagnosed primary immunodeficiency. Baseline inflammatory complications, autoimmune diseases, and lymphoproliferation are common in CVID patients [4, 6668], defining specific clinical phenotypes due to dysfunctional immune responses besides those seen upon recurrent infections [40]. Although the mechanisms underlying CVID-associated immune dysregulation remain largely unclear [68], previous reports have shown increased microbial translocation and systemic myeloid cell activation in CVID patients [6972], whose chronic monocyte activation appears related to persistence of T cell activation and the inflammatory and lymphoproliferative complications [68, 73]. IVIg therapy is currently the treatment of choice for CVID, and we have previously shown that IVIg modifies the phenotype and function of myeloid cells in vitro and in vivo [36, 37]. Although it is accepted that the main therapeutic benefit of IVIg in CVID patients is the presence of pathogen-specific antibodies [74], IVIg-mediated cellular re-programming might also contribute to improve the control of infections in CVID patients [10, 11, 75]. We now report that IVIg treatment of CVID patients provokes the acquisition of an anti-inflammatory profile in PBMC and monocytes, and that IVIg enhances the percentage of CD14+ monocytes with a transcriptional, phenotypic, and functional profile compatible with those of myeloid-derived suppressor cells (MDSC). In parallel, IVIg infusion led to a marked reduction of the intermediate and non-classic monocyte subsets at the transcriptomic and phenotypic levels, a finding that is in agreement with the phenotypic effects of IVIg described by Cavaliere et al. in CVID patients [54]. Our results indicate that monocytes are preferential IVIg targets in vivo, and that the IVIg-mediated changes in the relative levels of monocyte subsets might contribute to the anti-inflammatory and immunosuppressive effects of IVIg in CVID. Of note, no difference was observed between CVID patients with either “infections only” or “inflammatory” phenotype. However, given the size of the analyzed cohort, these results warrant further validation in an independent and larger cohort.
Compared to healthy individuals, CVID patients have been recently found to exhibit higher levels of low-density neutrophils, whose phenotype and suppressive activity is consistent with granulocytic MDSC [70] that might contribute to the immune dysregulation in CVID. To our knowledge, the present report provides the first evidence for an IVIg-mediated increase in blood M-MDSC in CVID patients in vivo. We have also analyzed the levels of PMN-MDSC (CD15 + CD11b + CD33 + HLA-DRlow) in IVIg-treated CVID patients, but the results were inconclusive, and a larger cohort of CVID patients is required to clarify this issue in future studies. Our findings are in line with a previous report describing an increase in CD33+/CD11b+/HLA-DR MDSC in immune thrombocytopenia (ITP) patients treated with both IVIg and dexamethasone after 6 days [76], and also agree with an enhancement of CD33+/CD11b+/HLA-DR cells in spleen cells from ITP patients exposed in vitro to IVIg for 90 h [77]. Our results, however, indicate that IVIg enhances monocytic MDSC levels in peripheral blood as soon as 6 h after infusion, indicating an acute effect. MDSC appear to originate mainly from an emergency myelopoiesis [78], and their enrichment might be due to monocyte reprogramming into an immunosuppressive state, early release of bone marrow immature myeloid cells into the circulation (emergency myelopoiesis), or a combination of both mechanisms [79]. Whether the IVIg-induced increase in monocytic MDSC in CVID reflects a re-programming of peripheral blood monocytic cells or is secondary to release of bone marrow progenitors has yet to be addressed. Although the latter cannot be ruled out, since IVIg-treated CVID patients do not show elevated monocyte counts, and considering that IVIg re-program monocytes and macrophages in vitro [36, 37], it is reasonable to assume that IVIg directly shapes peripheral blood monocytes at the transcriptional and phenotypic level in vivo in CVID patients. In any event, and regardless of its origin, the immunosuppressive character of IVIg-induced monocytic MDSC might help in protecting the host from the extensive tissue damage caused by the excessive monocyte activation usually observed in CVID [73]. Moreover, and since MDSC also appear to increase immune surveillance and innate immune responses [79], IVIg-induced MDSC might also contribute to maintain immune homeostasis and improve antimicrobial activities in CVID patients. Of note, we have not seen any differences in IVIg-induced MDSC increases between patients with distinct duration of IVIg treatment. However, considering the limited cohort we have analyzed, future studies should assess prospectively whether the duration of IVIg treatment has any effect on the IVIg-induced changes in MDSC population that we now report.
Regarding the IVIg-mediated decrease in the intermediate and non-classical monocyte subsets in CVID, seen at the phenotypic and transcriptional levels, our findings support the idea that IVIg can correct the imbalance of monocytes subsets seen in CVID patients, which exhibit increased levels of CD16+ monocytes [54, 73]. Indeed, our results corroborate previous findings on the ability of IVIg to diminish the number of non-classical monocytes in after 4 h in CVID patients [54, 8082], an effect that appears to be transient [80] and has been also observed in patients with Kawasaki disease [83]. Therefore, considering that CD16+ monocytes exhibit more pro-inflammatory ability than classical monocytes [50] and give rise to macrophages with a more pro-inflammatory gene profile [84], the expression of genes preferentially expressed in intermediate/non-classical monocytes (Fig. 2D) could be used as molecular markers for immediate/early responses to IVIg infusion.
An additional consequence of the IVIg-mediated M-MDSC increase in CVID patients is its potential involvement in the generation of regulatory T lymphocytes (Treg). Numerous reports have now established that IVIg enhances suppressive Treg [8588], an effect observed in immune thrombocytopenia [89], Guillain-Barré syndrome [90], Myasthenia Gravis [91], allergic airways disease [92], Kawasaki disease [93], and experimental autoimmune encephalomyelitis [94]. In fact, Treg expansion has been proposed as a biomarker to predict clinical response to IVIg therapy [95], and is thought to be one of the mechanisms by which IVIg restores homeostasis in patients with autoimmune and systemic inflammatory disorders. Since MDSC promote Treg expansion [96100] and recruitment [101], the IVIg-induced increase in peripheral blood MDSC that we have observed in CVID has additional implications, and might be a primary step in the immunosuppressive ability of IVIg. While the global IVIg-induced anti-inflammatory effects may be beneficial to restrain chronic immune activation and inflammation in the complex interplay of factors involved in oncogenesis in CVID population [102], a major concern of our data is the potential deleterious effects of IVIg on patients with established cancer. This latter aspect deserves further focused exploration.
In summary, we report that IVIg infusion has an immediate effect on the transcriptome, phenotype and function of peripheral blood monocytes in CVID patients, and that these IVIg-induced changes are compatible with IVIg promoting the acquisition of M-MDSC-like properties upon infusion. These results warrant further analysis of potential similar IVIg effects in other diseases, especially considering the ample immunomodulatory actions of MDSC and the large number of disorders that are currently treated with IVIg.

Declarations

Ethics Approval

This study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the Ethics Committee of Hospital Clínico San Carlos (Madrid, Spain), and all subjects provided signed informed consent (Project 19/284-E).
Informed consent was obtained from all individual participants included in the study.
The authors affirm that human research participants provided informed consent for publication of individual person’s data included in this study.

Conflict of Interest

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

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

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

e.Med Innere Medizin

Kombi-Abonnement

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

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Chapel H, Cunningham-Rundles C. Update in understanding common variable immunodeficiency disorders (CVIDs) and the management of patients with these conditions. Br J Haematol. 2009;145:709–27.PubMedPubMedCentralCrossRef Chapel H, Cunningham-Rundles C. Update in understanding common variable immunodeficiency disorders (CVIDs) and the management of patients with these conditions. Br J Haematol. 2009;145:709–27.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Notarangelo LD, Fischer A, Geha RS, Casanova JL, Chapel H, Conley ME, et al. Primary immunodeficiencies: 2009 update. J Allergy Clin Immunol. 2009;124(6):1161–78.PubMedPubMedCentralCrossRef Notarangelo LD, Fischer A, Geha RS, Casanova JL, Chapel H, Conley ME, et al. Primary immunodeficiencies: 2009 update. J Allergy Clin Immunol. 2009;124(6):1161–78.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Bogaert DJA, Dullaers M, Lambrecht BN, Vermaelen KY, De Baere E, Haerynck F. Genes associated with common variable immunodeficiency: one diagnosis to rule them all? J Med Genet. 2016;53(9):575–90.PubMedCrossRef Bogaert DJA, Dullaers M, Lambrecht BN, Vermaelen KY, De Baere E, Haerynck F. Genes associated with common variable immunodeficiency: one diagnosis to rule them all? J Med Genet. 2016;53(9):575–90.PubMedCrossRef
4.
Zurück zum Zitat Patuzzo G, Barbieri A, Tinazzi E, Veneri D, Argentino G, Moretta F, et al. Autoimmunity and infection in common variable immunodeficiency (CVID). Autoimmun Rev. 2016;15:877–82.PubMedCrossRef Patuzzo G, Barbieri A, Tinazzi E, Veneri D, Argentino G, Moretta F, et al. Autoimmunity and infection in common variable immunodeficiency (CVID). Autoimmun Rev. 2016;15:877–82.PubMedCrossRef
5.
Zurück zum Zitat Resnick ES, Moshier EL, Godbold JH, Cunningham-Rundles C. Morbidity and mortality in common variable immune deficiency over 4 decades. Blood. 2012;119(7):1650–7.PubMedPubMedCentralCrossRef Resnick ES, Moshier EL, Godbold JH, Cunningham-Rundles C. Morbidity and mortality in common variable immune deficiency over 4 decades. Blood. 2012;119(7):1650–7.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Chapel H, Lucas M, Lee M, Bjorkander J, Webster D, Grimbacher B, et al. Common Variable immunodeficiency disorders: division into distinct clinical phenotypes. Blood. 2008;112(2):277–86.PubMedCrossRef Chapel H, Lucas M, Lee M, Bjorkander J, Webster D, Grimbacher B, et al. Common Variable immunodeficiency disorders: division into distinct clinical phenotypes. Blood. 2008;112(2):277–86.PubMedCrossRef
7.
Zurück zum Zitat Aspalter RM, Sewell WAC, Dolman K, Farrant J, Webster ADB. Deficiency in circulating natural killer (NK) cell subsets in common variable immunodeficiency and X-linked agammaglobulinaemia. Clin Exp Immunol. 2000;121(3):506–14.PubMedPubMedCentralCrossRef Aspalter RM, Sewell WAC, Dolman K, Farrant J, Webster ADB. Deficiency in circulating natural killer (NK) cell subsets in common variable immunodeficiency and X-linked agammaglobulinaemia. Clin Exp Immunol. 2000;121(3):506–14.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Litzman J, Vlková M, Pikulová Z, Štikarovská D, Lokaj J. T and B lymphocyte subpopulations and activation/differentiation markers in patients with selective IgA deficiency. Clin Exp Immunol. 2007;147(2):249–54.PubMedPubMedCentralCrossRef Litzman J, Vlková M, Pikulová Z, Štikarovská D, Lokaj J. T and B lymphocyte subpopulations and activation/differentiation markers in patients with selective IgA deficiency. Clin Exp Immunol. 2007;147(2):249–54.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Scott-Taylor TH, Green MR, Raeiszadeh M, Workman S, Webster AD. Defective maturation of dendritic cells in common variable immunodeficiency. Clin Exp Immunol. 2006;145(3):420–7.PubMedPubMedCentralCrossRef Scott-Taylor TH, Green MR, Raeiszadeh M, Workman S, Webster AD. Defective maturation of dendritic cells in common variable immunodeficiency. Clin Exp Immunol. 2006;145(3):420–7.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Kaveri SV, Maddur MS, Hegde P, Lacroix-Desmazes S, Bayry J. Intravenous immunoglobulins in immunodeficiencies: more than mere replacement therapy. Clin Exp Immunol. 2011;164:2–5.PubMedPubMedCentralCrossRef Kaveri SV, Maddur MS, Hegde P, Lacroix-Desmazes S, Bayry J. Intravenous immunoglobulins in immunodeficiencies: more than mere replacement therapy. Clin Exp Immunol. 2011;164:2–5.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Perez EE, Orange JS, Bonilla F, Chinen J, Chinn IK, Dorsey M, et al. Update on the use of immunoglobulin in human disease: a review of evidence. J Allergy Clin Immunol. 2017;139(3):S1-46.PubMedCrossRef Perez EE, Orange JS, Bonilla F, Chinen J, Chinn IK, Dorsey M, et al. Update on the use of immunoglobulin in human disease: a review of evidence. J Allergy Clin Immunol. 2017;139(3):S1-46.PubMedCrossRef
13.
Zurück zum Zitat Kazatchkine MD, Kaveri SV. Immunomodulation of autoimmune and inflammatory diseases with intravenous immune globulin. N Engl J Med. 2001;345(10):747–55.PubMedCrossRef Kazatchkine MD, Kaveri SV. Immunomodulation of autoimmune and inflammatory diseases with intravenous immune globulin. N Engl J Med. 2001;345(10):747–55.PubMedCrossRef
14.
15.
Zurück zum Zitat Gelfand EW. Intravenous immune globulin in autoimmune and inflammatory diseases. N Engl J Med. 2012;367:2015–25.PubMedCrossRef Gelfand EW. Intravenous immune globulin in autoimmune and inflammatory diseases. N Engl J Med. 2012;367:2015–25.PubMedCrossRef
16.
Zurück zum Zitat Schwab I, Nimmerjahn F. Intravenous immunoglobulin therapy: how does IgG modulate the immune system? Nat Rev Immunol. 2013;13:176–89.PubMedCrossRef Schwab I, Nimmerjahn F. Intravenous immunoglobulin therapy: how does IgG modulate the immune system? Nat Rev Immunol. 2013;13:176–89.PubMedCrossRef
17.
Zurück zum Zitat Durandy A, Kaveri SV, Kuijpers TW, Basta M, Miescher S, Ravetch JV, et al. Intravenous immunoglobulins-understanding properties and mechanisms. Clin Exp Immunol. 2009;158:2–13.PubMedPubMedCentralCrossRef Durandy A, Kaveri SV, Kuijpers TW, Basta M, Miescher S, Ravetch JV, et al. Intravenous immunoglobulins-understanding properties and mechanisms. Clin Exp Immunol. 2009;158:2–13.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Tha-In T, Bayry J, Metselaar HJ, Kaveri SV, Kwekkeboom J. Modulation of the cellular immune system by intravenous immunoglobulin. Trends Immunol. 2008;29:608–15.PubMedCrossRef Tha-In T, Bayry J, Metselaar HJ, Kaveri SV, Kwekkeboom J. Modulation of the cellular immune system by intravenous immunoglobulin. Trends Immunol. 2008;29:608–15.PubMedCrossRef
19.
Zurück zum Zitat Ballow M. The IgG molecule as a biological immune response modifier: mechanisms of action of intravenous immune serum globulin in autoimmune and inflammatory disorders. J Allergy Clin Immunol. 2011;127:315–23.PubMedCrossRef Ballow M. The IgG molecule as a biological immune response modifier: mechanisms of action of intravenous immune serum globulin in autoimmune and inflammatory disorders. J Allergy Clin Immunol. 2011;127:315–23.PubMedCrossRef
20.
Zurück zum Zitat Negi VS, Elluru S, Sibéril S, Graff-Dubois S, Mouthon L, Kazatchkine MD, et al. Intravenous immunoglobulin: an update on the clinical use and mechanisms of action. J Clin Immunol. 2007;27:233–45.PubMedCrossRef Negi VS, Elluru S, Sibéril S, Graff-Dubois S, Mouthon L, Kazatchkine MD, et al. Intravenous immunoglobulin: an update on the clinical use and mechanisms of action. J Clin Immunol. 2007;27:233–45.PubMedCrossRef
21.
Zurück zum Zitat Tjon AS, van Gent R, Geijtenbeek TB, Kwekkeboom J. Differences in anti-inflammatory actions of intravenous immunoglobulin between mice and men: more than meets the eye. Front Immunol. 2015;6:197.PubMedPubMedCentralCrossRef Tjon AS, van Gent R, Geijtenbeek TB, Kwekkeboom J. Differences in anti-inflammatory actions of intravenous immunoglobulin between mice and men: more than meets the eye. Front Immunol. 2015;6:197.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Corbi AL, Sanchez-Ramon S, Dominguez-Soto A. The potential of intravenous immunoglobulins for cancer therapy: a road that is worth taking? Immunotherapy. 2016;8(5):601–12.PubMedCrossRef Corbi AL, Sanchez-Ramon S, Dominguez-Soto A. The potential of intravenous immunoglobulins for cancer therapy: a road that is worth taking? Immunotherapy. 2016;8(5):601–12.PubMedCrossRef
23.
Zurück zum Zitat Ben Mkaddem S, Aloulou M, Benhamou M, Monteiro RC. Role of FcγRIIIA (CD16) in IVIg-mediated anti-inflammatory function. J Clin Immunol. 2014;34(SUPPL. 1):S46-50.PubMedCrossRef Ben Mkaddem S, Aloulou M, Benhamou M, Monteiro RC. Role of FcγRIIIA (CD16) in IVIg-mediated anti-inflammatory function. J Clin Immunol. 2014;34(SUPPL. 1):S46-50.PubMedCrossRef
24.
Zurück zum Zitat Aloulou M, Ben Mkaddem S, Biarnes-Pelicot M, Boussetta T, Souchet H, Rossato E, et al. IgG1 and IVIg induce inhibitory ITAM signaling through FcγRIII controlling inflammatory responses. Blood. 2012;119(13):3084–96.PubMedCrossRef Aloulou M, Ben Mkaddem S, Biarnes-Pelicot M, Boussetta T, Souchet H, Rossato E, et al. IgG1 and IVIg induce inhibitory ITAM signaling through FcγRIII controlling inflammatory responses. Blood. 2012;119(13):3084–96.PubMedCrossRef
26.
Zurück zum Zitat Alijotas-Reig J, Esteve-Valverde E, Belizna C, Selva-O’Callaghan A, Pardos-Gea J, Quintana A, et al. Immunomodulatory therapy for the management of severe COVID-19. Beyond the anti-viral therapy: a comprehensive review. Autoimmun Rev. 2020;19:102569.PubMedPubMedCentralCrossRef Alijotas-Reig J, Esteve-Valverde E, Belizna C, Selva-O’Callaghan A, Pardos-Gea J, Quintana A, et al. Immunomodulatory therapy for the management of severe COVID-19. Beyond the anti-viral therapy: a comprehensive review. Autoimmun Rev. 2020;19:102569.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Cao W, Liu X, Bai T, Fan H, Hong K, Song H, et al. High-dose intravenous immunoglobulin as a therapeutic option for deteriorating patients with coronavirus disease 2019. Open Forum Infect Dis. 2020;7(3):1–6.CrossRef Cao W, Liu X, Bai T, Fan H, Hong K, Song H, et al. High-dose intravenous immunoglobulin as a therapeutic option for deteriorating patients with coronavirus disease 2019. Open Forum Infect Dis. 2020;7(3):1–6.CrossRef
29.
Zurück zum Zitat Prete M, Favoino E, Catacchio G, Racanelli V, Perosa F. SARS-CoV-2 infection complicated by inflammatory syndrome. Could high-dose human immunoglobulin for intravenous use (IVIG) be beneficial? Autoimmun Rev. 2020;19(7):102559.PubMedPubMedCentralCrossRef Prete M, Favoino E, Catacchio G, Racanelli V, Perosa F. SARS-CoV-2 infection complicated by inflammatory syndrome. Could high-dose human immunoglobulin for intravenous use (IVIG) be beneficial? Autoimmun Rev. 2020;19(7):102559.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Verdoni L, Mazza A, Gervasoni A, Martelli L, Ruggeri M, Ciuffreda M, et al. An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study. Lancet. 2020;395(10239):1771–8.PubMedPubMedCentralCrossRef Verdoni L, Mazza A, Gervasoni A, Martelli L, Ruggeri M, Ciuffreda M, et al. An outbreak of severe Kawasaki-like disease at the Italian epicentre of the SARS-CoV-2 epidemic: an observational cohort study. Lancet. 2020;395(10239):1771–8.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Riphagen S, Gomez X, Gonzalez-Martinez C, Wilkinson N, Theocharis P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet. 2020;395:1607–8.PubMedPubMedCentralCrossRef Riphagen S, Gomez X, Gonzalez-Martinez C, Wilkinson N, Theocharis P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet. 2020;395:1607–8.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Goto R, Inuzuka R, Shindo T, Namai Y, Oda Y, Harita Y, et al. Relationship between post-IVIG IgG levels and clinical outcomes in Kawasaki disease patients: new insight into the mechanism of action of IVIG. Clin Rheumatol. 2020;39(12):3747–55.PubMedCrossRef Goto R, Inuzuka R, Shindo T, Namai Y, Oda Y, Harita Y, et al. Relationship between post-IVIG IgG levels and clinical outcomes in Kawasaki disease patients: new insight into the mechanism of action of IVIG. Clin Rheumatol. 2020;39(12):3747–55.PubMedCrossRef
35.
Zurück zum Zitat von Hundelshausen P, Lorenz R, Siess W, Weber C. Vaccine-induced immune thrombotic thrombocytopenia (VITT): targeting pathomechanisms with Bruton tyrosine kinase inhibitors. Thromb Haemost. 2021;121(11):1395–9. https://doi.org/10.1055/a-1481-3039. von Hundelshausen P, Lorenz R, Siess W, Weber C. Vaccine-induced immune thrombotic thrombocytopenia (VITT): targeting pathomechanisms with Bruton tyrosine kinase inhibitors. Thromb Haemost. 2021;121(11):1395–9. https://​doi.​org/​10.​1055/​a-1481-3039.
36.
Zurück zum Zitat Dominguez-Soto A, de las Casas-Engel M, Bragado R, Medina-Echeverz J, Aragoneses-Fenoll L, Martin-Gayo E, et al. Intravenous immunoglobulin promotes antitumor responses by modulating macrophage polarization. J Immunol. 2014;193(10):5181–9.PubMedCrossRef Dominguez-Soto A, de las Casas-Engel M, Bragado R, Medina-Echeverz J, Aragoneses-Fenoll L, Martin-Gayo E, et al. Intravenous immunoglobulin promotes antitumor responses by modulating macrophage polarization. J Immunol. 2014;193(10):5181–9.PubMedCrossRef
37.
Zurück zum Zitat Domínguez-Soto Á, Simón-Fuentes M, de las Casas-Engel M, Cuevas VD, López-Bravo M, Domínguez-Andrés J, et al. IVIg promote cross-tolerance against inflammatory stimuli in vitro and in vivo. J Immunol. 2018;201(1):41–52.PubMedCrossRef Domínguez-Soto Á, Simón-Fuentes M, de las Casas-Engel M, Cuevas VD, López-Bravo M, Domínguez-Andrés J, et al. IVIg promote cross-tolerance against inflammatory stimuli in vitro and in vivo. J Immunol. 2018;201(1):41–52.PubMedCrossRef
38.
Zurück zum Zitat Seidel MG, Kindle G, Gathmann B, Quinti I, Buckland M, van Montfrans J, et al. The European Society for Immunodeficiencies (ESID) registry working definitions for the clinical diagnosis of inborn errors of immunity. J Allergy Clin Immunol Pract. 2019;7(6):1763–70.PubMedCrossRef Seidel MG, Kindle G, Gathmann B, Quinti I, Buckland M, van Montfrans J, et al. The European Society for Immunodeficiencies (ESID) registry working definitions for the clinical diagnosis of inborn errors of immunity. J Allergy Clin Immunol Pract. 2019;7(6):1763–70.PubMedCrossRef
39.
Zurück zum Zitat Conley ME, Notarangelo LD, Etzioni A. Diagnostic criteria for primary immunodeficiencies. Clin Immunol. 1999;93(3):190–7.PubMedCrossRef Conley ME, Notarangelo LD, Etzioni A. Diagnostic criteria for primary immunodeficiencies. Clin Immunol. 1999;93(3):190–7.PubMedCrossRef
40.
Zurück zum Zitat Chapel H, Lucas M, Patel S, Lee M, Cunningham-Rundles C, Resnick E, et al. Confirmation and improvement of criteria for clinical phenotyping in common variable immunodeficiency disorders in replicate cohorts. J Allergy Clin Immunol. 2012;130(5):1197–8. https://doi.org/10.1016/j.jaci.2012.05.046. Chapel H, Lucas M, Patel S, Lee M, Cunningham-Rundles C, Resnick E, et al. Confirmation and improvement of criteria for clinical phenotyping in common variable immunodeficiency disorders in replicate cohorts. J Allergy Clin Immunol. 2012;130(5):1197–8. https://​doi.​org/​10.​1016/​j.​jaci.​2012.​05.​046.
41.
Zurück zum Zitat Cuevas VD, Anta L, Samaniego R, Orta-Zavalza E, Vladimir de la Rosa J, Baujat G, et al. MAFB determines human macrophage anti-inflammatory polarization: relevance for the pathogenic mechanisms operating in multicentric carpotarsal osteolysis. J Immunol. 2017;198(5):2070–81.PubMedCrossRef Cuevas VD, Anta L, Samaniego R, Orta-Zavalza E, Vladimir de la Rosa J, Baujat G, et al. MAFB determines human macrophage anti-inflammatory polarization: relevance for the pathogenic mechanisms operating in multicentric carpotarsal osteolysis. J Immunol. 2017;198(5):2070–81.PubMedCrossRef
42.
Zurück zum Zitat Riera-Borrull M, Cuevas VD, Alonso B, Vega MA, Joven J, Izquierdo E, et al. Palmitate conditions macrophages for enhanced responses toward inflammatory stimuli via JNK activation. J Immunol. 2017;199(11):3858–69.PubMedCrossRef Riera-Borrull M, Cuevas VD, Alonso B, Vega MA, Joven J, Izquierdo E, et al. Palmitate conditions macrophages for enhanced responses toward inflammatory stimuli via JNK activation. J Immunol. 2017;199(11):3858–69.PubMedCrossRef
43.
Zurück zum Zitat Nieto C, Rayo I, de las Casas-Engel M, Izquierdo E, Alonso B, Béchade C, et al. Serotonin (5-HT) shapes the macrophage gene profile through the 5-HT 2B –dependent activation of the aryl hydrocarbon receptor. J Immunol. 2020;204(10):2808–17.PubMedCrossRef Nieto C, Rayo I, de las Casas-Engel M, Izquierdo E, Alonso B, Béchade C, et al. Serotonin (5-HT) shapes the macrophage gene profile through the 5-HT 2B –dependent activation of the aryl hydrocarbon receptor. J Immunol. 2020;204(10):2808–17.PubMedCrossRef
44.
Zurück zum Zitat Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–50.PubMedPubMedCentralCrossRef Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–50.PubMedPubMedCentralCrossRef
45.
46.
Zurück zum Zitat Cappuzzello C, Napolitano M, Arcelli D, Melillo G, Melchionna R, Di Vito L, et al. Gene expression profiles in peripheral blood mononuclear cells of chronic heart failure patients. Physiol Genomics. 2009;38(3):233–40.PubMedCrossRef Cappuzzello C, Napolitano M, Arcelli D, Melillo G, Melchionna R, Di Vito L, et al. Gene expression profiles in peripheral blood mononuclear cells of chronic heart failure patients. Physiol Genomics. 2009;38(3):233–40.PubMedCrossRef
47.
Zurück zum Zitat Shi M, Chen MS, Sekar K, Tan CK, Ooi LL, Hui KM. A blood-based three-gene signature for the non-invasive detection of early human hepatocellular carcinoma. Eur J Cancer. 2014;50(5):928–36.PubMedCrossRef Shi M, Chen MS, Sekar K, Tan CK, Ooi LL, Hui KM. A blood-based three-gene signature for the non-invasive detection of early human hepatocellular carcinoma. Eur J Cancer. 2014;50(5):928–36.PubMedCrossRef
48.
Zurück zum Zitat Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4+CD25+Foxp3+ T cells. Gastroenterology. 2008;135(1):234–43.PubMedCrossRef Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4+CD25+Foxp3+ T cells. Gastroenterology. 2008;135(1):234–43.PubMedCrossRef
49.
Zurück zum Zitat Zhou L, Miao K, Yin B, Li H, Fan J, Zhu Y, et al. Cardioprotective role of myeloid-derived suppressor cells in heart failure. Circulation. 2018;138(2):181–97.PubMedCrossRef Zhou L, Miao K, Yin B, Li H, Fan J, Zhu Y, et al. Cardioprotective role of myeloid-derived suppressor cells in heart failure. Circulation. 2018;138(2):181–97.PubMedCrossRef
51.
Zurück zum Zitat Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003;19(1):71–82.PubMedCrossRef Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003;19(1):71–82.PubMedCrossRef
52.
Zurück zum Zitat Ancuta P, Liu KY, Misra V, Wacleche VS, Gosselin A, Zhou X, et al. Transcriptional profiling reveals developmental relationship and distinct biological functions of CD16+ and CD16- monocyte subsets. BMC Genomics. 2009;10:403.PubMedPubMedCentralCrossRef Ancuta P, Liu KY, Misra V, Wacleche VS, Gosselin A, Zhou X, et al. Transcriptional profiling reveals developmental relationship and distinct biological functions of CD16+ and CD16- monocyte subsets. BMC Genomics. 2009;10:403.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Cavaliere FM, Prezzo A, Conti V, Bilotta C, Pulvirenti F, Iacobini M, et al. Intravenous immunoglobulin replacement induces an in vivo reduction of inflammatory monocytes and retains the monocyte ability to respond to bacterial stimulation in patients with common variable immunodeficiencies. Int Immunopharmacol. 2015;28(1):596–603.PubMedCrossRef Cavaliere FM, Prezzo A, Conti V, Bilotta C, Pulvirenti F, Iacobini M, et al. Intravenous immunoglobulin replacement induces an in vivo reduction of inflammatory monocytes and retains the monocyte ability to respond to bacterial stimulation in patients with common variable immunodeficiencies. Int Immunopharmacol. 2015;28(1):596–603.PubMedCrossRef
55.
Zurück zum Zitat Wong KL, Yeap WH, Tai JJY, Ong SM, Dang TM, Wong SC. The three human monocyte subsets: implications for health and disease. Immunol Res. 2012;53(1–3):41–57.PubMedCrossRef Wong KL, Yeap WH, Tai JJY, Ong SM, Dang TM, Wong SC. The three human monocyte subsets: implications for health and disease. Immunol Res. 2012;53(1–3):41–57.PubMedCrossRef
57.
Zurück zum Zitat Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37:208–20.PubMedPubMedCentralCrossRef Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37:208–20.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Weber R, Fleming V, Hu X, Nagibin V, Groth C, Altevogt P, et al. Myeloid-derived suppressor cells hinder the anti-cancer activity of immune checkpoint inhibitors. Front Immunol. 2018;9:1310.PubMedPubMedCentralCrossRef Weber R, Fleming V, Hu X, Nagibin V, Groth C, Altevogt P, et al. Myeloid-derived suppressor cells hinder the anti-cancer activity of immune checkpoint inhibitors. Front Immunol. 2018;9:1310.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, Ron D, et al. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity. 2005;22(5):633–42.PubMedCrossRef Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, Ron D, et al. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity. 2005;22(5):633–42.PubMedCrossRef
61.
62.
Zurück zum Zitat Solito S, Marigo I, Pinton L, Damuzzo V, Mandruzzato S, Bronte V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann N Y Acad Sci. 2014;1319(1):47–65.PubMedCrossRef Solito S, Marigo I, Pinton L, Damuzzo V, Mandruzzato S, Bronte V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann N Y Acad Sci. 2014;1319(1):47–65.PubMedCrossRef
63.
Zurück zum Zitat Waigel S, Rendon BE, Lamont G, Richie J, Mitchell RA, Yaddanapudi K. MIF inhibition reverts the gene expression profile of human melanoma cell line-induced MDSCs to normal monocytes. Genomics Data. 2016;7:240–2.PubMedPubMedCentralCrossRef Waigel S, Rendon BE, Lamont G, Richie J, Mitchell RA, Yaddanapudi K. MIF inhibition reverts the gene expression profile of human melanoma cell line-induced MDSCs to normal monocytes. Genomics Data. 2016;7:240–2.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Yaddanapudi K, Rendon BE, Lamont G, Kim EJ, Al Rayyan N, Richie J, et al. MIF is necessary for late-stage melanoma patient MDSC immune suppression and differentiation. Cancer Immunol Res. 2016;4(2):101–12.PubMedCrossRef Yaddanapudi K, Rendon BE, Lamont G, Kim EJ, Al Rayyan N, Richie J, et al. MIF is necessary for late-stage melanoma patient MDSC immune suppression and differentiation. Cancer Immunol Res. 2016;4(2):101–12.PubMedCrossRef
66.
Zurück zum Zitat Yakaboski E, Fuleihan RL, Sullivan KE, Cunningham-Rundles C, Feuille E. Lymphoproliferative disease in CVID: a report of types and frequencies from a US patient registry. J Clin Immunol. 2020;40(3):524–30.PubMedPubMedCentralCrossRef Yakaboski E, Fuleihan RL, Sullivan KE, Cunningham-Rundles C, Feuille E. Lymphoproliferative disease in CVID: a report of types and frequencies from a US patient registry. J Clin Immunol. 2020;40(3):524–30.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Fischer A, Provot J, Jais JP, Alcais A, Mahlaoui N, Adoue D, et al. Autoimmune and inflammatory manifestations occur frequently in patients with primary immunodeficiencies. J Allergy Clin Immunol. 2017;140(5):1388-1393.e8.PubMedCrossRef Fischer A, Provot J, Jais JP, Alcais A, Mahlaoui N, Adoue D, et al. Autoimmune and inflammatory manifestations occur frequently in patients with primary immunodeficiencies. J Allergy Clin Immunol. 2017;140(5):1388-1393.e8.PubMedCrossRef
68.
Zurück zum Zitat Fernando SL, Jang HSI, Li J. The immune dysregulation of common variable immunodeficiency disorders. Immunol Lett. 2021;230:21–6.PubMedCrossRef Fernando SL, Jang HSI, Li J. The immune dysregulation of common variable immunodeficiency disorders. Immunol Lett. 2021;230:21–6.PubMedCrossRef
69.
Zurück zum Zitat Hel Z, Huijbregts RPH, Xu J, Nechvatalova J, Vlkova M, Litzman J. Altered serum cytokine signature in common variable immunodeficiency. J Clin Immunol. 2014;34(8):971–8.PubMedPubMedCentralCrossRef Hel Z, Huijbregts RPH, Xu J, Nechvatalova J, Vlkova M, Litzman J. Altered serum cytokine signature in common variable immunodeficiency. J Clin Immunol. 2014;34(8):971–8.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Vlkova M, Chovancova Z, Nechvatalova J, Connelly AN, Davis MD, Slanina P, et al. Neutrophil and granulocytic myeloid-derived suppressor cell–mediated T cell suppression significantly contributes to immune dysregulation in common variable immunodeficiency disorders. J Immunol. 2019;202(1):93–104.PubMedCrossRef Vlkova M, Chovancova Z, Nechvatalova J, Connelly AN, Davis MD, Slanina P, et al. Neutrophil and granulocytic myeloid-derived suppressor cell–mediated T cell suppression significantly contributes to immune dysregulation in common variable immunodeficiency disorders. J Immunol. 2019;202(1):93–104.PubMedCrossRef
71.
Zurück zum Zitat Jørgensen SF, Trøseid M, Kummen M, Anmarkrud JA, Michelsen AE, Osnes LT, et al. Altered gut microbiota profile in common variable immunodeficiency associates with levels of lipopolysaccharide and markers of systemic immune activation. Mucosal Immunol. 2016;9(6):1455–65.PubMedCrossRef Jørgensen SF, Trøseid M, Kummen M, Anmarkrud JA, Michelsen AE, Osnes LT, et al. Altered gut microbiota profile in common variable immunodeficiency associates with levels of lipopolysaccharide and markers of systemic immune activation. Mucosal Immunol. 2016;9(6):1455–65.PubMedCrossRef
72.
Zurück zum Zitat Litzman J, Nechvatalova J, Xu J, Ticha O, Vlkova M, Hel Z. Chronic immune activation in common variable immunodeficiency (CVID) is associated with elevated serum levels of soluble CD14 and CD25 but not endotoxaemia. Clin Exp Immunol. 2012;170(3):321–32.PubMedPubMedCentralCrossRef Litzman J, Nechvatalova J, Xu J, Ticha O, Vlkova M, Hel Z. Chronic immune activation in common variable immunodeficiency (CVID) is associated with elevated serum levels of soluble CD14 and CD25 but not endotoxaemia. Clin Exp Immunol. 2012;170(3):321–32.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Barbosa RR, Silva SP, Silva SL, Tendeiro R, Melo AC, Pedro E, et al. Monocyte activation is a feature of common variable immunodeficiency irrespective of plasma lipopolysaccharide levels. Clin Exp Immunol. 2012;169(3):263–72.PubMedPubMedCentralCrossRef Barbosa RR, Silva SP, Silva SL, Tendeiro R, Melo AC, Pedro E, et al. Monocyte activation is a feature of common variable immunodeficiency irrespective of plasma lipopolysaccharide levels. Clin Exp Immunol. 2012;169(3):263–72.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Albin S, Cunningham-Rundles C. An update on the use of immunoglobulin for the treatment of immunodeficiency disorders. Immunotherapy. 2014;6:1113–26.PubMedCrossRef Albin S, Cunningham-Rundles C. An update on the use of immunoglobulin for the treatment of immunodeficiency disorders. Immunotherapy. 2014;6:1113–26.PubMedCrossRef
75.
Zurück zum Zitat Paquin-Proulx D, Santos BAN, Carvalho KI, Toledo-Barros M, Barreto de Oliveira AK, Kokron CM, et al. IVIg immune reconstitution treatment alleviates the state of persistent immune activation and suppressed CD4 T cell counts in CVID. PLoS One. 2013;8(10):e75199. https://doi.org/10.1371/journal.pone.0075199. Paquin-Proulx D, Santos BAN, Carvalho KI, Toledo-Barros M, Barreto de Oliveira AK, Kokron CM, et al. IVIg immune reconstitution treatment alleviates the state of persistent immune activation and suppressed CD4 T cell counts in CVID. PLoS One. 2013;8(10):e75199. https://​doi.​org/​10.​1371/​journal.​pone.​0075199.
77.
Zurück zum Zitat Aslam R, Burack WR, Segel GB, McVey M, Spence SA, Semple JW. Intravenous immunoglobulin treatment of spleen cells from patients with immune thrombocytopenia significantly increases the percentage of myeloid-derived suppressor cells. Br J Haematol. 2018;181:262–4.PubMedCrossRef Aslam R, Burack WR, Segel GB, McVey M, Spence SA, Semple JW. Intravenous immunoglobulin treatment of spleen cells from patients with immune thrombocytopenia significantly increases the percentage of myeloid-derived suppressor cells. Br J Haematol. 2018;181:262–4.PubMedCrossRef
78.
Zurück zum Zitat Janols H, Bergenfelz C, Allaoui R, Larsson A-M, Rydén L, Björnsson S, et al. A high frequency of MDSCs in sepsis patients, with the granulocytic subtype dominating in gram-positive cases. J Leukoc Biol. 2014;96(5):685–93.PubMedCrossRef Janols H, Bergenfelz C, Allaoui R, Larsson A-M, Rydén L, Björnsson S, et al. A high frequency of MDSCs in sepsis patients, with the granulocytic subtype dominating in gram-positive cases. J Leukoc Biol. 2014;96(5):685–93.PubMedCrossRef
79.
Zurück zum Zitat Cuenca AG, Delano MJ, Kelly-Scumpia KM, Moreno C, Scumpia PO, LaFace DM, et al. A paradoxical role for myeloid-derived suppressor cells in sepsis and trauma. Mol Med. 2011;17(3–4):281–92.PubMedCrossRef Cuenca AG, Delano MJ, Kelly-Scumpia KM, Moreno C, Scumpia PO, LaFace DM, et al. A paradoxical role for myeloid-derived suppressor cells in sepsis and trauma. Mol Med. 2011;17(3–4):281–92.PubMedCrossRef
80.
Zurück zum Zitat Siedlar M, Strach M, Bukowska-Strakova K, Lenart M, Szaflarska A, Weglarczyk K, et al. Preparations of intravenous immunoglobulins diminish the number and proinflammatory response of CD14+CD16++ monocytes in common variable immunodeficiency (CVID) patients. Clin Immunol. 2011;139(2):122–32.PubMedCrossRef Siedlar M, Strach M, Bukowska-Strakova K, Lenart M, Szaflarska A, Weglarczyk K, et al. Preparations of intravenous immunoglobulins diminish the number and proinflammatory response of CD14+CD16++ monocytes in common variable immunodeficiency (CVID) patients. Clin Immunol. 2011;139(2):122–32.PubMedCrossRef
81.
Zurück zum Zitat Tjon ASW, Metselaar HJ, te Boekhorst PAW, van Hagen PM, Kwekkeboom J. High-dose intravenous immunoglobulin does not reduce the numbers of circulating CD14+CD16++ monocytes in patients with inflammatory disorders. Clin Immunol. 2012;145:11–2.PubMedCrossRef Tjon ASW, Metselaar HJ, te Boekhorst PAW, van Hagen PM, Kwekkeboom J. High-dose intravenous immunoglobulin does not reduce the numbers of circulating CD14+CD16++ monocytes in patients with inflammatory disorders. Clin Immunol. 2012;145:11–2.PubMedCrossRef
82.
Zurück zum Zitat Siedlar M, Ziegler-Heitbrock L. Commentary to the letter of Tjon et al. Clin Immunol. 2012;145:141.PubMedCrossRef Siedlar M, Ziegler-Heitbrock L. Commentary to the letter of Tjon et al. Clin Immunol. 2012;145:141.PubMedCrossRef
83.
Zurück zum Zitat Katayama K, Matsubara T, Fujiwara M, Koga M, Furukawa S. CD14+CD16+ monocyte subpopulation in Kawasaki disease. Clin Exp Immunol. 2000;121(3):566–70.PubMedPubMedCentralCrossRef Katayama K, Matsubara T, Fujiwara M, Koga M, Furukawa S. CD14+CD16+ monocyte subpopulation in Kawasaki disease. Clin Exp Immunol. 2000;121(3):566–70.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Gonzalez-Dominguez E, Dominguez-Soto A, Nieto C, Luis Flores-Sevilla J, Pacheco-Blanco M, Campos-Pena V, et al. Atypical Activin A and IL-10 production impairs human CD16(+) monocyte differentiation into anti-inflammatory macrophages. J Immunol. 2016;196(3):1327–37.PubMedCrossRef Gonzalez-Dominguez E, Dominguez-Soto A, Nieto C, Luis Flores-Sevilla J, Pacheco-Blanco M, Campos-Pena V, et al. Atypical Activin A and IL-10 production impairs human CD16(+) monocyte differentiation into anti-inflammatory macrophages. J Immunol. 2016;196(3):1327–37.PubMedCrossRef
85.
Zurück zum Zitat Maddur MS, Kaveri SV, Bayry J. Circulating normal IgG as stimulator of regulatory T cells: lessons from intravenous immunoglobulin. Trends Immunol. Elsevier Ltd. 2017;38:789–92.PubMedCrossRef Maddur MS, Kaveri SV, Bayry J. Circulating normal IgG as stimulator of regulatory T cells: lessons from intravenous immunoglobulin. Trends Immunol. Elsevier Ltd. 2017;38:789–92.PubMedCrossRef
87.
Zurück zum Zitat Trinath J, Hegde P, Sharma M, Maddur MS, Rabin M, Vallat JM, et al. Intravenous immunoglobulin expands regulatory T cells via induction of cyclooxygenase-2-dependent prostaglandin E2 in human dendritic cells. Blood. 2013;122(8):1419–27.PubMedCrossRef Trinath J, Hegde P, Sharma M, Maddur MS, Rabin M, Vallat JM, et al. Intravenous immunoglobulin expands regulatory T cells via induction of cyclooxygenase-2-dependent prostaglandin E2 in human dendritic cells. Blood. 2013;122(8):1419–27.PubMedCrossRef
88.
Zurück zum Zitat Tjon ASW, Tha-In T, Metselaar HJ, van Gent R, van der Laan LJW, Groothuismink ZMA, et al. Patients treated with high-dose intravenous immunoglobulin show selective activation of regulatory T cells. Clin Exp Immunol. 2013;173(2):259–67.PubMedPubMedCentralCrossRef Tjon ASW, Tha-In T, Metselaar HJ, van Gent R, van der Laan LJW, Groothuismink ZMA, et al. Patients treated with high-dose intravenous immunoglobulin show selective activation of regulatory T cells. Clin Exp Immunol. 2013;173(2):259–67.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Wang SC, Yang KD, Lin CY, Huang AY, Hsiao CC, Lin MT, et al. Intravenous immunoglobulin therapy enhances suppressive regulatory T cells and decreases innate lymphoid cells in children with immune thrombocytopenia. Pediatr Blood Cancer. 2020;67(2):e28075. https://doi.org/10.1002/pbc.28075. Wang SC, Yang KD, Lin CY, Huang AY, Hsiao CC, Lin MT, et al. Intravenous immunoglobulin therapy enhances suppressive regulatory T cells and decreases innate lymphoid cells in children with immune thrombocytopenia. Pediatr Blood Cancer. 2020;67(2):e28075. https://​doi.​org/​10.​1002/​pbc.​28075.
90.
91.
Zurück zum Zitat Xu W, Ren M, Ghosh S, Qian K, Luo Z, Zhang A, et al. Defects of CTLA-4 are associated with regulatory T cells in myasthenia gravis implicated by intravenous immunoglobulin therapy. Mediat Inflamm. 2020;2020:3645157. https://doi.org/10.1155/2020/3645157. Xu W, Ren M, Ghosh S, Qian K, Luo Z, Zhang A, et al. Defects of CTLA-4 are associated with regulatory T cells in myasthenia gravis implicated by intravenous immunoglobulin therapy. Mediat Inflamm. 2020;2020:3645157. https://​doi.​org/​10.​1155/​2020/​3645157.
92.
Zurück zum Zitat Massoud AH, Kaufman GN, Xue D, Béland M, Dembele M, Piccirillo CA, et al. Peripherally generated Foxp3 + regulatory T cells mediate the immunomodulatory effects of IVIg in allergic airways disease. J Immunol. 2017;198(7):2760–71.PubMedCrossRef Massoud AH, Kaufman GN, Xue D, Béland M, Dembele M, Piccirillo CA, et al. Peripherally generated Foxp3 + regulatory T cells mediate the immunomodulatory effects of IVIg in allergic airways disease. J Immunol. 2017;198(7):2760–71.PubMedCrossRef
93.
Zurück zum Zitat Hirabayashi Y, Takahashi Y, Xu Y, Akane K, Villalobos IB, Okuno Y, et al. Lack of CD4+CD25+FOXP3+ regulatory T cells is associated with resistance to intravenous immunoglobulin therapy in patients with Kawasaki disease. Eur J Pediatr. 2013;172(6):833–7.PubMedCrossRef Hirabayashi Y, Takahashi Y, Xu Y, Akane K, Villalobos IB, Okuno Y, et al. Lack of CD4+CD25+FOXP3+ regulatory T cells is associated with resistance to intravenous immunoglobulin therapy in patients with Kawasaki disease. Eur J Pediatr. 2013;172(6):833–7.PubMedCrossRef
94.
Zurück zum Zitat Ephrem A, Chamat S, Miquel C, Fisson S, Mouthon L, Caligiuri G, et al. Expansion of CD4+CD25+ regulatory T cells by intravenous immunoglobulin: a critical factor in controlling experimental autoimmune encephalomyelitis. Blood. 2008;111(2):715–22.PubMedCrossRef Ephrem A, Chamat S, Miquel C, Fisson S, Mouthon L, Caligiuri G, et al. Expansion of CD4+CD25+ regulatory T cells by intravenous immunoglobulin: a critical factor in controlling experimental autoimmune encephalomyelitis. Blood. 2008;111(2):715–22.PubMedCrossRef
95.
Zurück zum Zitat Maddur MS, Stephen-Victor E, Das M, Prakhar P, Sharma VK, Singh V, et al. Regulatory T cell frequency, but not plasma IL-33 levels, represents potential immunological biomarker to predict clinical response to intravenous immunoglobulin therapy. J Neuroinflamm 2017;14(1):58. https://doi.org/10.1186/s12974-017-0818-5. Maddur MS, Stephen-Victor E, Das M, Prakhar P, Sharma VK, Singh V, et al. Regulatory T cell frequency, but not plasma IL-33 levels, represents potential immunological biomarker to predict clinical response to intravenous immunoglobulin therapy. J Neuroinflamm 2017;14(1):58. https://​doi.​org/​10.​1186/​s12974-017-0818-5.
96.
Zurück zum Zitat Zoso A, Mazza EMC, Bicciato S, Mandruzzato S, Bronte V, Serafini P, et al. Human fibrocytic myeloid-derived suppressor cells express IDO and promote tolerance via Treg-cell expansion. Eur J Immunol. 2014;44(11):3307–19.PubMedCrossRef Zoso A, Mazza EMC, Bicciato S, Mandruzzato S, Bronte V, Serafini P, et al. Human fibrocytic myeloid-derived suppressor cells express IDO and promote tolerance via Treg-cell expansion. Eur J Immunol. 2014;44(11):3307–19.PubMedCrossRef
97.
Zurück zum Zitat Serafini P, Mgebroff S, Noonan K, Borrello I. Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells. Cancer Res. 2008;68(13):5439–49.PubMedPubMedCentralCrossRef Serafini P, Mgebroff S, Noonan K, Borrello I. Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells. Cancer Res. 2008;68(13):5439–49.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Groth C, Hu X, Weber R, Fleming V, Altevogt P, Utikal J, et al. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br J Cancer. Nature Publishing Group. 2019;120:16–25.PubMedCrossRef Groth C, Hu X, Weber R, Fleming V, Altevogt P, Utikal J, et al. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br J Cancer. Nature Publishing Group. 2019;120:16–25.PubMedCrossRef
99.
Zurück zum Zitat Hatziioannou A, Alissafi T, Verginis P. Myeloid-derived suppressor cells and T regulatory cells in tumors: unraveling the dark side of the force. J Leukoc Biol. 2017;102(2):407–21.PubMedCrossRef Hatziioannou A, Alissafi T, Verginis P. Myeloid-derived suppressor cells and T regulatory cells in tumors: unraveling the dark side of the force. J Leukoc Biol. 2017;102(2):407–21.PubMedCrossRef
100.
101.
Zurück zum Zitat Schlecker E, Stojanovic A, Eisen C, Quack C, Falk CS, Umansky V, et al. Tumor- infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J Immunol. 2012;189(12):5602–11.PubMedCrossRef Schlecker E, Stojanovic A, Eisen C, Quack C, Falk CS, Umansky V, et al. Tumor- infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J Immunol. 2012;189(12):5602–11.PubMedCrossRef
Metadaten
Titel
Intravenous Immunoglobulins Promote an Expansion of Monocytic Myeloid-Derived Suppressor Cells (MDSC) in CVID Patients
verfasst von
Miriam Simón-Fuentes
Silvia Sánchez-Ramón
Lidia Fernández-Paredes
Bárbara Alonso
Kissy Guevara-Hoyer
Miguel A. Vega
Angel L. Corbí
Ángeles Domínguez-Soto
Publikationsdatum
29.04.2022
Verlag
Springer US
Erschienen in
Journal of Clinical Immunology / Ausgabe 5/2022
Print ISSN: 0271-9142
Elektronische ISSN: 1573-2592
DOI
https://doi.org/10.1007/s10875-022-01277-7

Weitere Artikel der Ausgabe 5/2022

Journal of Clinical Immunology 5/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

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