Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Immunobiology ; 228(3): 152364, 2023 05.
Article En | MEDLINE | ID: mdl-36881973

Factor H is a pivotal complement regulatory protein that is preferentially produced by the liver and circulates in high concentrations in serum. There has been an increasing interest in the extrahepatic production of complement factors, including by cells of the immune system, since this contributes to non-canonical functions of local complement activation and regulation. Here we investigated the production and regulation of factor H and its splice variant factor H-like protein 1 (FHL-1) by human myeloid cells. As validation, we confirmed the predominant presence of intact factor H in serum, despite a strong but comparable mRNA expression of CFH and FHL1 in liver. Comparable levels of CFH and FHL1 were also observed in renal tissue, although a dominant staining for FHL-1 was shown within the proximal tubules. Human in vitro generated pro- and anti-inflammatory macrophages both expressed and produced factor H/FHL-1, but this was strongest in pro-inflammatory macrophages. Production was not affected by LPS activation, but was increased upon stimulation with IFN-γ or CD40L. Importantly, in both macrophage subsets mRNA expression of FHL1 was significantly higher than CFH. Moreover, production of FHL-1 protein could be confirmed using precipitation and immunoblotting of culture supernatants. These data identify macrophages as producers of factor H and FHL-1, thereby potentially contributing to local complement regulation at sites of inflammation.


Complement Activation , Complement Factor H , Humans , Complement Factor H/genetics , Myeloid Cells/metabolism , RNA, Messenger , Muscle Proteins , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins
2.
Immunobiology ; 227(4): 152246, 2022 07.
Article En | MEDLINE | ID: mdl-35843030

The complement system does not only play an important role in the defence against microorganism and pathogens, but also contributes to the regulation of innate and adaptive immunity. Especially activation fragments C3a and C5a and complement activation at the interface of antigen presenting cell (APC) and T cell, were shown to have a role in T cell activation and proliferation. Whereas most complement factors are produced by the liver, properdin, a positive regulator of the C3 convertase, is mainly produced by myeloid cells. Here we show that properdin can be detected in myeloid cell infiltrate during human renal allograft rejection. In vitro, properdin is produced and secreted by human immature dendritic cells (iDCs), which is further increased by CD40-L-matured DCs (mDCs). Transfection with a specific properdin siRNA reduced properdin secretion by iDCs and mDCs, without affecting the expression of co-stimulatory markers CD80 and CD86. Co-culture of properdin siRNA-transfected iDCs and mDCs with human allogeneic T cells resulted in reduced T cell proliferation, especially under lower DC-T cell ratio's (1:30 and 1:90 ratio). In addition, T cell cytokines were altered, including a reduced TNF-α and IL-17 secretion by T cells co-cultured with properdin siRNA-transfected iDCs. Taken together, these results indicate a local role for properdin during the interaction of DCs and allogeneic T cells, contributing to the shaping of T cell proliferation and activation.


Kidney Transplantation , Properdin , Cells, Cultured , Dendritic Cells , Humans , Properdin/genetics , Properdin/metabolism , RNA, Small Interfering , T-Lymphocytes
3.
Eur J Immunol ; 52(4): 597-608, 2022 04.
Article En | MEDLINE | ID: mdl-35092629

Properdin, the only known positive regulator of the complement system, stabilizes the C3 convertase, thereby increasing its half-life. In contrast to most other complement factors, properdin is mainly produced extrahepatically by myeloid cells. Recent data suggest a role for properdin as a pattern recognition molecule. Here, we confirmed previous findings of properdin binding to different necrotic cells including Jurkat T cells. Binding can occur independent of C3, as demonstrated by HAP-1 C3 KO cells, excluding a role for endogenous C3. In view of the cellular source of properdin, interaction with myeloid cells was examined. Properdin bound to the surface of viable monocyte-derived pro- and anti-inflammatory macrophages, but not to DCs. Binding was demonstrated for purified properdin as well as fractionated P2, P3, and P4 properdin oligomers. Binding contributed to local complement activation as determined by C3 and C5b-9 deposition on the cell surfaces and seems a prerequisite for alternative pathway activation. Interaction of properdin with cell surfaces could be inhibited with the tick protein Salp20 and by different polysaccharides, depending on sulfation and chain length. These data identify properdin as a factor interacting with different cell surfaces, being either dead or alive, contributing to the local stimulation of complement activation.


Complement C3-C5 Convertases , Properdin , Complement Activation , Complement C3-C5 Convertases/metabolism , Complement Membrane Attack Complex , Complement Pathway, Alternative , Humans , Necrosis , Properdin/metabolism
4.
J Immunol Methods ; 479: 112748, 2020 04.
Article En | MEDLINE | ID: mdl-31958452

Gene silencing using small interfering ribonucleic acids (siRNA) is a powerful method to interfere with gene expression, allowing for the functional exploration of specific genes. siRNA interference can be applied in both cell lines, as well as in primary, non-dividing cell types like dendritic cells. However, the efficacy in different cell types is variable and requires optimization. Here, we showed that the type of culture medium used during lipid-based siRNA-mediated transfection acts as a critical factor, affecting dendritic cell activation. Transfection of immature monocyte-derived dendritic cells in RPMI medium, but not in IMDM, showed increased transcript levels of pro-inflammatory cytokines. Moreover, the expression of co-stimulatory molecules was enhanced, thereby increasing the T cell stimulatory capacity. Our data demonstrates that the choice of medium should be critically examined as one of the variables while optimizing cell transfection.


Culture Media/metabolism , Dendritic Cells/immunology , T-Lymphocytes/immunology , Cell Differentiation , Cells, Cultured , Culture Media/chemistry , Humans , Lymphocyte Activation , RNA, Small Interfering/genetics , Transfection
5.
Nephrol Dial Transplant ; 32(12): 2132-2141, 2017 Dec 01.
Article En | MEDLINE | ID: mdl-29045706

BACKGROUND: Kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) are promising biomarkers for monitoring delayed graft function (DGF) after kidney transplantation. Here we investigated localization and distribution of KIM-1 and NGAL staining in renal allograft biopsies and studied their association with histological features, functional DGF (fDGF) and the tubular function slope (TFS), a functioning proximal tubular epithelial cell (PTEC) marker. METHODS: Day 10 protocol biopsies of 64 donation after circulatory death recipients were stained for KIM-1 and NGAL and the positive area was quantified using ImageJ software. Biopsies were scored according to Banff and acute tubular necrosis (ATN) criteria. A 99mtechnetium-mercaptoacetyltriglycine (99mTc-MAG3)-renography was performed to calculate TFS. RESULTS: KIM-1 staining was located on the brush border of tubular epithelial cells (TECs) and correlated with denudation, while NGAL was present more focally in a cytoplasmic distribution. KIM-1 and NGAL staining were not correlated and no co-localization was observed. Quantitative stainings were not associated with fDGF, but KIM-1 tended to be higher in patients with prolonged fDGF (≥21 days; P = 0.062). No correlation was observed between the quantitative tissue stainings and urinary KIM-1 or NGAL. Quantitative KIM-1 staining was inversely correlated with the TFS (Spearman's ρ = -0.53; P < 0.001), whereas NGAL was not. The latter finding might be because cortical NGAL staining is dependent on filtration and subsequent reabsorption by functioning PTECs. Staining of NGAL was indeed restricted to PTECs, as shown by co-localization with a PTEC-specific lectin. CONCLUSIONS: KIM-1 and NGAL staining showed different localization and distribution. Quantitative KIM-1 staining was inversely correlated with functioning PTECs.


Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Delayed Graft Function/diagnosis , Epithelial Cells/pathology , Hepatitis A Virus Cellular Receptor 1/metabolism , Kidney Transplantation/adverse effects , Kidney Tubules, Proximal/pathology , Aged , Animals , Biopsy , Delayed Graft Function/metabolism , Epithelial Cells/metabolism , Female , Humans , Kidney Tubules, Proximal/injuries , Kidney Tubules, Proximal/metabolism , Lipocalin-2/metabolism , Male , Middle Aged , Rats , Rats, Inbred Lew , Staining and Labeling , Transplantation, Homologous
6.
J Am Soc Nephrol ; 25(8): 1710-22, 2014 Aug.
Article En | MEDLINE | ID: mdl-24610930

Ischemia/reperfusion injury (IRI) is a central phenomenon in kidney transplantation and AKI. Integrity of the renal peritubular capillary network is an important limiting factor in the recovery from IRI. MicroRNA-126 (miR-126) facilitates vascular regeneration by functioning as an angiomiR and by modulating mobilization of hematopoietic stem/progenitor cells. We hypothesized that overexpression of miR-126 in the hematopoietic compartment could protect the kidney against IRI via preservation of microvascular integrity. Here, we demonstrate that hematopoietic overexpression of miR-126 increases neovascularization of subcutaneously implanted Matrigel plugs in mice. After renal IRI, mice overexpressing miR-126 displayed a marked decrease in urea levels, weight loss, fibrotic markers, and injury markers (such as kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin). This protective effect was associated with a higher density of the peritubular capillary network in the corticomedullary junction and increased numbers of bone marrow-derived endothelial cells. Hematopoietic overexpression of miR-126 increased the number of circulating Lin(-)/Sca-1(+)/cKit(+) hematopoietic stem and progenitor cells. Additionally, miR-126 overexpression attenuated expression of the chemokine receptor CXCR4 on Lin(-)/Sca-1(+)/cKit(+) cells in the bone marrow and increased renal expression of its ligand stromal cell-derived factor 1, thus favoring mobilization of Lin(-)/Sca-1(+)/cKit(+) cells toward the kidney. Taken together, these results suggest overexpression of miR-126 in the hematopoietic compartment is associated with stromal cell-derived factor 1/CXCR4-dependent vasculogenic progenitor cell mobilization and promotes vascular integrity and supports recovery of the kidney after IRI.


Acute Kidney Injury/prevention & control , Hematopoietic Stem Cells/physiology , Kidney/blood supply , MicroRNAs/physiology , Neovascularization, Physiologic/physiology , Reperfusion Injury/prevention & control , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Cell Movement/physiology , Chemokine CXCL12/metabolism , Kidney/metabolism , Kidney/pathology , Male , Mice, Inbred C57BL , Receptors, CXCR4/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
7.
Am J Physiol Renal Physiol ; 305(6): F901-10, 2013 Sep 15.
Article En | MEDLINE | ID: mdl-23825073

Endothelial cells (ECs) are highly susceptible to hypoxia and easily affected upon ischemia-reperfusion (I/R) during renal transplantation. Pericytes and angiopoeitins play important role in modulating EC function. In the present study, we investigate the effect of renal I/R on the dynamics of angiopoietin expression and its association with pericytes and fibrosis development. Male Lewis rats were subjected to unilateral renal ischemia for 45 min followed by removal of the contralateral kidney. Rats were killed at different time points after reperfusion. Endothelial integrity (RECA-1), pericytes [platelet-derived growth factor receptor-ß (PDGFR-ß)], angiopoietin-2 (Ang-2)/angiopoietin-1 (Ang-1) expression, and interstitial collagen deposition (Sirius red and α-smooth muscle actin) were assessed using immunohistochemistry and RT-PCR. Our study shows an increase in protein expression of Ang-2 starting at 5 h and remaining elevated up to 72 h, with a consequently higher Ang-2/Ang-1 ratio after renal I/R (P < 0.05 at 48 h). This was accompanied by an increase in protein expression of the pericytic marker PDGFR-ß and a loss of ECs (both at 72 h after I/R, P < 0.05). Nine weeks after I/R, when renal function was restored, we observed normalization of the Ang-2/Ang-1 ratio and PDGFR-ß expression and increase in cortical ECs, which was accompanied by fibrosis. Renal I/R induces a dysbalance of Ang-2/Ang-1 accompanied by proliferation of pericytes, EC loss, and development of fibrosis. The Ang-2/Ang-1 balance was reversed to baseline at 9 wk after renal I/R, which coincided with restoration of cortical ECs and pericytes. Our findings suggest that angiopoietins and pericytes play an important role in renal microvascular remodeling and development of fibrosis.


Angiopoietins/metabolism , Pericytes/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Angiopoietin-1/biosynthesis , Angiopoietin-2/biosynthesis , Animals , Endothelial Cells , Fibrosis , Kidney Diseases/pathology , Male , Pericytes/metabolism , Rats , Rats, Inbred Lew , Receptor, Platelet-Derived Growth Factor beta/biosynthesis
8.
Transplantation ; 94(9): 903-10, 2012 Nov 15.
Article En | MEDLINE | ID: mdl-23047455

BACKGROUND: Allogeneic islets of Langerhans transplantation is hampered in its success as a curative treatment of type 1 diabetes by the absence of potent, specific, and nontoxic immunosuppressive drugs. Here, we assessed whether donor bone marrow-derived dexamethasone-treated dendritic cells (dexDCs) could prolong islet allograft survival in a full major histocompatibility complex mismatch rat model. METHODS: Rodent allogeneic islet transplantation was performed from DA rats to Lewis rats and vice versa. Permanently immature dendritic cells were generated from the bone marrow of DA and Lewis rats by treatment with dexamethasone. Animals were either vehicle or donor dexDCs pretreated. Serum was used to monitor glucose, C-peptide, and alloreactive antibodies. RESULTS: The transplantation of DA islets into Lewis recipients showed direct graft failure with reduced numbers of ß-cells when rats were pretreated with donor dexDCs. In the reverse model (Lewis islets into DA recipients), dexDC-treated DA recipients even showed a significantly accelerated rejection of Lewis islets. Immunohistochemical analysis of allograft tissue of dexDC-treated recipients showed a predominant natural killer cell infiltration and a presence of antibody reactivity in the absence of complement deposition. Alloreactive antibodies were solely found in dexDC-treated recipients. CONCLUSION: Our study shows that pretreatment with donor-derived dexDCs induces an antibody-mediated rejection in this islet transplantation rodent model.


Dendritic Cells/drug effects , Dendritic Cells/transplantation , Dexamethasone/pharmacology , Diabetes Mellitus, Experimental/surgery , Graft Rejection/immunology , Islets of Langerhans Transplantation/immunology , Animals , Antibodies/blood , Cell Movement/physiology , Dendritic Cells/pathology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Female , Killer Cells, Natural/pathology , Male , Rats , Rats, Inbred Lew , Rats, Inbred Strains , Streptozocin/adverse effects , T-Lymphocytes/pathology , Time Factors
9.
Transpl Immunol ; 27(1): 55-8, 2012 Aug.
Article En | MEDLINE | ID: mdl-22709941

Local activation of the complement system has been associated with ischemia/reperfusion injury following kidney transplantation and tubular injury under proteinuric conditions. The soluble terminal complement complex sC5b-9 is a stable end-product of the complement cascade, and as such a promising urinary biomarker. In the early post-transplant period we found high urinary levels of sC5b-9, significantly correlating with the degree of proteinuria, suggesting activation of filtered complement components at the tubular epithelial surface of the kidney. However, when mimicking proteinuria in vitro by exposing serum (or blood) to urine (both negative for sC5b-9), we found extensive generation of sC5b-9 in urine. This process was inhibited by EDTA, confirming activation of the complement system. In conclusion, although sC5b-9 is an attractive urinary biomarker, one should be aware of the risk of extra-renal complement activation independent of a renal contribution. This may be of special interest when measuring urinary sC5b-9 following kidney transplantation in which procedure-related (microscopic) hematuria and proteinuria are common.


Complement Membrane Attack Complex/urine , Kidney Transplantation , Reperfusion Injury/urine , Biomarkers/urine , Edetic Acid/pharmacology , Female , Hematuria , Humans , Male , Middle Aged , Proteinuria
10.
Transplantation ; 86(9): 1275-82, 2008 Nov 15.
Article En | MEDLINE | ID: mdl-19005410

BACKGROUND: Dendritic cells (DC) can exert powerful immune stimulatory as well as regulatory functions and are therefore important tools for therapeutic strategies. Dexamethasone (Dex) was previously shown to inhibit DC maturation and to induce regulatory properties both in vitro and in vivo. Here, we investigated the immunoregulatory role of DexDC in two different rat acute rejection models of kidney transplantation. METHODS: Rat DC were generated from BN and DA bone marrow in the presence of the corticosteroid, Dex. The function of Dex-modulated DC was analyzed in vitro and in vivo, using a BN to LEW and a DA to LEW renal transplantation model in the absence of other forms of immunosuppression. T cells of transplanted rats were isolated and restimulated with donor mature DC (lipopolysaccharide [LPS] or CD40L activated). T-cell responsiveness was analyzed by proliferative capacity and IFN-gamma production. RESULTS: Stimulation of Dex-modulated rat DC with LPS resulted in normal IL-10 production, whereas synthesis of IL-12 was impaired. In accordance, the capacity of LPS-DexDC to stimulate T-cell activation was decreased. In both renal transplantation models, treatment with donor-derived LPS-DexDC induced a significant donor-specific T-cell hyporesponse. However, pretreatment did not result in a prolonged graft survival. CONCLUSIONS: In two fully mismatched kidney transplantation models, donor-derived LPS-DexDC induce a donor-specific T-cell hyporesponse. However, in this setting allograft survival was not improved, suggesting an important role for T cells with indirect alloreactivity. Understanding the underlying mechanism involved in the rejection process will improve the development of a cell-based immunotherapy.


Adrenal Cortex Hormones/pharmacology , Dendritic Cells/immunology , Dexamethasone/pharmacology , Graft Rejection/immunology , Kidney Transplantation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Animals , Cell Communication/immunology , Cell Proliferation , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Male , Models, Animal , Rats , Rats, Inbred BN , Rats, Inbred Lew , T-Lymphocytes/metabolism
11.
J Immunol ; 180(11): 7613-21, 2008 Jun 01.
Article En | MEDLINE | ID: mdl-18490764

Cells that undergo apoptosis or necrosis are promptly removed by phagocytes. Soluble opsonins such as complement can opsonize dying cells, thereby promoting their removal by phagocytes and modulating the immune response. The pivotal role of the complement system in the handling of dying cells has been demonstrated for the classical pathway (via C1q) and lectin pathway (via mannose-binding lectin and ficolin). Herein we report that the only known naturally occurring positive regulator of complement, properdin, binds predominantly to late apoptotic and necrotic cells, but not to early apoptotic cells. This binding occurs independently of C3b, which is additional to the standard model wherein properdin binds to preexisting clusters of C3b on targets and stabilizes the convertase C3bBb. By binding to late apoptotic or necrotic cells, properdin serves as a focal point for local amplification of alternative pathway complement activation. Furthermore, properdin exhibits a strong interaction with DNA that is exposed on the late stage of dying cells. Our data indicate that direct recognition of dying cells by properdin is essential to drive alternative pathway complement activation.


Complement C3b/metabolism , Complement Pathway, Alternative , Properdin/metabolism , Apoptosis , Cell Line, Tumor , Complement C1q/immunology , Complement C1q/metabolism , Complement C3 Convertase, Alternative Pathway , Complement C3b/immunology , Humans , Immunologic Factors/immunology , Immunologic Factors/metabolism , Jurkat Cells , Necrosis , Phagocytosis , Properdin/immunology
12.
J Immunol Methods ; 335(1-2): 46-52, 2008 Jun 01.
Article En | MEDLINE | ID: mdl-18384807

The CD40-CD40L interaction plays a critical role in cell-mediated immune responses. Blocking this interaction has been shown to be beneficial in the treatment of various diseases studied in murine models. Although rats are widely used to test therapeutic strategies in several disease models, a monoclonal antibody (mAb) to block the CD40-CD40L interaction in rats is not broadly available. In the present study we generated Armenian hamster fibroblasts expressing rat CD40L and used these to generate a novel anti-rat CD40L mAb (AS1). In vitro studies showed that AS1 was able to block CD40L-induced DC maturation and B cell proliferation. Most importantly, in vivo, AS1 inhibited B cell responses in a dose-dependent fashion, as measured by the production of OVA specific antibodies after subcutaneous immunization with OVA. AS1 was shown to be a powerful tool to modulate Ag presentation in vitro and in vivo. Elucidating the effect of AS1 in various rat models for human diseases will provide more insight into blocking the CD40-CD40L interaction as a therapeutic strategy to prevent human diseases.


Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/pharmacology , CD40 Antigens/metabolism , CD40 Ligand/antagonists & inhibitors , Immunity, Cellular/drug effects , Animals , Antibodies, Monoclonal/pharmacokinetics , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD40 Ligand/genetics , CD40 Ligand/immunology , Cells, Cultured , Cricetinae , Cricetulus , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dose-Response Relationship, Immunologic , Fibroblasts/immunology , Humans , Immunoglobulin G/blood , Lymphocyte Activation/drug effects , Male , Mice , Ovalbumin/immunology , Rats , Rats, Inbred Lew , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors , Transfection
13.
Mol Immunol ; 45(9): 2641-50, 2008 May.
Article En | MEDLINE | ID: mdl-18262271

Dendritic cells (DC) play an important role in immune responses and have been studied extensively in human and mouse models. CD40 triggering of DC has a pivotal role in their maturation process, obtaining the unique capacity to induce strong CD4 and CD8 T cell activation. Although rat models are frequently used for the understanding of the underlying mechanism of human diseases, relatively little is known about rat DC. To investigate the effect of CD40 triggering on rat DC, we cloned the rat CD40L gene and generated murine fibroblasts with stable expression (L-rCD40L). DC stimulated by L-rCD40L cells exhibited a strong T cell stimulatory capacity, associated with higher amounts of IFN-gamma as compared to LPS-stimulated DC. Analysis of cytokine production showed that LPS induced both IL-12 and IL-10 production, whereas CD40L induced high amounts of IL-12, but little IL-10 production by rat DC. This implies that the difference found in T cell stimulatory capacity by the stimulated DC is due to the cytokine profile of the DC at the time of T cell activation.


CD40 Ligand/immunology , Dendritic Cells/immunology , Interleukin-10/metabolism , Interleukin-12/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , CD40 Ligand/metabolism , Dendritic Cells/metabolism , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-12/immunology , Lipopolysaccharides/immunology , Male , Mice , Mice, Transgenic , Rats , T-Lymphocytes/metabolism
14.
Eur J Immunol ; 37(10): 2803-11, 2007 Oct.
Article En | MEDLINE | ID: mdl-17899552

Deficiency in complement proteins such as C1q is associated with the development of systemic lupus erythematosus (SLE). Here, we show that the differentiation of dendritic cells (DC) in the presence of C1q (C1qDC) gives rise to CD1a(+)/DC-SIGN(+) cells with high phagocytic capacity and low expression of CD80, CD83 and CD86. Further, when C1qDC were exposed to LPS, a significant reduction in the production of IL-6, TNF-alpha and IL-10 occurred with a limited up-regulation of CD80, CD83 and CD86. In addition, C1qDC were less responsive to activation by CD40L in terms of IL-12p70 secretion and CD86 expression. C1qDC showed an impaired ability to stimulate alloreactive T cells, with a reduced production of IFN-gamma. In conclusion, we have shown that C1q is a potent modulator of DC, resulting in cells characterized by an impaired capacity of cytokine production and an impaired up-regulation of costimulatory molecules, leading to a limited T cell response. Therefore, we hypothesize that, next to a pivotal role in the safe clearance of apoptotic cells, C1q regulates the threshold of DC activation and thereby prevents hyperactivation of the overall immune response.


Complement C1q/physiology , Dendritic Cells/immunology , Cell Adhesion Molecules/biosynthesis , Cell Differentiation/immunology , Cells, Cultured , Complement Pathway, Classical/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Lectins, C-Type/biosynthesis , Lupus Erythematosus, Systemic/immunology , Receptors, Cell Surface/biosynthesis
15.
Eur J Immunol ; 37(6): 1594-9, 2007 Jun.
Article En | MEDLINE | ID: mdl-17474153

We have recently shown that in vitro polarized M-CSF-driven anti-inflammatory macrophages (MPhi2) have the unique capacity to preferentially bind and ingest early apoptotic cells. However, these data are based on in vitro polarized cells and it is unclear whether MPhi2-like cells exist in vivo. Here we used CD163 as a cell surface marker to distinguish MPhi2 from the pro-inflammatory MPhi1. We show that human peritoneal MPhi (pMPhi) freshly isolated from patients on peritoneal dialysis have the phenotypical characteristics of MPhi2, including CD163 surface expression and lack of CD16. Like MPhi2, pMPhi have the capacity for endocytosis and macropinocytosis, are able to preferentially bind and ingest early apoptotic cells, and produce large amounts of IL-10 upon stimulation with LPS. Moreover, upon LPS stimulation both pMPhi and MPhi2 down-regulate CD86, resulting in a reduced capacity to stimulate proliferation of allogeneic T cells and an inhibition of Th1 cytokine release of these T cells. Our data provide the evidence for the first time that in vitro polarized MPhi2 exist in vivo, and human pMPhi resemble the anti-inflammatory MPhi2. We propose that pMPhi have the potential to maintain an anti-inflammatory condition in the peritoneal cavity.


Macrophage Colony-Stimulating Factor/pharmacology , Macrophages, Peritoneal/physiology , Macrophages/physiology , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Cell Adhesion Molecules/analysis , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dextrans/metabolism , Endocytosis/drug effects , Endocytosis/physiology , Flow Cytometry , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , GPI-Linked Proteins , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Isoquinolines/metabolism , Jurkat Cells , Lectins, C-Type/analysis , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Phagocytosis/drug effects , Phagocytosis/physiology , Pinocytosis/drug effects , Pinocytosis/physiology , Receptors, Cell Surface/analysis , Receptors, IgG/analysis
16.
J Am Soc Nephrol ; 17(6): 1724-34, 2006 Jun.
Article En | MEDLINE | ID: mdl-16687629

IgA nephropathy (IgAN) is characterized by glomerular co-deposition of IgA and complement components. Earlier studies showed that IgA activates the alternative pathway of complement, whereas more recent data also indicate activation of the lectin pathway. The lectin pathway can be activated by binding of mannose-binding lectin (MBL) and ficolins to carbohydrate ligands, followed by activation of MBL-associated serine proteases and C4. This study examined the potential role of the lectin pathway in IgAN. Renal biopsies of patients with IgAN (n=60) showed mesangial deposition of IgA1 but not IgA2. Glomerular deposition of MBL was observed in 15 (25%) of 60 cases with IgAN and showed a mesangial pattern. All MBL-positive case, but none of the MBL-negative cases showed glomerular co-deposition of L-ficolin, MBL-associated serine proteases, and C4d. Glomerular deposition of MBL and L-ficolin was associated with more pronounced histologic damage, as evidenced by increased mesangial proliferation, extracapillary proliferation, glomerular sclerosis, and interstitial infiltration, as well as with significantly more proteinuria. Patients who had IgAN with or without glomerular MBL deposition did not show significant differences in serum levels of MBL, L-ficolin, or IgA or in the size distribution of circulating IgA. Furthermore, in vitro experiments showed clear binding of MBL to polymeric but not monomeric patient IgA, without a significant difference between both groups. Together, these findings strongly point to a role for the lectin pathway of complement in glomerular complement activation in IgAN and suggest a contribution for both MBL and L-ficolin in the progression of the disease.


Complement Activation , Glomerulonephritis, IGA/blood , Glomerulonephritis, IGA/pathology , Kidney Diseases/complications , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Lectins/metabolism , Adult , Biopsy , Complement System Proteins , Disease Progression , Female , Humans , Immunoglobulin A/blood , Immunoglobulin A/chemistry , Kidney Diseases/blood , Male , Middle Aged , Ficolins
17.
Blood ; 107(12): 4930-7, 2006 Jun 15.
Article En | MEDLINE | ID: mdl-16497970

Efficient clearance of apoptotic cells seems to be a prerequisite to prevent the development of autoimmunity. Here we identify that macrophage colony-stimulating factor (M-CSF)-driven macrophages (Mø2s) are potent phagocytes that have the unique capacity to preferentially bind and ingest early apoptotic cells. This macrophage subset has intrinsic anti-inflammatory properties, characterized by high interleukin-10 (IL-10) production in the absence of proinflammatory cytokines, such as IL-6 and tumor necrosis factor-alpha (TNF-alpha). Importantly, whereas the IL-6 and TNF-alpha production by granulocyte-macrophage (GM)-CSF-driven macrophages (Mø1s) is inhibited upon uptake of apoptotic cells, the anti-inflammatory status of Mø2 is retained during phagocytosis. Mø2s were shown to use CD14 to tether apoptotic cells, whereas recognition of phosphatidylserine (PS) contributed to uptake of early apoptotic cells. Mø2s showed more potent macropinocytosis compared with dendritic cells (DCs) and Mø1s, and uptake of apoptotic cells was inhibited by a macropinocytosis inhibitor. Our studies suggest that, under steady-state conditions, IL-10-producing Mø2s are prominently involved in the clearance of early apoptotic cells.


Apoptosis/immunology , Autophagy/immunology , Dendritic Cells/immunology , Interleukin-10/immunology , Macrophages/immunology , Phagocytosis/immunology , Apoptosis/drug effects , Autophagy/drug effects , Cells, Cultured , Cytokines/immunology , Cytokines/pharmacology , Dendritic Cells/cytology , Humans , Lipopolysaccharide Receptors/immunology , Macrophages/cytology , Phagocytosis/drug effects , Phosphatidylserines/immunology , Pinocytosis/drug effects , Pinocytosis/immunology
18.
Eur J Immunol ; 34(9): 2589-98, 2004 Sep.
Article En | MEDLINE | ID: mdl-15307191

Deficiency of mannose-binding lectin (MBL), a recognition molecule of the lectin pathway of complement, is associated with increased susceptibility to infections. The high frequency of MBL deficiency suggests that defective MBL-mediated innate immunity can be compensated by alternative defense strategies. To examine this hypothesis, complement activation by MBL-binding ligands was studied. The results show that the prototypic MBL ligand mannan can induce complement activation via both the lectin pathway and the classical pathway. Furthermore, antibody binding to mannan restored complement activation in MBL-deficient serum in a C1q-dependent manner. Cooperation between the classical pathway and the lectin pathway was also observed for complement activation by protein 60 from Listeria monocytogenes. MBL pathway analysis at the levels of C4 and C5b-9 in the presence of classical pathway inhibition revealed a large variation of MBL pathway activity, depending on mbl2 gene polymorphisms. MBL pathway dysfunction in variant allele carriers is associated with reduced MBL ligand binding and a relative increase of low-molecular-mass MBL. These findings indicate that antibody-mediated classical pathway activation can compensate for impaired target opsonization via the MBL pathway in MBL-deficient individuals, and imply that MBL deficiency may become clinically relevant in absence of a concomitant adaptive immune response.


Antibodies/physiology , Mannose-Binding Lectin/deficiency , Bacterial Proteins/physiology , Complement C1q/physiology , Complement C4/metabolism , Complement Pathway, Classical , Humans , Lectins/physiology , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/physiology , Promoter Regions, Genetic
19.
J Immunol ; 172(10): 6482-9, 2004 May 15.
Article En | MEDLINE | ID: mdl-15128841

Sanglifehrin A (SFA) is a recently developed immunosuppressant that belongs to the family of immunophilin-binding ligands. SFA is a cyclophilin A-binding immunosuppressive drug with a novel, but unidentified, mechanism of action. Several reports exist about the effect of SFA on T cells, but its effect on the initiators of the immune response, i.e., dendritic cells (DCs), is relatively unknown. Therefore, we examined the effect of SFA on the differentiation and function of human monocyte-derived DCs. Unlike the well-known cyclophilin A-binding immunosuppressant cyclosporin A, which did not affect DC phenotype, differentiation of DCs in the presence of SFA resulted in CD14-CD1a DCs with normal DC morphology, viability, and a proper capacity to activate allogeneic T cells. However, DCs generated in the presence of SFA demonstrated reduced macropinocytosis and lectin-mediated endocytosis, which was in line with a decreased expression of C-type lectins, including mannose receptor, C1qRP, DC-ASGPR, and especially, DC-SIGN. In contrast, FcalphaRI (CD89) and FcgammaRII (CD32) were increased by SFA. The explicit effect of SFA on the expression of Ag uptake receptors and Ag capture by DCs makes SFA unique among immunophilin-binding immunosuppressive drugs.


Cyclophilins/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Endocytosis/drug effects , Endocytosis/immunology , Fluorescein-5-isothiocyanate/analogs & derivatives , Immunosuppressive Agents/pharmacology , Lactones/pharmacology , Receptors, Antigen/metabolism , Spiro Compounds/pharmacology , Antigens, CD/biosynthesis , Cell Differentiation/immunology , Cells, Cultured , Cyclosporine/metabolism , Cyclosporine/pharmacology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dextrans/antagonists & inhibitors , Dextrans/metabolism , Fluorescein-5-isothiocyanate/metabolism , Humans , Immunophenotyping , Lactones/metabolism , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/biosynthesis , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Mannose/antagonists & inhibitors , Mannose/metabolism , Receptors, Antigen/biosynthesis , Receptors, Fc/biosynthesis , Receptors, IgG/biosynthesis , Serum Albumin/antagonists & inhibitors , Serum Albumin/metabolism , Spiro Compounds/metabolism , T-Lymphocytes/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
...