Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Commun Biol ; 6(1): 207, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36813842

ABSTRACT

CD47 recognized by its macrophage receptor SIRPα serves as a "don't eat-me" signal protecting viable cells from phagocytosis. How this is abrogated by apoptosis-induced changes in the plasma membrane, concomitantly with exposure of phosphatidylserine and calreticulin "eat-me" signals, is not well understood. Using STORM imaging and single-particle tracking, we interrogate how the distribution of these molecules on the cell surface correlates with plasma membrane alteration, SIRPα binding, and cell engulfment by macrophages. Apoptosis induces calreticulin clustering into blebs and CD47 mobility. Modulation of integrin affinity impacts CD47 mobility on the plasma membrane but not the SIRPα binding, whereas CD47/SIRPα interaction is suppressed by cholesterol destabilization. SIRPα no longer recognizes CD47 localized on apoptotic blebs. Overall, the data suggest that disorganization of the lipid bilayer at the plasma membrane, by inducing inaccessibility of CD47 possibly due to a conformational change, is central to the phagocytosis process.


Subject(s)
CD47 Antigen , Calreticulin , Humans , Apoptosis , Calreticulin/metabolism , CD47 Antigen/chemistry , CD47 Antigen/metabolism , Cell Membrane/metabolism
2.
FEBS Open Bio ; 11(10): 2693-2704, 2021 10.
Article in English | MEDLINE | ID: mdl-34328698

ABSTRACT

SREC-II (scavenger receptor expressed by endothelial cells II) is a membrane protein encoded by the SCARF2 gene, with high homology to class F scavenger receptor SR-F1, but no known scavenging function. We produced the extracellular domain of SREC-II in a recombinant form and investigated its capacity to interact with common scavenger receptor ligands, including acetylated low-density lipoprotein (AcLDL) and maleylated or acetylated BSA (MalBSA or AcBSA). Whereas no binding was observed for AcLDL, SREC-II ectodomain interacted strongly with MalBSA and bound with high affinity to AcBSA, a property shared with the SR-F1 ectodomain. SREC-II ectodomain also interacted with two SR-F1-specific ligands, complement C1q and calreticulin, with affinities in the 100 nm range. We proceeded to generate a stable CHO cell line overexpressing full-length SREC-II; binding of MalBSA to these cells was significantly increased compared with nontransfected CHO cells. In contrast, no increase in binding could be detected for C1q and calreticulin. We show for the first time that SREC-II has the capacity to interact with the common scavenger receptor ligand MalBSA. In addition, our data highlight similarities and differences in the ligand binding properties of SREC-II in soluble form and at the cell surface, and show that endogenous protein ligands of the ectodomain of SREC-II, such as C1q and calreticulin, are shared with the corresponding domain of SR-F1.


Subject(s)
Endothelial Cells , Scavenger Receptors, Class F , Animals , Cricetinae , Cricetulus , Endothelial Cells/metabolism , Ligands , Receptors, Scavenger , Scavenger Receptors, Class F/genetics , Scavenger Receptors, Class F/metabolism
3.
Front Immunol ; 11: 544, 2020.
Article in English | MEDLINE | ID: mdl-32296440

ABSTRACT

The scavenger receptor SR-F1 binds to and mediates the internalization of a wide range of ligands, and is involved in several immunological processes. We produced recombinant SR-F1 ectodomain and fragments deleted from the last 2 or 5 C-terminal epidermal growth factor-like modules and investigated their role in the binding of acetylated low density lipoprotein (AcLDL), complement C1q, and calreticulin (CRT). C1q measured affinity was in the 100 nM range and C1q interaction occurs via its collagen-like region. We identified two different binding regions on SR-F1: the N-terminal moiety interacts with C1q and CRT whereas the C-terminal moiety binds AcLDL. The role of SR-F1 N-linked glycans was also tested by mutating each of the three glycosylated asparagines. The three mutants retained binding activities for both AcLDL and C1q. A stable THP-1 cell line overexpressing SR-F1 was generated and C1q was shown to bind more strongly to the surface of SR-F1 overexpressing macrophages, with C1q/SR-F1 colocalization observed in some membrane areas. We also observed a higher level of CRT internalization for THP-1 SR-F1 cells. Increasing SR-F1 negatively modulated the uptake of apoptotic cells. Indeed, THP-1 cells overexpressing SR-F1 displayed a lower phagocytic capacity as compared with mock-transfected cells, which could be partially restored by addition of C1q in the extracellular milieu. Our data shed some light on the role of SR-F1 in efferocytosis, through its capacity to bind C1q and CRT, two proteins involved in this process.


Subject(s)
Apoptosis/immunology , Complement C1q/immunology , Macrophages/immunology , Phagocytosis/immunology , Scavenger Receptors, Class F/immunology , Calreticulin/immunology , Cell Communication/immunology , Complement C1q/metabolism , Humans , Scavenger Receptors, Class F/metabolism , THP-1 Cells
5.
J Exp Med ; 216(11): 2669-2687, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31492810

ABSTRACT

Neutrophils produce high levels of reactive oxygen species (ROS) by NADPH oxidase that are crucial for host defense but can lead to tissue injury when produced in excess. We previously described that proliferating cell nuclear antigen (PCNA), a nuclear scaffolding protein pivotal in DNA synthesis, controls neutrophil survival through its cytosolic association with procaspases. We herein showed that PCNA associated with p47phox, a key subunit of NADPH oxidase, and that this association regulated ROS production. Surface plasmon resonance and crystallography techniques demonstrated that the interdomain-connecting loop of PCNA interacted directly with the phox homology (PX) domain of the p47phox. PCNA inhibition by competing peptides or by T2AA, a small-molecule PCNA inhibitor, decreased NADPH oxidase activation in vitro. Furthermore, T2AA provided a therapeutic benefit in mice during trinitro-benzene-sulfonic acid (TNBS)-induced colitis by decreasing oxidative stress, accelerating mucosal repair, and promoting the resolution of inflammation. Our data suggest that targeting PCNA in inflammatory neutrophils holds promise as a multifaceted antiinflammatory strategy.


Subject(s)
Cytosol/metabolism , NADPH Oxidase 2/metabolism , NADPH Oxidases/metabolism , Neutrophils/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Colitis/chemically induced , Colitis/prevention & control , Enzyme Activation/drug effects , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/genetics , Protein Binding , Reactive Oxygen Species/metabolism , Small Molecule Libraries/pharmacology , Trinitrobenzenesulfonic Acid
6.
Kidney Int ; 96(2): 397-408, 2019 08.
Article in English | MEDLINE | ID: mdl-31142442

ABSTRACT

Granulomatosis with polyangiitis (GPA) is an autoimmune vasculitis associated with anti-neutrophil-cytoplasmic antibodies (ANCA) against proteinase 3 leading to kidney damage. Neutrophils from those patients have increased expression of membrane proteinase 3 during apoptosis. Here we examined whether neutrophils from patients with GPA have dysregulated protein expressions associated with apoptosis. A global proteomic analysis was performed comparing neutrophils from patients with GPA, with healthy individuals under basal conditions and during apoptosis. At disease onset, the cytosolic proteome of neutrophils of patients with GPA before treatment was significantly different from healthy controls, and this dysregulation was more pronounced following ex vivo apoptosis. Proteins involved in cell death/survival were altered in neutrophils of patients with GPA. Several proteins identified were PR3-binding partners involved in the clearance of apoptotic cells, namely calreticulin, annexin-A1 and phospholipid scramblase 1. These proteins form a platform at the membrane of apoptotic neutrophils in patients with GPA but not healthy individuals and this was associated with the clinical presentation of GPA. Thus, our study shows that neutrophils from patients with GPA have an intrinsic dysregulation in proteins involved in apoptotic cell clearance, which could contribute to the unabated inflammation and autoimmunity in GPA. Hence, harnessing these dysregulated pathways could lead to novel biomarkers and targeted therapeutic opportunities to treat kidney disease.


Subject(s)
Annexin A1/metabolism , Apoptosis/immunology , Autoimmunity , Granulomatosis with Polyangiitis/immunology , Neutrophils/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Annexin A1/immunology , Antibodies, Antineutrophil Cytoplasmic/immunology , Biomarkers/metabolism , Calreticulin/immunology , Calreticulin/metabolism , Female , Granulomatosis with Polyangiitis/blood , Granulomatosis with Polyangiitis/diagnosis , Humans , Male , Middle Aged , Myeloblastin/immunology , Myeloblastin/metabolism , Neutrophils/metabolism , Phospholipid Transfer Proteins/immunology , Phospholipid Transfer Proteins/metabolism , Proteomics , Signal Transduction/immunology , Young Adult
7.
Nanomedicine ; 18: 292-302, 2019 06.
Article in English | MEDLINE | ID: mdl-30368000

ABSTRACT

Nanodiamonds are promising nanomedicines for diagnostic and therapeutic applications. As nanodiamonds are mainly administered intravenously, it is critical to understand the humoral immune response upon exposure to nanodiamonds. Here, we report the interactions of pristine, oxidized, and PEG-functionalized nanodiamonds with human complement, an important part of our humoral innate immunity. In particular, we report the nanodiamond binding properties of the recognition protein of the classical complement pathway: C1q, which also takes part in many other physiological and pathological processes. Our results show similar trends in the effects of C1q on the three types of nanodiamonds. Complement activation assays using human serum show that the nanodiamonds trigger slight activities via the alternative pathway and no response via the classical pathway. Nevertheless, surface plasmon resonance shows that C1q binds the nanodiamonds and transmission electron microscopy reveals their agglutination. Studies with macrophages further show that C1q attachment affects their phagocytosis and cytokine response.


Subject(s)
Complement Activation , Complement C1q/metabolism , Immunity, Innate , Nanodiamonds/chemistry , Agglutination , Dynamic Light Scattering , Humans , Macrophages/metabolism , Nanodiamonds/ultrastructure , THP-1 Cells , Thermogravimetry
8.
Front Immunol ; 9: 818, 2018.
Article in English | MEDLINE | ID: mdl-29755460

ABSTRACT

Proteinase 3 (PR3) is the autoantigen in granulomatosis with polyangiitis, an autoimmune necrotizing vasculitis associated with anti-neutrophil cytoplasmic antibodies (ANCAs). Moreover, PR3 is a serine protease whose membrane expression can potentiate inflammatory diseases such as ANCA-associated vasculitis and rheumatoid arthritis. During apoptosis, PR3 is co-externalized with phosphatidylserine (PS) and is known to modulate the clearance of apoptotic cells through a calreticulin (CRT)-dependent mechanism. The complement protein C1q is one mediator of efferocytosis, the clearance of altered self-cells, particularly apoptotic cells. Since PR3 and C1q are both involved in the clearance of apoptotic cells and immune response modulation and share certain common ligands (i.e., CRT and PS), we examined their possible interaction. We demonstrated that C1q binding was increased on apoptotic rat basophilic leukemia (RBL) cells that expressed PR3, and we demonstrated the direct interaction between purified C1q and PR3 molecules as shown by surface plasmon resonance. To better understand the functional consequence of this partnership, we tested C1q-dependent phagocytosis of the RBL cell line expressing PR3 and showed that PR3 impaired C1q enhancement of apoptotic cell uptake. These findings shed new light on the respective roles of C1q and PR3 in the elimination of apoptotic cells and suggest a novel potential axis to explore in autoimmune diseases characterized by a defect in apoptotic cell clearance and in the resolution of inflammation.


Subject(s)
Apoptosis , Complement C1q/immunology , Myeloblastin/immunology , Animals , Cell Line, Tumor , Cell Membrane/immunology , Humans , Inflammation , Leukemia, Basophilic, Acute/immunology , Myeloblastin/genetics , Neutrophils/immunology , Phagocytosis , Protein Binding , Rats
9.
Front Immunol ; 8: 1034, 2017.
Article in English | MEDLINE | ID: mdl-28878781

ABSTRACT

Calreticulin (CRT) is a well-known "eat-me" signal harbored by dying cells participating in their recognition by phagocytes. CRT is also recognized to deeply impact the immune response to altered self-cells. In this study, we focus on the role of the newly exposed CRT following cell death induction. We show that if CRT increases at the outer face of the plasma membrane and is well recognized by C1q even when phosphatidylserine is not yet detected, CRT is also released in the surrounding milieu and is able to interact with phagocytes. We observed that exogenous CRT is endocytosed by THP1 macrophages through macropinocytosis and that internalization is associated with a particular phenotype characterized by an increase of cell spreading and migration, an upregulation of CD14, an increase of interleukin-8 release, and a decrease of early apoptotic cell uptake. Importantly, CRT-induced pro-inflammatory phenotype was confirmed on human monocytes-derived macrophages by the overexpression of CD40 and CD274, and we found that monocyte-derived macrophages exposed to CRT display a peculiar polarization notably associated with a downregulation of the histocompatibility complex of class II molecules hampering its description through the classical M1/M2 dichotomy. Altogether our results highlight the role of soluble CRT with strong possible consequences on the macrophage-mediated immune response to dying cell.

10.
Sci Rep ; 6: 35561, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759041

ABSTRACT

Cytosolic proliferating cell nuclear antigen (PCNA), a scaffolding protein involved in DNA replication, has been described as a key element in survival of mature neutrophil granulocytes, which are non-proliferating cells. Herein, we demonstrated an active export of PCNA involved in cell survival and chemotherapy resistance. Notably, daunorubicin-resistant HL-60 cells (HL-60R) have a prominent cytosolic PCNA localization due to increased nuclear export compared to daunorubicin-sensitive HL-60 cells (HL-60S). By interacting with nicotinamide phosphoribosyltransferase (NAMPT), a protein involved in NAD biosynthesis, PCNA coordinates glycolysis and survival, especially in HL-60R cells. These cells showed a dramatic increase in intracellular NAD+ concentration as well as glycolysis including increased expression and activity of hexokinase 1 and increased lactate production. Furthermore, this functional activity of cytoplasmic PCNA was also demonstrated in patients with acute myeloid leukemia (AML). Our data uncover a novel pathway of nuclear export of PCNA that drives cell survival by increasing metabolism flux.


Subject(s)
Cytosol/metabolism , Leukemia, Myeloid, Acute/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Animals , Cell Survival , DNA Replication , Daunorubicin/therapeutic use , Drug Resistance , Glycolysis , HL-60 Cells , Hexokinase/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Nicotinamide Phosphoribosyltransferase/metabolism , Proliferating Cell Nuclear Antigen/genetics , Protein Binding , Protein Transport
11.
J Biol Chem ; 291(20): 10476-89, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26961880

ABSTRACT

Proteinase 3 (PR3), the autoantigen in granulomatosis with polyangiitis, is expressed at the plasma membrane of resting neutrophils, and this membrane expression increases during both activation and apoptosis. Using surface plasmon resonance and protein-lipid overlay assays, this study demonstrates that PR3 is a phosphatidylserine-binding protein and this interaction is dependent on the hydrophobic patch responsible for membrane anchorage. Molecular simulations suggest that PR3 interacts with phosphatidylserine via a small number of amino acids, which engage in long lasting interactions with the lipid heads. As phosphatidylserine is a major component of microvesicles (MVs), this study also examined the consequences of this interaction on MV production and function. PR3-expressing cells produced significantly fewer MVs during both activation and apoptosis, and this reduction was dependent on the ability of PR3 to associate with the membrane as mutating the hydrophobic patch restored MV production. Functionally, activation-evoked MVs from PR3-expressing cells induced a significantly larger respiratory burst in human neutrophils compared with control MVs. Conversely, MVs generated during apoptosis inhibited the basal respiratory burst in human neutrophils, and those generated from PR3-expressing cells hampered this inhibition. Given that membrane expression of PR3 is increased in patients with granulomatosis with polyangiitis, MVs generated from neutrophils expressing membrane PR3 may potentiate oxidative damage of endothelial cells and promote the systemic inflammation observed in this disease.


Subject(s)
Cell-Derived Microparticles/metabolism , Myeloblastin/metabolism , Phosphatidylserines/metabolism , Animals , Apoptosis , Cell Line , Granulomatosis with Polyangiitis/enzymology , Granulomatosis with Polyangiitis/etiology , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Myeloblastin/chemistry , Neutrophils/metabolism , Phospholipid Transfer Proteins/metabolism , Rats , Respiratory Burst
12.
FEBS Open Bio ; 5: 99-106, 2015.
Article in English | MEDLINE | ID: mdl-25737835

ABSTRACT

Eukaryotic EnguLfment and cell MOtility (ELMO) proteins form an evolutionary conserved family of regulators involved in small GTPase dependent actin remodeling processes that regulates the guanine exchange factor activity of some of the Downstream Of CrK (DOCK) family members. Gathered data strongly suggest that DOCK activation by ELMO and the subsequent signaling result from a subtle balance in the binding of partners to ELMO. Among its putative upward modulators, the Hematopoietic cell kinase (Hck), a member of the Src kinase superfamily, has been identified as a binding partner and a specific tyrosine kinase for ELMO1. Indeed, Hck is implicated in distinct molecular signaling pathways governing phagocytosis, cell adhesion, and migration of hematopoietic cells. Although ELMO1 has been shown to interact with the regulatory Src Homology 3 (SH3) domain of Hck, no direct evidence indicating the mode of interaction between Hck and ELMO1 have been provided in the literature. In the present study, we report convergent pieces of evidence that demonstrate the specific interaction between the SH3 domain of Hck and the polyproline motif of ELMO1. Our results also suggest that the tyrosine-phosphorylation state of ELMO1 tail might act as a putative modulator of Hck kinase activity towards ELMO1 that in turn participates in DOCK180 activation and further triggers subsequent signaling towards actin remodeling.

13.
J Innate Immun ; 6(4): 426-34, 2014.
Article in English | MEDLINE | ID: mdl-24557008

ABSTRACT

C1q has been shown to recognize apoptotic cells, to enhance their uptake and to modulate cytokine release by phagocytes and thus promote immune tolerance. Surface-exposed calreticulin (CRT), known as a C1q receptor, is also considered to be an early eat-me signal that enhances the phagocytosis of apoptotic cells and is capable of eliciting an immunogenic response. However, the molecular mechanisms that trigger these functions are not clear. We hypothesized that CRT and C1q might act together in these processes. We first showed, by means of fluorescence resonance energy transfer (FRET), that CRT interacts with the C1q globular region at the surface of early apoptotic cells. Next, we pointed out that knockdown of CRT on early apoptotic HeLa cells impairs the enhancement effect of C1q on their uptake by THP-1 monocyte-derived macrophages. Furthermore, a deficiency of CRT induces contrasting effects on cytokine release by THP-1 macrophages, increasing interleukin (IL)-6 and monocyte chemotactic protein 1/CCL2 and decreasing IL-8. Remarkably, these effects were greatly reduced when apoptotic cells were opsonized by C1q, which counterbalanced the effect of the CRT deficiency. These results demonstrate that CRT-C1q interaction is involved in the C1q bridging function and they highlight the particular ability of C1q to control the phagocyte inflammatory status, i.e. by integrating the molecular changes that could occur at the surface of dying cells.


Subject(s)
Calreticulin/metabolism , Complement C1q/metabolism , Macrophages/physiology , Apoptosis , Calreticulin/genetics , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Fluorescence Resonance Energy Transfer , Gene Expression Regulation/genetics , HeLa Cells , Humans , Immune Tolerance , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Phagocytosis/genetics , Protein Binding/genetics
14.
J Biol Chem ; 287(51): 42620-33, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23086952

ABSTRACT

C1q, a key component of the classical complement pathway, is a major player in the response to microbial infection and has been shown to detect noxious altered-self substances such as apoptotic cells. In this work, using complementary experimental approaches, we identified the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as a C1q partner when exposed at the surface of human pathogenic bacteria Streptococcus pneumoniae and human apoptotic cells. The membrane-associated GAPDH on HeLa cells bound the globular regions of C1q as demonstrated by pulldown and cell surface co-localization experiments. Pneumococcal strains deficient in surface-exposed GAPDH harbored a decreased level of C1q recognition when compared with the wild-type strains. Both recombinant human and pneumococcal GAPDHs interacted avidly with C1q as measured by surface plasmon resonance experiments (K(D) = 0.34-2.17 nm). In addition, GAPDH-C1q complexes were observed by transmission electron microscopy after cross-linking. The purified pneumococcal GAPDH protein activated C1 in an in vitro assay unlike the human form. Deposition of C1q, C3b, and C4b from human serum at the surface of pneumococcal cells was dependent on the presence of surface-exposed GAPDH. This ability of C1q to sense both human and bacterial GAPDHs sheds new insights on the role of this important defense collagen molecule in modulating the immune response.


Subject(s)
Cell Membrane/enzymology , Complement C1q/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Streptococcus pneumoniae/enzymology , Apoptosis , Cell Membrane Structures/metabolism , Complement Activation , Complement C1q/chemistry , Complement C1q/ultrastructure , Glyceraldehyde-3-Phosphate Dehydrogenases/ultrastructure , HeLa Cells , Humans , Immobilized Proteins/metabolism , Kinetics , Ligands , Mutation/genetics , Plasminogen/metabolism , Protein Binding , Protein Transport , Solubility , Solutions , Surface Plasmon Resonance
15.
J Immunol ; 189(5): 2574-83, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22844112

ABSTRACT

Proteinase 3 (PR3) is the target of anti-neutrophil cytoplasm Abs in granulomatosis with polyangiitis, a form of systemic vasculitis. Upon neutrophil apoptosis, PR3 is coexternalized with phosphatidylserine and impaired macrophage phagocytosis. Calreticulin (CRT), a protein involved in apoptotic cell recognition, was found to be a new PR3 partner coexpressed with PR3 on the neutrophil plasma membrane during apoptosis, but not after degranulation. The association between PR3 and CRT was demonstrated in neutrophils by confocal microscopy and coimmunoprecipitation. Evidence for a direct interaction between PR3 and the globular domain of CRT, but not with its P domain, was provided by surface plasmon resonance spectroscopy. Phagocytosis of apoptotic neutrophils from healthy donors was decreased after blocking lipoprotein receptor-related protein (LRP), a CRT receptor on macrophages. In contrast, neutrophils from patients with granulomatosis with polyangiitis expressing high membrane PR3 levels showed a lower rate of phagocytosis than those from healthy controls not affected by anti-LRP, suggesting that the LRP-CRT pathway was disturbed by PR3-CRT association. Moreover, phagocytosis of apoptotic PR3-expressing cells potentiated proinflammatory cytokine in vitro by human monocyte-derived macrophages and in vivo by resident murine peritoneal macrophages, and diverted the anti-inflammatory response triggered by the phagocytosis of apoptotic cells after LPS challenge in thioglycolate-elicited murine macrophages. Therefore, membrane PR3 expressed on apoptotic neutrophils might amplify inflammation and promote autoimmunity by affecting the anti-inflammatory "reprogramming" of macrophages.


Subject(s)
Apoptosis/immunology , Autoantigens/metabolism , Calreticulin/metabolism , Granulomatosis with Polyangiitis/immunology , Macrophages/immunology , Microscopic Polyangiitis/immunology , Myeloblastin/metabolism , Neutrophils/immunology , Adjuvants, Immunologic/physiology , Animals , Granulomatosis with Polyangiitis/enzymology , Granulomatosis with Polyangiitis/pathology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Macrophages/enzymology , Macrophages/pathology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopic Polyangiitis/enzymology , Microscopic Polyangiitis/pathology , Neutrophils/enzymology , Neutrophils/pathology , Rats
16.
J Mol Biol ; 408(2): 277-90, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21352829

ABSTRACT

Both C1q and calreticulin (CRT) are involved in the recognition of apoptotic cells. CRT was initially characterized as a receptor for the C1q collagen-like fragment (CLF), whereas C1q was shown to bind apoptotic cells through its globular region (GR). Using purified CRT and recombinant CRT domains, we now provide unambiguous experimental evidence that, in addition to its CLF, the C1q GR also binds CRT and that both types of interactions are mediated by the CRT globular domain. Surface plasmon resonance analyses revealed that the C1q CLF and GR domains each bind individually to immobilized CRT and its globular domain with K(D) values of (2.6-8.3) × 10(-7) M. Further evidence that CRT binds to the C1q GR was obtained by electron microscopy. The role of CRT in the recognition of apoptotic HeLa cells by C1q was analyzed. The C1q GR partially colocalized with CRT on the surface of early apoptotic cells, and siRNA (small interfering RNA)-induced CRT deficiency resulted in increased apoptotic cell binding to C1q. The interaction between CRT and phosphatidylserine (PS), a known C1q ligand on apoptotic cells, was also investigated. The polar head of PS was shown to bind to CRT with a 10-fold higher affinity (K(D)=1.5 × 10(-5) M) than that determined for C1q, and, accordingly, the C1q GR-PS interaction was impaired in the presence of CRT. Together, these observations indicate that CRT, C1q, and PS are all closely involved in the uptake of apoptotic cells and strongly suggest a combinatorial role of these three molecules in the recognition step.


Subject(s)
Apoptosis , Calreticulin/metabolism , Cell Communication , Complement C1q/metabolism , Phosphatidylserines/metabolism , Blotting, Western , Calreticulin/antagonists & inhibitors , Calreticulin/genetics , Flow Cytometry , HeLa Cells , Humans , Membrane Proteins/metabolism , Phagocytosis , Protein Binding , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface Plasmon Resonance
17.
PLoS One ; 6(3): e17886, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21423620

ABSTRACT

In the endoplasmic reticulum, calreticulin acts as a chaperone and a Ca(2+)-signalling protein. At the cell surface, it mediates numerous important biological effects. The crystal structure of the human calreticulin globular domain was solved at 1.55 Å resolution. Interactions of the flexible N-terminal extension with the edge of the lectin site are consistently observed, revealing a hitherto unidentified peptide-binding site. A calreticulin molecular zipper, observed in all crystal lattices, could further extend this site by creating a binding cavity lined by hydrophobic residues. These data thus provide a first structural insight into the lectin-independent binding properties of calreticulin and suggest new working hypotheses, including that of a multi-molecular mechanism.


Subject(s)
Calreticulin/chemistry , Calreticulin/metabolism , Peptides/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Lectins/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Surface Properties
18.
ACS Nano ; 5(2): 730-7, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21214219

ABSTRACT

The classical pathway of complement is an essential component of the human innate immune system involved in the defense against pathogens as well as in the clearance of altered self-components. Activation of this pathway is triggered by C1, a multimolecular complex comprising a recognition protein C1q associated with a catalytic subunit C1s-C1r-C1r-C1s. We report here the direct observation of organized binding of C1 components C1q and C1s-C1r-C1r-C1s on carbon nanotubes, an ubiquitous component in nanotechnology research. Electron microscopy imaging showed individual multiwalled carbon nanotubes with protein molecules organized along the length of the sidewalls, often over 1 µm long. Less well-organized protein attachment was also observed on double-walled carbon nanotubes. Protein-solubilized nanotubes continued to attract protein molecules after their surface was fully covered. Despite the C1q binding properties, none of the nanotubes activated the C1 complex. We discuss these results on the adsorption mechanisms of macromolecules on carbon nanotubes and the possibility of using carbon nanotubes for structural studies of macromolecules. Importantly, the observations suggest that carbon nanotubes may interfere with the human immune system when entering the bloodstream. Our results raise caution in the applications of carbon nanotubes in biomedicine but may also open possibilities of novel applications concerning the many biochemical processes involving the versatile C1 macromolecule.


Subject(s)
Complement C1/chemistry , Immunity, Innate , Nanotubes, Carbon/chemistry , Complement C1/immunology , Complement C1/metabolism , Crystallization , Humans , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/immunology , Protein Subunits/metabolism , Surface Properties
19.
Front Immunol ; 2: 92, 2011.
Article in English | MEDLINE | ID: mdl-22566881

ABSTRACT

C1q, the ligand-binding unit of the C1 complex of complement, is a pattern recognition molecule with the unique ability to sense an amazing variety of targets, including a number of altered structures from self, such as apoptotic cells. The three-dimensional structure of its C-terminal globular domain, responsible for its recognition function, has been solved by X-ray crystallography, revealing a tightly packed heterotrimeric assembly with marked differences in the surface patterns of the subunits, and yielding insights into its versatile binding properties. In conjunction with other approaches, this same technique has been used recently to decipher the mechanisms that allow this domain to interact with various non-immune self ligands, including molecules known to provide eat-me signals on apoptotic cells, such as phosphatidylserine and DNA. These investigations provide evidence for a common binding area for these ligands located in subunit C of the C1q globular domain, and suggest that ligand recognition through this area down-regulates C1 activation, hence contributing to the control of the inflammatory reaction. The purpose of this article is to give an overview of these advances which represent a first step toward understanding the recognition mechanisms of C1q and their biological implications.

20.
FEBS J ; 277(23): 4956-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21054788

ABSTRACT

CD91 plays an important role in the scavenging of apoptotic material, possibly through binding to soluble pattern-recognition molecules. In this study, we investigated the interaction of CD91 with mannan-binding lectin (MBL), ficolins and lung surfactant proteins. Both MBL and L-ficolin were found to bind CD91. The MBL-CD91 interaction was time- and concentration-dependent and could be inhibited by known ligands of CD91. MBL-associated serine protease 3 (MASP-3) also inhibited binding between MBL and CD91, suggesting that the site of interaction is located at or near the MASP-MBL interaction site. This was confirmed by using MBL mutants deficient for MASP binding that were unable to interact with CD91. These findings demonstrate that MBL and L-ficolin interact with CD91, strongly suggesting that they have the potential to function as soluble recognition molecules for scavenging microbial and apoptotic material by CD91.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/metabolism , Mannose-Binding Lectin/chemistry , Mannose-Binding Lectin/metabolism , Mannose-Binding Protein-Associated Serine Proteases/chemistry , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Amino Acid Substitution , Binding Sites/genetics , Calcium/metabolism , Humans , In Vitro Techniques , Kinetics , Lectins/chemistry , Lectins/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-1 , Mannose-Binding Lectin/genetics , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface Plasmon Resonance , Ficolins
SELECTION OF CITATIONS
SEARCH DETAIL
...