Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 237
Filter
Add more filters

Publication year range
1.
Cell ; 180(6): 1081-1097.e24, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32142650

ABSTRACT

Understanding molecular mechanisms that dictate B cell diversity is important for targeting B cells as anti-cancer treatment. Through the single-cell dissection of B cell heterogeneity in longitudinal samples of patients with breast cancer before and after neoadjuvant chemotherapy, we revealed that an ICOSL+ B cell subset emerges after chemotherapy. Using three immunocompetent mouse models, we recapitulated the subset switch of human tumor-infiltrating B cells during chemotherapy. By employing B-cell-specific deletion mice, we showed that ICOSL in B cells boosts anti-tumor immunity by enhancing the effector to regulatory T cell ratio. The signature of ICOSL+ B cells is imprinted by complement-CR2 signaling, which is triggered by immunogenic cell death. Moreover, we identified that CD55, a complement inhibitory protein, determines the opposite roles of B cells in chemotherapy. Collectively, we demonstrated a critical role of the B cell subset switch in chemotherapy response, which has implications in designing novel anti-cancer therapies. VIDEO ABSTRACT.


Subject(s)
B-Lymphocytes/immunology , Breast Neoplasms/immunology , Inducible T-Cell Co-Stimulator Ligand/metabolism , Animals , Antineoplastic Agents/metabolism , B-Lymphocytes/metabolism , CD55 Antigens/immunology , CD55 Antigens/metabolism , Cell Line, Tumor , Complement System Proteins/metabolism , Disease Models, Animal , Female , Humans , Inducible T-Cell Co-Stimulator Ligand/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Receptors, Complement 3d/immunology , Receptors, Complement 3d/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology
2.
Trends Genet ; 34(2): 133-141, 2018 02.
Article in English | MEDLINE | ID: mdl-29249333

ABSTRACT

Severe malaria is caused by the Apicomplexan parasite Plasmodium falciparum, and results in significant global morbidity and mortality, particularly among young children and pregnant women. P. falciparum exclusively infects human erythrocytes during clinical illness, and several natural erythrocyte polymorphisms are protective against severe malaria. Since erythrocytes are enucleated and lack DNA, genetic approaches to understand erythrocyte determinants of malaria infection have historically been limited. This review highlights recent advances in the use of hematopoietic stem cells to facilitate genetic screening for malaria host factors. While challenges still exist, this approach holds promise for gaining new insights into host-pathogen interactions in malaria.


Subject(s)
CD55 Antigens/genetics , Disease Resistance/genetics , Host-Pathogen Interactions/immunology , Hyaluronan Receptors/genetics , Malaria, Falciparum/genetics , Plasmodium falciparum/metabolism , CD55 Antigens/immunology , Cell Differentiation , Child , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/parasitology , Female , Gene Expression , Genome-Wide Association Study , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/parasitology , Humans , Hyaluronan Receptors/immunology , Immunity, Innate , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Pregnancy , Primary Cell Culture , Severity of Illness Index
3.
Transgenic Res ; 30(1): 11-21, 2021 02.
Article in English | MEDLINE | ID: mdl-33387103

ABSTRACT

Decay accelerating factor (DAF), a key complement activation control protein, is a 70 kDa membrane bound glycoprotein which controls extent of formation of the C3 and C5 convertases by accelerating their decay. Using clustered regularly-interspaced short palindromic repeats, (CRISPR)/associated protein 9 (Cas9) genome editing we generated a novel DAF deficient (Daf-/-) rat model. The present study describes the renal and extrarenal phenotype of this model and assesses renal response to complement-dependent injury induced by administration of a complement-fixing antibody (anti-Fx1A) against the glomerular epithelial cell (podocyte). Rats generated were healthy, viable and able to reproduce normally. Complete absence of DAF was documented in renal as well as extra-renal tissues at both protein and mRNA level compared to Daf+/+ rats. Renal histology in Daf-/- rats showed no differences regarding glomerular or tubulointerstitial pathology compared to Daf+/+ rats. Moreover, there was no difference in urine protein excretion (ratio of urine albumin to creatinine) or in serum creatinine and urea levels. In Daf-/- rats, proteinuria was significantly increased following binding of anti-Fx1A antibody to podocytes while increased C3b deposition was observed. The DAF knock-out rat model developed validates the role of this complement cascade regulator in immune-mediated podocyte injury. Given the increasing role of dysregulated complement activation in various forms of kidney disease and the fact that the rat is the preferred animal for renal pathophysiology studies, the rat DAF deficient model may serve as a useful tool to study the role of this complement activation regulator in complement-dependent forms of kidney injury.


Subject(s)
Acute Kidney Injury/genetics , CD55 Antigens/genetics , Complement Activation/genetics , Podocytes/metabolism , Acute Kidney Injury/pathology , Albuminuria , Animals , Antibodies, Anti-Idiotypic/pharmacology , CD55 Antigens/deficiency , CD55 Antigens/immunology , CRISPR-Cas Systems/genetics , Complement Activation/immunology , Complement C3-C5 Convertases/genetics , Complement C5/genetics , Gene Knockout Techniques , Heymann Nephritis Antigenic Complex/genetics , Heymann Nephritis Antigenic Complex/immunology , Humans , Podocytes/pathology , Rats
4.
Am J Pathol ; 189(7): 1386-1401, 2019 07.
Article in English | MEDLINE | ID: mdl-31103439

ABSTRACT

Recent studies traced inflammatory bowel disease in some patients to deficiency of CD55 [decay-accelerating factor (DAF)], but the mechanism underlying the linkage remained unclear. Herein, we studied the importance of DAF in enabling processes that program tolerance in the gut and the eye, two immune-privileged sites where immunosuppressive responses are continuously elicited. Unlike oral feeding or ocular injection of ovalbumin in wild-type (WT) mice, which induced dominant immune tolerance, identical treatment of DAF-/- mice or DAF-/- to WT bone marrow chimeras did not. While 10% to 30% of mesenteric and submandibular lymph node CD4+ cells became robust T-regulatory cells (Tregs) in WT forkhead box P3 (Foxp3)-green fluorescent protein mice, few in either site became Tregs with little suppressor activity in DAF-/- Foxp3-green fluorescent protein mice. Phenotyping of CD103+ dendritic cells (DCs) from the ovalbumin-fed DAF-/- mice showed impaired expression of inducer of costimulation (ICOS) ligand, programmed death receptor 1-ligand 1 (PD1-L1), CxxxC chemokine receptor 1 (Cx3CR1), CCR7, and CCR9. Analyses of elicited DAF-/- Foxp3+ Tregs showed reduced expression of interferon regulatory factor 8 (IRF-8)/aldehyde dehydrogenase 1 family member A2 (Aldh1a2) and glycoprotein A repetitions predominant/latency-associated protein associated with Treg transforming growth factor-ß production and presentation, as well as integrin ß6/integrin ß8 associated with Treg and CD103+ DC transforming growth factor-ß release. Thus, DAF is required for the properties of CD103+ DCs and their naïve CD4+ cell partners that together program tolerance.


Subject(s)
Antigens, CD/immunology , Autoimmune Diseases/immunology , CD55 Antigens/immunology , Dendritic Cells/immunology , Immune Tolerance , Integrin alpha Chains/immunology , Animals , Antigens, CD/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , CD55 Antigens/genetics , Dendritic Cells/pathology , Integrin alpha Chains/genetics , Integrin beta Chains/genetics , Integrin beta Chains/immunology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
5.
Cell Microbiol ; 21(10): e13085, 2019 10.
Article in English | MEDLINE | ID: mdl-31290210

ABSTRACT

Staphylococcus aureus is frequently isolated from patients with community-acquired pneumonia and acute respiratory distress syndrome (ARDS). ARDS is associated with staphylococcal phosphatidylinositol-specific phospholipase C (PI-PLC); however, the role of PI-PLC in the pathogenesis and progression of ARDS remains unknown. Here, we showed that recombinant staphylococcal PI-PLC possesses enzyme activity that causes shedding of glycosylphosphatidylinositol-anchored CD55 and CD59 from human umbilical vein endothelial cell surfaces and triggers cell lysis via complement activity. Intranasal infection with PI-PLC-positive S. aureus resulted in greater neutrophil infiltration and increased pulmonary oedema compared with a plc-isogenic mutant. Although indistinguishable proinflammatory genes were induced, the wild-type strain activated higher levels of C5a in lung tissue accompanied by elevated albumin instillation and increased lactate dehydrogenase release in bronchoalveolar lavage fluid compared with the plc- mutant. Following treatment with cobra venom factor to deplete complement, the wild-type strain with PI-PLC showed a reduced ability to trigger pulmonary permeability and tissue damage. PI-PLC-positive S. aureus induced the formation of membrane attack complex, mainly on type II pneumocytes, and reduced the level of CD55/CD59, indicating the importance of complement regulation in pulmonary injury. In conclusion, S. aureus PI-PLC sensitised tissue to complement activation leading to more severe tissue damage, increased pulmonary oedema, and ARDS progression.


Subject(s)
Bacterial Proteins/metabolism , Complement System Proteins/metabolism , Phosphoinositide Phospholipase C/metabolism , Pulmonary Edema/immunology , Pulmonary Edema/microbiology , Respiratory Distress Syndrome/microbiology , Staphylococcal Infections/immunology , Staphylococcus aureus/enzymology , Alveolar Epithelial Cells/enzymology , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/microbiology , Animals , Bacterial Proteins/genetics , CD55 Antigens/immunology , CD59 Antigens/immunology , Cytokines/metabolism , Glycosylphosphatidylinositols/immunology , Glycosylphosphatidylinositols/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Phosphoinositide Phospholipase C/genetics , Pulmonary Edema/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/metabolism , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
6.
Clin Exp Immunol ; 197(3): 376-386, 2019 09.
Article in English | MEDLINE | ID: mdl-31091357

ABSTRACT

Anti-phospholipid syndrome (APS) is characterized by recurrent pathological pregnancy, arterial or venous thrombosis in the presence of anti-phospholipid antibody (aPL). Complement activation is recognized as an intermediate link leading to placental thrombosis and placental inflammation in APS model mice. Decay accelerating factor (DAF, CD55), MAC-inhibitory protein (MAC-IP, CD59) and membrane co-factor protein (MCP, CD46) are important complement inhibitory proteins (CIPs) highly expressed in normal placenta to curb excessive complement activation and its mediated injuries. Anti-ß2 glycoprotein I (anti-ß2GPI) antibody is an important aPL. We found that placental DAF and CD46 decreased in ß2GPI passively immunized APS model mice, accompanied by C3 deposition, neutrophil infiltration and increased proinflammatory cytokine levels detected in its placenta. Progesterone supplement can up-regulate DAF but not CD46 expression, curb C3 activation and decrease proinflammatory cytokines levels to reduce fetal loss frequency. Progesterone receptor antagonist (mifepristone) or knock-down DAF with specific siRNA, above the protective effects of progesterone, were significantly weakened. Another sex hormone, oestrogen, has no significant effect on placental DAF and C3 contents and fetal loss frequency in the APS mice model. This may be an important mechanism by which progesterone induces maternal-fetal immune tolerance. At the same time, it may provide evidence for the use of progesterone in APS abortion patients.


Subject(s)
Antiphospholipid Syndrome/immunology , CD55 Antigens/immunology , Placenta/immunology , Placenta/injuries , Pregnancy Complications/immunology , Progesterone/pharmacology , Up-Regulation/drug effects , Animals , Antiphospholipid Syndrome/pathology , Female , Mice , Mice, Inbred BALB C , Placenta/pathology , Pregnancy , Pregnancy Complications/drug therapy , Pregnancy Complications/pathology , Up-Regulation/immunology
7.
Anal Biochem ; 570: 51-55, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30771337

ABSTRACT

Scintillation proximity assay (SPA) is a type of radioimmunoassay (RIA). We apply ultrasound enhancement to the general SPA. All assay procedures, including the antibody coating and radiolabeled antigen binding are achieved by simply mixing then standing for 5 min in an ultrasound chamber. No additional incubation time is required. To further demonstrate the capability of the UE-SPA, a quantitative measurement of CD55 in various grades of colon tumors was assessed on human tissue slides. The results showed a significant correlation between CD55 expression and tumorigenesis. In conclusion, we confirmed that UE-SPA is a reliable, rapid and alternative to RIA.


Subject(s)
CD55 Antigens/analysis , Radioimmunoassay/methods , Antibodies, Monoclonal/immunology , CD55 Antigens/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Sonication
8.
Xenotransplantation ; 26(4): e12516, 2019 07.
Article in English | MEDLINE | ID: mdl-30989742

ABSTRACT

Xenotransplantation research has made considerable progress in recent years, largely through the increasing availability of pigs with multiple genetic modifications. We suggest that a pig with nine genetic modifications (ie, currently available) will provide organs (initially kidneys and hearts) that would function for a clinically valuable period of time, for example, >12 months, after transplantation into patients with end-stage organ failure. The national regulatory authorities, however, will likely require evidence, based on in vitro and/or in vivo experimental data, to justify the inclusion of each individual genetic modification in the pig. We provide data both from our own experience and that of others on the advantages of pigs in which (a) all three known carbohydrate xenoantigens have been deleted (triple-knockout pigs), (b) two human complement-regulatory proteins (CD46, CD55) and two human coagulation-regulatory proteins (thrombomodulin, endothelial cell protein C receptor) are expressed, (c) the anti-apoptotic and "anti-inflammatory" molecule, human hemeoxygenase-1 is expressed, and (d) human CD47 is expressed to suppress elements of the macrophage and T-cell responses. Although many alternative genetic modifications could be made to an organ-source pig, we suggest that the genetic manipulations we identify above will all contribute to the success of the initial clinical pig kidney or heart transplants, and that the beneficial contribution of each individual manipulation is supported by considerable experimental evidence.


Subject(s)
Animals, Genetically Modified/genetics , Graft Rejection/prevention & control , Swine/genetics , Transplantation, Heterologous , Animals , Animals, Genetically Modified/immunology , CD47 Antigen/genetics , CD47 Antigen/immunology , CD55 Antigens/genetics , CD55 Antigens/immunology , Endothelial Protein C Receptor/genetics , Endothelial Protein C Receptor/immunology , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knock-In Techniques , Gene Knockout Techniques , Graft Rejection/immunology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , Humans , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/immunology , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/deficiency , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/immunology , Swine/immunology , Thrombomodulin/genetics , Thrombomodulin/immunology
9.
J Immunol ; 198(12): 4837-4845, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28500075

ABSTRACT

Deficiency in the membrane-bound complement regulators CD55 and CD59 exacerbates renal ischemia-reperfusion injury (IRI) in mouse models, but the effect of increasing CD55 and CD59 activity has not been examined. In this study, we investigated the impact of overexpression of human (h) CD55 ± hCD59 or treatment with soluble rhCD55 in a mouse model of renal IRI. Unilaterally nephrectomised mice were subjected to 18 (mild IRI) or 22 min (moderate IRI) warm renal ischemia, and analyzed 24 h after reperfusion for renal function (serum creatinine and urea), complement deposition (C3b/c and C9), and infiltration of neutrophils and macrophages. Transgenic mice expressing hCD55 alone were protected against mild renal IRI, with reduced creatinine and urea levels compared with wild type littermates. However, the renal function of the hCD55 mice was not preserved in the moderate IRI model, despite a reduction in C3b/c and C9 deposition and innate cell infiltration. Mice expressing both hCD55 and hCD59, on the other hand, were protected in the moderate IRI model, with significant reductions in all parameters measured. Wild type mice treated with rhCD55 immediately after reperfusion were also protected in the moderate IRI model. Thus, manipulation of CD55 activity to increase inhibition of the C3 and C5 convertases is protective against renal IRI, and the additional expression of hCD59, which regulates the terminal complement pathway, provides further protection. Therefore, anti-complement therapy using complement regulatory proteins may provide a potential clinical option for preventing tissue and organ damage in renal IRI.


Subject(s)
CD55 Antigens/genetics , CD55 Antigens/therapeutic use , CD59 Antigens/genetics , Kidney Diseases/therapy , Reperfusion Injury/therapy , Animals , CD55 Antigens/immunology , Complement Activation , Creatinine/blood , Humans , Kidney Diseases/immunology , Kidney Diseases/physiopathology , Macrophages/immunology , Mice , Mice, Transgenic , Neutrophils/immunology , Reperfusion Injury/immunology , Reperfusion Injury/physiopathology , Urea/blood
10.
J Immunol ; 197(4): 1127-36, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27357152

ABSTRACT

CD55/DAF, one of the regulators of complement activation, is known to limit excess complement activation on the host cell surface by accelerating the decay of C3 convertase. We reported previously that hepatitis C virus (HCV) infection or virus core protein expression upregulates CD55 expression. CD55 associates with HCV particles, potentially protecting HCV from lysis in circulation. An increase in CD55 on the surface of HCV-infected cells may inhibit complement-mediated cell killing. In this study, we show that Abs against cancer cell surface proteins induce complement-dependent cytolysis or Ab-dependent cell-mediated cytotoxicity of immortalized human hepatocytes in the presence of CD55-blocking Ab. CD55 has a secreted isoform (sCD55) that is generated by alternative splicing. We observed that sCD55 is induced in HCV-infected or HCV replicon-harboring cells, as well as in liver biopsy samples from chronically HCV-infected patients. Conditioned medium from HCV-infected hepatoma cells (Huh7.5 cells) or immortalized human hepatocytes inhibited C3 convertase activity and complement-dependent cytolysis of sheep blood erythrocytes. Chronically HCV-infected patient sera inhibited C3 convertase activity, further implicating HCV-specific impairment of complement function in infected humans. CD55-blocking Ab inhibited erythrocyte lysis by conditioned medium, suggesting that CD55/sCD55 impairs convertase activity. Together, our data show that HCV infection induces sCD55 expression in HCV-infected cell culture-conditioned medium and inhibits C3 convertase activity. This may have implications for modulating complement-mediated immune function in the microenvironment and on HCV-harboring cells.


Subject(s)
CD55 Antigens/metabolism , Complement Activation/physiology , Cytotoxicity, Immunologic/immunology , Hepatitis C/immunology , CD55 Antigens/immunology , Complement C3-C5 Convertases/metabolism , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hepacivirus , Hepatocytes/immunology , Hepatocytes/virology , Humans , Polymerase Chain Reaction , Protein Isoforms
11.
PLoS Pathog ; 9(5): e1003351, 2013 May.
Article in English | MEDLINE | ID: mdl-23675299

ABSTRACT

Nucleotide-binding oligomerization domain (NOD) 2 is a cytosolic protein that plays a defensive role in bacterial infection by sensing peptidoglycans. C5a, which has harmful effects in sepsis, interacts with innate proteins. However, whether NOD2 regulates C5a generation during sepsis remains to be determined. To address this issue, cecal ligation & puncture (CLP)-induced sepsis was compared in wild type and Nod2-/- mice. Nod2-/- mice showed lower levels of C5a, IL-10, and IL-1ß in serum and peritoneum, but higher survival rate during CLP-induced sepsis compared to wild type mice. Injection of recombinant C5a decreased survival rates of Nod2-/- mice rate during sepsis, whereas it did not alter those in wild type mice. These findings suggest a novel provocative role for NOD2 in sepsis, in contrast to its protective role during bacterial infection. Furthermore, we found that NOD2-mediated IL-10 production by neutrophils enhanced C5a generation by suppressing CD55 expression on neutrophils in IL-1ß-dependent and/or IL-1ß-independent manners, thereby aggravating CLP-induced sepsis. SB203580, a receptor-interacting protein 2 (RIP2) inhibitor downstream of NOD2, reduced C5a generation by enhancing CD55 expression on neutrophils, resulting in attenuation of polymicrobial sepsis. Therefore, we propose a novel NOD2-mediated complement cascade regulatory pathway in sepsis, which may be a useful therapeutic target.


Subject(s)
CD55 Antigens/immunology , Complement C5a/immunology , Neutrophils/immunology , Nod2 Signaling Adaptor Protein/immunology , Sepsis/immunology , Animals , Blotting, Western , CD55 Antigens/metabolism , Complement Activation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukin-10/biosynthesis , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Real-Time Polymerase Chain Reaction , Sepsis/metabolism
12.
Mult Scler ; 21(13): 1644-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25698168

ABSTRACT

BACKGROUND: It is unclear why AQP4-IgG primarily causes central nervous system lesions by activating complement, but generally spares peripheral AQP4-expressing organs. OBJECTIVES: To determine whether peripheral AQP4-expressing cells are protected from complement-mediated damage by expressing complement regulators. METHODS: Human tissue and cultured human cells were immunostained for aquaporin-4 (AQP4), CD46, CD55 and CD59. We also determined the vulnerability to AQP4-IgG and complement-mediated damage of astrocytes cultured alone or co-cultured with endothelial cells. RESULTS: In normal brain, astrocyte end-feet express AQP4, but are devoid of CD46, CD55 and CD59. Immunoreactivity for CD46, CD55 and CD59 is not increased in or around neuromyelitis optica lesions. In kidney AQP4 is co-expressed with CD46 and CD55, in stomach AQP4 is co-expressed with CD46 and in skeletal muscle AQP4 is co-expressed with CD46. Astrocytes cultured alone co-express AQP4 and CD59 but, in astrocyte-endothelial co-cultures, AQP4 is found in cell processes devoid of CD59. Astrocytes co-cultured with endothelial cells are more vulnerable to AQP4-IgG and complement-mediated lysis than astrocytes cultured alone. CONCLUSIONS: Complement regulators protect peripheral organs, but not the central nervous system, from AQP4-IgG and complement-mediated damage. Our findings may explain why neuromyelitis optica primarily damages the central nervous system, but spares peripheral organs.


Subject(s)
Aquaporin 4/immunology , Astrocytes/metabolism , Brain/metabolism , Complement System Proteins/immunology , Neuromyelitis Optica/immunology , Aquaporin 4/metabolism , Astrocytes/immunology , Brain/immunology , CD55 Antigens/immunology , CD55 Antigens/metabolism , CD59 Antigens/immunology , CD59 Antigens/metabolism , Cells, Cultured , Coculture Techniques , Complement Pathway, Classical/immunology , Complement System Proteins/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gastric Mucosa/metabolism , Humans , Immunoglobulin G/immunology , Kidney/metabolism , Membrane Cofactor Protein/immunology , Membrane Cofactor Protein/metabolism , Muscle, Skeletal/metabolism , Neuromyelitis Optica/metabolism
13.
Xenotransplantation ; 22(4): 310-6, 2015.
Article in English | MEDLINE | ID: mdl-26174749

ABSTRACT

We describe the incidence of early graft failure (EGF, defined as loss of function from any cause within 3 days after transplant) in a large cohort of GalTKO pig organs transplanted into baboons in three centers, and the effect of additional expression of a human complement pathway-regulatory protein, CD46 or CD55 (GalTKO.hCPRP). Baboon recipients of life-supporting GalTKO kidney (n = 7) or heterotopic heart (n = 14) grafts received either no immunosuppression (n = 4), or one of several partial or full immunosuppressive regimens (n = 17). Fourteen additional baboons received a GalTKO.hCPRP kidney (n = 5) or heart (n = 9) and similar treatment regimens. Immunologic, pathologic, and coagulation parameters were measured at frequent intervals. EGF of GalTKO organs occurred in 9/21 baboons (43%). hCPRP expression reduced the GalTKO EGF incidence to 7% (1/14; P < 0.01 vs. GalTKO alone). At 30 mins, complement deposits were more intense in organs in which EGF developed (P < 0.005). The intensity of peri-transplant platelet activation (as ß-thromboglobulin release) correlated with EGF, as did the cumulative coagulation score (P < 0.01). We conclude that (i) the transgenic expression of a hCPRP on the vascular endothelium of a GalTKO pig reduces the incidence of EGF and reduces complement deposition, (ii) complement deposition and platelet activation correlate with early GalTKO organ failure, and (iii) the expression of a hCPRP reduces EGF but does not prevent systemic coagulation activation. Additional strategies will be required to control coagulation activation.


Subject(s)
CD55 Antigens/immunology , Galactosyltransferases/deficiency , Graft Rejection/prevention & control , Membrane Cofactor Protein/immunology , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified , CD55 Antigens/genetics , Complement Activation , Disaccharides/immunology , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knockout Techniques , Graft Rejection/immunology , Graft Survival/immunology , Heart Transplantation/adverse effects , Heart Transplantation/methods , Humans , Kidney Transplantation/adverse effects , Kidney Transplantation/methods , Membrane Cofactor Protein/genetics , Papio , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Swine , Transplantation, Heterologous/adverse effects
14.
J Immunol ; 191(12): 5895-903, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24198281

ABSTRACT

Unlike other helper T cells, the costimulatory ligands responsible for T regulatory type 1 (Tr1) cell differentiation remain undefined. Understanding the molecular interactions driving peripheral Tr1 differentiation is important because Tr1s potently regulate immune responses by IL-10 production. In this study, we show that costimulation of human naive CD4(+) cells through CD97/CD55 interaction drives Tr1 activation, expansion, and function. T cell activation and expansion was equipotent with CD55 or CD28 costimulation; however, CD55 costimulation resulted in two IL-10-secreting populations. Most IL-10 was secreted by the minor Tr1 population (IL-10(high)IFN-γ(-)IL-4(-), <5% cells) that expresses Tr1 markers CD49b, LAG-3, and CD226. This Tr1 phenotype was not restimulated by CD28. However, on CD55 restimulation, Tr1s proliferated and maintained their differentiated IL-10(high) phenotype. The Tr1s significantly suppressed effector T cell function in an IL-10-dependent manner. The remaining (>95%) cells adopted a Th1-like IFN-γ(+) phenotype. However, in contrast to CD28-derived Th1s, CD55-derived Th1s demonstrated increased plasticity with the ability to coexpress IL-10 when restimulated through CD55 or CD28. These data identify CD55 as a novel costimulator of human Tr1s and support a role for alternative costimulatory pathways in determining the fate of the growing number of T helper populations. This study demonstrates that CD55 acts as a potent costimulator and activator of human naive CD4(+) cells, resulting in the differentiation of a discrete Tr1 population that inhibits T cell function in an IL-10-dependent manner and maintains the Tr1 phenotype upon restimulation.


Subject(s)
CD55 Antigens/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/analysis , CD28 Antigens/immunology , Cell Division , Cells, Cultured , Humans , Immunophenotyping , Interferon-gamma/analysis , Interleukin-10/metabolism , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lymphocyte Activation , Lymphopoiesis , Receptors, G-Protein-Coupled , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/chemistry , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/chemistry , Th1 Cells/immunology , Th1 Cells/metabolism
15.
Am J Physiol Gastrointest Liver Physiol ; 306(12): G1056-64, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24763553

ABSTRACT

Engineering cancer cells to express heterologous antigen α-gal and induce the destruction of tumor cells depending on the complement cascade may be a promising strategy of tumor therapy. However, the feasibility and effect of using α-gal to induce colorectal adenocarcinoma cell line cytolysis is not yet known. In this study, we evaluated α-gal expression's ability to sensitize human colorectal adenocarcinoma cell lines to complement attack in cell lines LoVo, SW620, and Ls-174T. Nearly all α-gal-expressing LoVo and SW620 cells were killed by normal human serum (NHS), but α-gal-expressing Ls-174T cells showed no significant lysis. We analyzed the expression levels of membrane-bound complement regulatory proteins (mCRPs) on the three cell lines, and their protective role in α-gal-mediated activation of the complement. LoVo showed no expression of any of the three proteins. CD59 was strongly expressed by SW620 and Ls-174T. CD46 and CD55 varied between the two cell lines. CD46 on SW620 was only half the intensity of CD46 on Ls-174T. Ls-174T showed a notable expression of CD55, while expression of CD55 on SW620 was not detected. The sensitivity of Ls-174T expressing α-gal to NHS greatly increased following the downregulation of CD46 and CD55 with short hairpin RNA (shRNA). However, there is no increase in cell killing when CD59 expression was diminished. Our findings suggest that the use of α-gal as antigen to induce tumor cell killing may be a potential therapeutic strategy in colon cancer and that CD55 plays a primary role in conferring resistance to lysis.


Subject(s)
CD55 Antigens/metabolism , Colonic Neoplasms/metabolism , Complement System Proteins/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , CD55 Antigens/immunology , Colonic Neoplasms/immunology , Complement Activation/immunology , Complement System Proteins/immunology , Down-Regulation/immunology , Down-Regulation/physiology , Humans , Tumor Cells, Cultured
16.
Blood ; 120(17): 3436-43, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22966167

ABSTRACT

Despite the potent immunosuppressive activity that mesenchymal stem cells (MSCs) display in vitro, recent clinical trial results are disappointing, suggesting that MSC viability and/or function are greatly reduced after infusion. In this report, we demonstrated that human MSCs activated complement of the innate immunity after their contact with serum. Although all 3 known intrinsic cell-surface complement regulators were present on MSCs, activated complement overwhelmed the protection of these regulators and resulted in MSCs cytotoxicity and dysfunction. In addition, autologous MSCs suffered less cellular injury than allogeneic MSCs after contacting serum. All 3 complement activation pathways were involved in generating the membrane attack complex to directly injure MSCs. Supplementing an exogenous complement inhibitor, or up-regulating MSC expression levels of CD55, one of the cell-surface complement regulators, helped to reduce the serum-induced MSC cytotoxicity. Finally, adoptively transferred MSCs in complement deficient mice or complement-depleted mice showed reduced cellular injury in vivo compared with those in wild type mice. These results indicate that complement is integrally involved in recognizing and injuring MSCs after their infusion, suggesting that autologous MSCs may have ad-vantages over allogeneic MSCs, and that inhibiting complement activation could be a novel strategy to improve existing MSC-based therapies.


Subject(s)
Complement Activation/drug effects , Complement Inactivating Agents/pharmacology , Complement System Proteins/immunology , Immunoglobulin G/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Adoptive Transfer , Animals , CD55 Antigens/genetics , CD55 Antigens/immunology , Complement Activation/immunology , Complement Inactivating Agents/therapeutic use , Humans , Immunity, Innate/drug effects , Immunoglobulin G/therapeutic use , Immunomodulation , Mesenchymal Stem Cells/pathology , Mice , Primary Cell Culture , Serum , Transplantation, Autologous , Transplantation, Homologous , Up-Regulation
17.
Blood ; 118(4): 1008-14, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21652682

ABSTRACT

Recent studies have indicated a role of complement in regulating T-cell immunity but the mechanism of action of complement in this process remains to be clarified. Here we studied mice deficient in decay-accelerating factor (DAF), a key membrane complement regulator whose deficiency led to increased complement-dependent T-cell immune responses in vivo. By crossing OT-II and OT-I T-cell receptor transgenic mice with DAF-knockout mice, we found that lack of DAF on T cells did not affect their responses to antigen stimulation. Similarly, lack of DAF on antigen-presenting cells (APCs) of naive mice did not alter their T-cell stimulating activity. In contrast, APCs from DAF-knockout mice treated with inflammatory stimuli were found to be more potent T-cell stimulators than cells from similarly treated wild-type mice. Acquisition of higher T-cell stimulating activity by APCs in challenged DAF-knockout mice required C3 and C5aR and was correlated with decreased surface PD-L1 and/or increased CD40 expression. These findings implied that DAF suppressed T-cell immunity as a complement regulator in the context of inflammation but did not play an intrinsic role on T cells or APCs. Collectively, our data suggest a systemic and indirect role of complement in T-cell immunity.


Subject(s)
Antigen-Presenting Cells/immunology , CD55 Antigens/immunology , Complement Activation/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/metabolism , Blotting, Western , CD55 Antigens/metabolism , Cell Separation , Flow Cytometry , Inflammation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/metabolism
18.
J Infect Dis ; 206(4): 495-503, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22693232

ABSTRACT

Infection due to 2009 pandemic H1N1 influenza A virus (A[H1N1]pdm09) is commonly manifested as mild infection but occasionally as severe pneumonia. We hypothesized that host genetic variations may contribute to disease severity. An initially small-scale genome-wide association study guided the selection of CD55 single-nucleotide polymorphisms in 425 Chinese patients with severe (n = 177) or mild (n = 248) disease. Carriers of rs2564978 genotype T/T were significantly associated with severe infection (odds ratio, 1.75; P = .011) under a recessive model, after adjustment for clinical confounders. An allele-specific effect on CD55 expression was revealed and ascribed to a promoter indel variation, which was in complete linkage disequilibrium with rs2564978. The promoter variant with deletion exhibited significantly lower transcriptional activity. We further demonstrated that CD55 can protect respiratory epithelial cells from complement attack. Additionally, A(H1N1)pdm09 infection promoted CD55 expression. In conclusion, CD55 polymorphisms are associated with severe A(H1N1)pdm09 infection. CD55 may exert a substantial impact on the disease severity of A(H1N1)pdm09 infection.


Subject(s)
CD55 Antigens/genetics , Genetic Predisposition to Disease , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/genetics , Influenza, Human/pathology , Polymorphism, Single Nucleotide , Adolescent , Adult , Aged , Aged, 80 and over , CD55 Antigens/immunology , China , Female , Gene Frequency , Humans , INDEL Mutation , Influenza, Human/immunology , Influenza, Human/virology , Male , Middle Aged , Promoter Regions, Genetic , Severity of Illness Index , Young Adult
19.
Eur J Immunol ; 41(8): 2436-46, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21674476

ABSTRACT

We have reported that during complement-mediated cytolysis of B cells promoted by the CD20 mAbs rituximab or ofatumumab (OFA), long, thin structures that we call streamers (≥ 3 cell diameters) are rapidly generated and grow out from the cell surface. Streamers appear before cells are killed and contain opsonizing mAbs and membrane lipids. By exploiting the differential Ca(2+) requirements of discrete steps in the complement cascade, we determined that mAb-opsonized cells first tagged with C3b using C5-depleted serum are killed on addition of serum and EDTA, but the cells do not produce streamers. Also, cells first opsonized with OFA are lysed in serum containing Mg-EGTA by the alternative complement pathway but streamers are not produced. These findings indicate that Ca(2+) influx is necessary for streamer formation. Other mAbs that promote complement-mediated cytolysis also induce streamers on target cells. Streamer-like structures called nanotubes have been reported in several cellular systems, and are thought to promote intercellular communication/signaling. We tested whether this signaling could influence the susceptibility of neighboring cells contacted by streamers to complement attack and found that complement-mediated cytolysis of OFA-opsonized cells increases the resistance of unopsonized indicator cell populations to subsequent lysis when these cells are exposed to OFA and complement.


Subject(s)
Antibodies, Monoclonal/immunology , Calcium/immunology , Complement Membrane Attack Complex/immunology , Cytotoxicity, Immunologic/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived/immunology , Antigens, CD20/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD55 Antigens/immunology , CD55 Antigens/metabolism , Calcium/antagonists & inhibitors , Calcium/metabolism , Cell Line , Cell Line, Tumor , Chelating Agents/pharmacology , Complement Activation/immunology , Complement C3b/immunology , Complement C3b/metabolism , Complement C5/immunology , Complement C5/metabolism , Complement Membrane Attack Complex/metabolism , Complement Pathway, Alternative/immunology , Cytotoxicity, Immunologic/drug effects , Egtazic Acid/pharmacology , Flow Cytometry , Humans , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Melitten/pharmacology , Microscopy, Fluorescence , Phosphatidylserines/immunology , Phosphatidylserines/metabolism , Rituximab , Serum/immunology , Serum/metabolism
20.
J Immunol ; 185(10): 6128-37, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20956343

ABSTRACT

Vaccinia virus encodes a structural and functional homolog of human complement regulators named vaccinia virus complement control protein (VCP). This four-complement control protein domain containing secretory protein is known to inhibit complement activation by supporting the factor I-mediated inactivation of complement proteins, proteolytically cleaved form of C3 (C3b) and proteolytically cleaved form of C4 (C4b) (termed cofactor activity), and by accelerating the irreversible decay of the classical and to a limited extent of the alternative pathway C3 convertases (termed decay-accelerating activity [DAA]). In this study, we have mapped the VCP domains important for its cofactor activity and DAA by swapping its individual domains with those of human decay-accelerating factor (CD55) and membrane cofactor protein (MCP; CD46). Our data indicate the following: 1) swapping of VCP domain 2 or 3, but not 1, with homologous domains of decay-accelerating factor results in loss in its C3b and C4b cofactor activities; 2) swapping of VCP domain 1, but not 2, 3, or 4 with corresponding domains of MCP results in abrogation in its classical pathway DAA; and 3) swapping of VCP domain 1, 2, or 3, but not 4, with homologous MCP domains have marked effect on its alternative pathway DAA. These functional data together with binding studies with C3b and C4b suggest that in VCP, domains 2 and 3 provide binding surface for factor I interaction, whereas domain 1 mediates dissociation of C2a and Bb from the classical and alternative pathway C3 convertases, respectively.


Subject(s)
Complement System Proteins/metabolism , Vaccinia virus/chemistry , Vaccinia virus/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , CD55 Antigens/chemistry , CD55 Antigens/immunology , CD55 Antigens/metabolism , Complement System Proteins/chemistry , Complement System Proteins/immunology , Electrophoresis, Polyacrylamide Gel , Humans , Protein Structure, Tertiary , Surface Plasmon Resonance , Vaccinia virus/immunology , Viral Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL