1
|
Saxena R, Gottlin EB, Campa MJ, He YW, Patz EF. Complement regulators as novel targets for anti-cancer therapy: A comprehensive review. Semin Immunol 2025; 77:101931. [PMID: 39826189 DOI: 10.1016/j.smim.2025.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/04/2025] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Cancer remains a formidable global health challenge requiring the continued exploration of innovative therapeutic approaches. While traditional treatment strategies including surgery, chemotherapy, and radiation therapy have had some success, primarily in early-stage disease, the quest for more targeted, personalized, safer, and effective therapies remains an ongoing pursuit. Over the past decade, significant advances in the field of tumor immunology have dramatically shifted a focus towards immunotherapy, although the ability to harness and coopt the immune system to treat cancer is still just beginning to be realized. One important area that has yet to be fully explored is the complement system, an integral part of innate immunity that has gathered attention recently as a source of potential targets for anti-cancer therapy. The complement system has a complex and context dependent role in cancer biology in that it not only contributes to immune surveillance but also may promote tumor progression. Complement regulators, including CD46, CD55, CD59, and complement factor H, exercise defined control over complement activation, and have also been acknowledged for their role in the tumor microenvironment. This review explores the intricate role of complement regulators in cancer development and progression, examining their potential as therapeutic targets, current strategies, challenges, and the evolving landscape of clinical research.
Collapse
Affiliation(s)
- Ruchi Saxena
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Elizabeth B Gottlin
- Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael J Campa
- Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - You-Wen He
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Edward F Patz
- Department of Radiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
2
|
Bharti R, Dey G, Lin F, Lathia J, Reizes O. CD55 in cancer: Complementing functions in a non-canonical manner. Cancer Lett 2022; 551:215935. [PMID: 36216147 PMCID: PMC11019835 DOI: 10.1016/j.canlet.2022.215935] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/02/2022]
Abstract
CD55, or decay accelerating factor, is a membrane lipid microdomain-associated, GPI-anchored protein implicated in the shielding of cells from complement-mediated attack via accelerating decay of C3 and C5. Loss of CD55 is associated with a number of pathologies due to hyperactivation of the complement system. CD55 is also implicated in cancer progression thought to be driven via its role in cell shielding mechanisms. We now appreciate that CD55 can signal intracellularly to promote malignant transformation, cancer progression, cell survival, angiogenesis, and inhibition of apoptosis. Outside-in signaling via CD55 is mediated by signaling pathways including JNK, JAK/STAT, MAPK/NF-κB, and LCK. Moreover, CD55 is enriched in the cancer stem cell (CSC) niche of multiple tumors including breast, ovarian, cervical, and can be induced by chemotherapeutics and hypoxic environments. CSCs are implicated in tumor recurrence and chemoresistance. Here, we review the unexpected roles of CD55 in cancer including the roles of canonical and noncanonical pathways that CD55 orchestrates. We will highlight opportunities for therapeutic targeting CD55 and gaps in the field that require more in-depth mechanistic insights.
Collapse
Affiliation(s)
- Rashmi Bharti
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Goutam Dey
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Feng Lin
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Justin Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Ofer Reizes
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
3
|
Complement Decay-Accelerating Factor is a modulator of influenza A virus lung immunopathology. PLoS Pathog 2021; 17:e1009381. [PMID: 34197564 PMCID: PMC8248730 DOI: 10.1371/journal.ppat.1009381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Clearance of viral infections, such as SARS-CoV-2 and influenza A virus (IAV), must be fine-tuned to eliminate the pathogen without causing immunopathology. As such, an aggressive initial innate immune response favors the host in contrast to a detrimental prolonged inflammation. The complement pathway bridges innate and adaptive immune system and contributes to the response by directly clearing pathogens or infected cells, as well as recruiting proinflammatory immune cells and regulating inflammation. However, the impact of modulating complement activation in viral infections is still unclear. In this work, we targeted the complement decay-accelerating factor (DAF/CD55), a surface protein that protects cells from non-specific complement attack, and analyzed its role in IAV infections. We found that DAF modulates IAV infection in vivo, via an interplay with the antigenic viral proteins hemagglutinin (HA) and neuraminidase (NA), in a strain specific manner. Our results reveal that, contrary to what could be expected, DAF potentiates complement activation, increasing the recruitment of neutrophils, monocytes and T cells. We also show that viral NA acts on the heavily sialylated DAF and propose that the NA-dependent DAF removal of sialic acids exacerbates complement activation, leading to lung immunopathology. Remarkably, this mechanism has no impact on viral loads, but rather on the host resilience to infection, and may have direct implications in zoonotic influenza transmissions. Exacerbated complement activation and immune deregulation are at the basis of several pathologies induced by respiratory viruses. Here, we report that complement decay-accelerating factor (DAF), which inhibits complement activation in healthy cells, increases disease severity upon influenza A virus (IAV) infection. Remarkably, DAF interaction with IAV proteins, hemagglutinin (HA) and neuraminidase (NA), resulted in excessive complement activation and recruitment of innate and adaptive immune cells, without affecting viral loads. Furthermore, we observed that viral NA directly cleaves DAF and promotes complement activation, providing a possible link between IAV-DAF interaction and pathology. Therefore, our results unveil a novel pathway that could modulate disease severity, which may help to understand the increased pathogenicity of zoonotic and pandemic IAV infections.
Collapse
|
4
|
Yin X, Shen C, Yin Y, Cai Z, Wang J, Zhao Z, Chen X, Chen Z, Chen H, Zhang B. Overexpression of CD55 correlates with tumor progression and poor prognosis in gastric stromal tumors. Onco Targets Ther 2019; 12:4703-4712. [PMID: 31417272 PMCID: PMC6594005 DOI: 10.2147/ott.s195182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/13/2019] [Indexed: 02/05/2023] Open
Abstract
Backgrounds: Accumulating evidences have demonstrated that CD55 can protect cells from complement-mediated attack, and is involved in tumor dedifferentiation, migration, invasiveness, and metastasis. However, the role of CD55 in gastrointestinal stromal tumors (GISTs) has not been investigated. Aims: Our study aimed to analyze the expression of CD55 in gastric GISTs and its correlations with clinicopathologic characteristics and prognosis. Materials and methods: A total of 118 gastric GIST patients were included in our study. CD55 expression in GIST tissue samples was evaluated using immunohistochemistry. Cumulative survival was conducted using the Kaplan-Meier method. Cox regression analyses were performed to identify factors associated with progression-free survival (PFS) for patients with gastric GISTs. Results: Of 118 gastric GISTs patients included in our study, 44 (37.3%) were positive for CD55 expression. Positive CD55 expression in gastric GISTs was closely associated with tumor size (13.52±7.35 vs 5.07±1.90 cm, respectively; P<0.001), Ki 67 labeling index (P=0.001), mitotic counts (P=0.005), NIH risk classification (P<0.001), PLR (P<0.001), and metastasis at initial diagnosis (P=0.002). Kaplan-Meier analyses revealed that tumor size (P<0.001), mitotic counts (P<0.001), Ki 67 labeling index (P<0.001), PLR (P<0.001), metastasis at initial diagnosis (P=0.031), and CD55 expression (P<0.001) were statistically significant risk factors affecting PFS of patients with gastric GISTs. Cox multivariate survival analysis showed that mitotic counts, Ki 67 labeling index, and CD55 expression were independent predictors of PFS for gastric GISTs. Conclusion: CD55 may be a potential prognostic marker in gastric GISTs patients.
Collapse
Affiliation(s)
- Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Chaoyong Shen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Zhaolun Cai
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Jian Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Zhou Zhao
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Xin Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Zhixin Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Huijiao Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| |
Collapse
|
5
|
Rebl A, Goldammer T. Under control: The innate immunity of fish from the inhibitors' perspective. FISH & SHELLFISH IMMUNOLOGY 2018; 77:328-349. [PMID: 29631025 DOI: 10.1016/j.fsi.2018.04.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 06/08/2023]
Abstract
The innate immune response involves a concerted network of induced gene products, preformed immune effectors, biochemical signalling cascades and specialised cells. However, the multifaceted activation of these defensive measures can derail or overshoot and, if left unchecked, overwhelm the host. A plenty of regulatory devices therefore mediate the fragile equilibrium between pathogen defence and pathophysiological manifestations. Over the past decade in particular, an almost complete set of teleostean sequences orthologous to mammalian immunoregulatory factors has been identified in various fish species, which prove the remarkable conservation of innate immune-control concepts among vertebrates. This review will present the current knowledge on more than 50 teleostean regulatory factors (plus additional fish-specific paralogs) that are of paramount importance for controlling the clotting cascade, the complement system, pattern-recognition pathways and cytokine-signalling networks. A special focus lies on those immunoregulatory features that have emerged as potential biomarker genes in transcriptome-wide research studies. Moreover, we report on the latest progress in elucidating control elements that act directly with immune-gene-encoding nucleic acids, such as transcription factors, hormone receptors and micro- and long noncoding RNAs. Investigations into the function of teleostean inhibitory factors are still mainly based on gene-expression profiling or overexpression studies. However, in support of structural and in-vitro analyses, evidence from in-vivo trials is also available and revealed many biochemical details on piscine immune regulation. The presence of multiple gene copies in fish adds a degree of complexity, as it is so far hardly understood if they might play distinct roles during inflammation. The present review addresses this and other open questions that should be tackled by fish immunologists in future.
Collapse
Affiliation(s)
- Alexander Rebl
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Fish Genetics Unit, Dummerstorf, Germany.
| | - Tom Goldammer
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Fish Genetics Unit, Dummerstorf, Germany
| |
Collapse
|
6
|
Apyrase Elicits Host Antimicrobial Responses and Resolves Infection in Burns. J Burn Care Res 2018; 37:e501-e507. [PMID: 27058579 DOI: 10.1097/bcr.0000000000000335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The authors previously reported that adenosine triphosphate (ATP) stimulates biofilm formation and removal of the ATP could reduce biofilm formation. The main objective of this study was to evaluate the effects of the ATP-hydrolyzing enzyme, apyrase, on control of Acinetabacter baumannii infection in the burn wound as well as to assess host skin antimicrobial responses. The authors found that apyrase stimulated nitric oxide formation at the wound site and reduced CD55 expression, thereby inducing the assembly of membrane attack complexes. Apyrase treatment nearly eradicated multidrug-resistant A. baumannii from burn wounds in the absence of antibiotics. Apyrase may be an effective therapy against antibiotic-resistant bacterial infections in burns.
Collapse
|
7
|
The role of decay accelerating factor in environmentally induced and idiopathic systemic autoimmune disease. Autoimmune Dis 2014; 2014:452853. [PMID: 24592327 PMCID: PMC3921935 DOI: 10.1155/2014/452853] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/19/2013] [Indexed: 01/05/2023] Open
Abstract
Decay accelerating factor (DAF) plays a complex role in the immune system through complement-dependent and -independent regulation of innate and adaptive immunity. Over the past five years there has been accumulating evidence for a significant role of DAF in negatively regulating adaptive T-cell responses and autoimmunity in both humans and experimental models. This review discusses the relationship between DAF and the complement system and highlights major advances in our understanding of the biology of DAF in human disease, particularly systemic lupus erythematosus. The role of DAF in regulation of idiopathic and environmentally induced systemic autoimmunity is discussed including studies showing that reduction or absence of DAF is associated with autoimmunity. In contrast, DAF-mediated T cell activation leads to cytokine expression consistent with T regulatory cells. This is supported by studies showing that interaction between DAF and its molecular partner, CD97, modifies expression of autoimmunity promoting cytokines. These observations are used to develop a hypothetical model to explain how DAF expression may impact T cell differentiation via interaction with CD97 leading to T regulatory cells, increased production of IL-10, and immune tolerance.
Collapse
|
8
|
Membrane-bound complement regulatory proteins as biomarkers and potential therapeutic targets for SLE. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:55-81. [PMID: 23402019 DOI: 10.1007/978-1-4614-4118-2_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
For the last two decades, there had been remarkable advancement in understanding the role of complement regulatory proteins in autoimmune disorders and importance of complement inhibitors as therapeutics. Systemic lupus erythematosus is a prototype of systemic autoimmune disorders. The disease, though rare, is potentially fatal and afflicts women at their reproductive age. It is a complex disease with multiorgan involvement, and each patient presents with a different set of symptoms. The diagnosis is often difficult and is based on the diagnostic criteria set by the American Rheumatology Association. Presence of antinuclear antibodies and more specifically antidouble-stranded DNA indicates SLE. Since the disease is multifactorial and its phenotypes are highly heterogeneous, there is a need to identify multiple noninvasive biomarkers for SLE. Lack of validated biomarkers for SLE disease activity or response to treatment is a barrier to the efficient management of the disease, drug discovery, as well as development of new therapeutics. Recent studies with gene knockout mice have suggested that membrane-bound complement regulatory proteins (CRPs) may critically determine the sensitivity of host tissues to complement injury in autoimmune and inflammatory disorders. Case-controlled and followup studies carried out in our laboratory suggest an intimate relation between the level of DAF, MCP, CR1, and CD59 transcripts and the disease activity in SLE. Based on comparative evaluation of our data on these four membrane-bound complement regulatory proteins, we envisaged CR1 and MCP transcripts as putative noninvasive disease activity markers and the respective proteins as therapeutic targets for SLE. Following is a brief appraisal on membrane-bound complement regulatory proteins DAF, MCP, CR1, and CD59 as biomarkers and therapeutic targets for SLE.
Collapse
|
9
|
Billig EMW, O'Meara WP, Riley EM, McKenzie FE. Developmental allometry and paediatric malaria. Malar J 2012; 11:64. [PMID: 22394452 PMCID: PMC3331816 DOI: 10.1186/1475-2875-11-64] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 03/06/2012] [Indexed: 12/02/2022] Open
Abstract
WHO estimates that 80% of mortality due to malaria occurs among infants and young children. Though it has long been established that malaria disproportionately affects children under age five, our understanding of the underlying biological mechanisms for this distribution remains incomplete. Many studies use age as an indicator of exposure, but age may affect malaria burden independently of previous exposure. Not only does the severity of malaria infection change with age, but the clinical manifestation of disease does as well: younger children are more likely to suffer severe anaemia, while older children are more likely to develop cerebral malaria. Intensity of transmission and acquired immunity are important determinants of this age variation, but age differences remain consistent over varying transmission levels. Thus, age differences in clinical presentation may involve inherent age-related factors as well as still-undiscovered facets of acquired immunity, perhaps including the rates at which relevant aspects of immunity are acquired. The concept of "allometry" - the relative growth of a part in relation to that of an entire organism or to a standard - has not previously been applied in the context of malaria infection. However, because malaria affects a number of organs and cells, including the liver, red blood cells, white blood cells, and spleen, which may intrinsically develop at rates partly independent of each other and of a child's overall size, developmental allometry may influence the course and consequences of malaria infection. Here, scattered items of evidence have been collected from a variety of disciplines, aiming to suggest possible research paths for investigating exposure-independent age differences affecting clinical outcomes of malaria infection.
Collapse
Affiliation(s)
- Erica M W Billig
- Fogarty International Center, National Institutes of Health, Building 16, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
10
|
Spendlove I, Sutavani R. The role of CD97 in regulating adaptive T-cell responses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 706:138-48. [PMID: 21618833 DOI: 10.1007/978-1-4419-7913-1_12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
CD97 was identified as an early activation marker on T cells, having low expression on naive T cells. This is a common feature of molecules that have a role in T-cell function. It was subsequently identified as a ligand for CD55, which has been previously identified as an innate regulator of complement. The interaction of this receptor-ligand pair has been shown to provide a potent costimulatory signal to human T cells, despite their modest affinity. Though both CD97 and CD55 are expressed on T cells as well as antigen presenting cells (APCs), their interaction is significant when CD97 on APCs interacts with CD55 on T cells. The converse interaction is poorly defined and may be less significant. A unique aspect of the interaction of CD97 with CD55 is the stimulation of naive T cells, leading to the induction of IL-10 producing cells that behave like Trl regulatory cells. This raises a number of questions regarding the dual functions of CD55; regulating complement and stimulating T cells via CD97 interaction and any potential overlap in the consequences of these dual roles.
Collapse
Affiliation(s)
- Ian Spendlove
- The University of Nottingham, Academic Clinical Oncology, The City Hospital, Hucknall Road, Nottingham, Ng5 1PB, UK.
| | | |
Collapse
|
11
|
Expression of the decay-accelerating factor (CD55) in renal transplants--a possible prediction marker of allograft survival. Transplantation 2009; 88:457-64. [PMID: 19696627 DOI: 10.1097/tp.0b013e3181b0517d] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Decay-accelerating factor (CD55) accelerates the decay of C3 and C5 convertases, participating in classical and alternative complement activation pathways. Complement activation plays a major role in antibody-mediated rejection of allografts (AMR); C4d is used as a marker of AMR. Emerging evidence suggests an important role of CD55 in the pathogenesis of AMR. The aim of this study was to investigate the expression of CD55 in renal allografts and to correlate it with the expression of C4d, allograft survival, changes in serum creatinine (SC). METHODS More than 200 renal allograft biopsies, performed for allograft dysfunction, were assessed for peritubular capillary (PTC) C4d and CD55 expression. RESULTS We found significant correlation between changes in SC and PTC CD55 staining pattern in patients with no PTC C4d staining. In these patients, SC increased from baseline by 2.2+0.34, 1.7+0.36, and 0.93+0.24 mg/dL in negative, focal, and diffuse PTC CD55 staining subgroups, respectively. Survival of renal allografts was better in diffuse PTC CD55 staining subgroup than in negative PTC CD55 staining subgroup. CONCLUSIONS These data suggest that CD55 expression has a protective effect on PTC C4d negative renal allografts, and the pattern of PTC CD55 expression may be used as a potential marker of renal allograft survival in patients with no evidence of AMR.
Collapse
|
12
|
Davitz MA. Decay-accelerating factor (DAF): a review of its function and structure. ACTA MEDICA SCANDINAVICA. SUPPLEMENTUM 2009; 715:111-21. [PMID: 2438906 DOI: 10.1111/j.0954-6820.1987.tb09911.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
13
|
Parker CJ. Isolation and functional assay of the membrane complement inhibitors CD55 (DAF) and CD59 (MIRL). CURRENT PROTOCOLS IN IMMUNOLOGY 2008; Chapter 13:13.5.1-13.5.18. [PMID: 18432718 DOI: 10.1002/0471142735.im1305s11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The complement system is the primary effector of humoral immunity. Because of its enormous destructive capacity, mechanisms for confining the activity of the system to the desired target and elaborate safeguards for protecting self against complement-mediated injury have evolved. Human cells, particularly those found at sites of inflammation (e.g., hematopoietic and endothelial cells), express highly specialized membrane constituents that act independently or in concert with plasma regulatory proteins to inhibit the functional activity of complement. Decay-accelerating factor (DAF), or CD55, directly inhibits the formation and stability of the amplification C3 and C5 convertases of both the classical and the alternative pathways. Failure of a cell to regulate the amplification C3 and C5 convertases allows the generation of the potentially cytolytic membrane attack complex (MAC), or C5b-9 (consisting of the complement components C5b, C6, C7, C8, and C9). The primary cellular regulator of the MAC is the membrane inhibitor of reactive lysis (MIRL), or CD59, which restricts complement-mediated lysis by blocking assembly of the MAC (primarily at the stage of C9 binding and polymerization). This unit provides a basic protocol for isolating CD55 and CD59, along with two support protocols describing separate functional assays for CD59 and CD55.
Collapse
Affiliation(s)
- C J Parker
- University of Utah School of Medicine and The Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
14
|
Parker CJ. Paroxysmal nocturnal hemoglobinuria: an historical overview. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2008; 2008:93-103. [PMID: 19074065 DOI: 10.1182/asheducation-2008.1.93] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The clinical hallmark of paroxysmal nocturnal hemoglobinuria (PNH) is episodic hemoglobinuria, and it was this feature that captured the attention of European physicians in the latter half of the 19th century, resulting in careful observational studies that established PNH as an entity distinct from paroxysmal cold hemoglobinuria and march hemoglobinuria. Curiosity about the etiology of the nocturnal aspects of the hemoglobinuria led the German physician Paul Strübing to develop the prescient hypothesis that the erythrocytes of PNH are abnormally sensitive to hemolysis when the plasma is acidified during sleep because of accumulation of carbon dioxide and lactic acid as a result of slowing of the circulation. Investigation of the intricate pathophysiology that underlies the abnormal sensitivity of PNH erythrocytes to hemolysis in acidified serum produced a number of remarkable scientific achievements that involved discovery of the alternative pathway of complement, identification of the membrane proteins that regulate complement, discovery of a novel mechanism for attachment of proteins to the cell surface, and identification of the genetic basis of the disease. These discoveries were made steadily over a period of more than 100 years, and each generation of physicians and scientists made important contributions to the field. The mysteries of PNH have been solved in a particularly satisfying way because the precision and orderliness of the solutions made clearly understandable what had seemed at the times prior to resolution to be problems of nearly insurmountable complexity. The history of PNH is an inspirational reminder of the elegant complexity of nature, the rewards of curiosity and the power and beauty of science.
Collapse
Affiliation(s)
- Charles J Parker
- Division of Hematology and Bone Marrow Transplantation, University of Utah School of Medicine, Salt Lake City, UT 84148, USA.
| |
Collapse
|
15
|
Mikesch JH, Buerger H, Simon R, Brandt B. Decay-accelerating factor (CD55): a versatile acting molecule in human malignancies. Biochim Biophys Acta Rev Cancer 2006; 1766:42-52. [PMID: 16784816 DOI: 10.1016/j.bbcan.2006.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2005] [Revised: 04/07/2006] [Accepted: 04/19/2006] [Indexed: 10/24/2022]
Abstract
The decay-accelerating factor (DAF, CD55) physiologically serves as an inhibitor of the complement system. Moreover, DAF is broadly expressed in malignant tumors. Here, DAF seems to dispose of several different functions reaching far beyond its immunological role, e.g., promotion of tumorigenesis, decrease of complement mediated tumor cell lysis, autocrine loops for cell rescue and evasion of apoptosis, neoangiogenesis, invasiveness, cell motility, and metastasis via oncogenic tyrosine kinase pathway activation, and specific seven-span transmembrane receptors (CD97) binding. Furthermore, DAF has already been included in diagnostic or therapeutic studies. Thereby, studies applying monoclonal anti-DAF antibodies and anti-DAF vaccination for a targeted therapy have been enrolled recently.
Collapse
Affiliation(s)
- Jan-Henrik Mikesch
- Department of Haematology-Oncology, University Hospital, Westf.-Wilhelms-Univ. Münster, Germany
| | | | | | | |
Collapse
|
16
|
Affiliation(s)
- Charles J Parker
- Hematology/Oncology Section, VA Medical Center, Salt Lake City, UT 84148, USA.
| |
Collapse
|
17
|
Pérez de la Lastra JM, Harris CL, Hinchliffe SJ, Holt DS, Rushmere NK, Morgan BP. Pigs express multiple forms of decay-accelerating factor (CD55), all of which contain only three short consensus repeats. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:2563-73. [PMID: 10946283 DOI: 10.4049/jimmunol.165.5.2563] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We report the cloning of cDNAs encoding multiple isoforms of the pig analogue of human decay-accelerating factor (DAF; CD55). Screening of a pig muscle cDNA library using a human DAF probe identified a single clone that encoded a DAF-like molecule comprising three short consensus repeats (SCR) homologous with the amino-terminal three SCR in human DAF, a serine/threonine-rich (ST) region, and sequence compatible with a transmembrane domain and cytoplasmic tail. Northern blot and RT-PCR analysis showed that pig DAF was expressed in a wide range of tissues. Additional isoforms of DAF were sought using RT-PCR and 3'-rapid amplification of cDNA ends followed by sequencing. Isoforms containing a GPI anchor and with differing lengths of ST region were identified; no isoform containing a fourth SCR was found. Cloning of the GPI-anchored isoform from granulocytes confirmed that it was identical with the original transmembrane isoform through the three SCR and first portion of ST and was derived from a frame shift caused by splicing out 176 bp of sequence. A panel of mAbs was generated and used to analyze the distribution and anchoring of pig DAF in circulating cells. Pig DAF was expressed on all circulating cells and was transmembrane anchored on erythrocytes, but completely or partially GPI anchored on granulocytes and mononuclear cells. The transmembrane isoform of pig DAF was expressed on Chinese hamster ovary cells and was shown to affect regulatory activity for the classical pathway of human complement, but was only marginally active against pig serum.
Collapse
Affiliation(s)
- J M Pérez de la Lastra
- Complement Biology Group, Department of Medical Biochemistry, University of Wales College of Medicine, Heath Park, Cardiff, United Kingdom
| | | | | | | | | | | |
Collapse
|
18
|
Harris CL, Spiller OB, Morgan BP. Human and rodent decay-accelerating factors (CD55) are not species restricted in their complement-inhibiting activities. Immunology 2000; 100:462-70. [PMID: 10929073 PMCID: PMC2327033 DOI: 10.1046/j.1365-2567.2000.00066.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Homologous complement activation is restricted on cells by the complement regulators, decay-accelerating factor (DAF), membrane cofactor protein (MCP) and CD59. These proteins act in concert with other membrane structures to protect cells from homologous complement attack. In contrast, cells are usually sensitive to heterologous complement attack. It has been suggested that species-specific restriction of complement activation can be attributed to the inability of regulators to inhibit across species. We have investigated the capacities of human, rat and mouse analogues of DAF to regulate homologous and heterologous complement. Cells transfected with cDNA encoding these analogues were protected from heterologous complement attack. C3b-deposition experiments indicated that whilst cells were best protected by DAF from the same species, all three analogues inhibited human, rat and mouse complement. Comparable results were obtained in haemolysis assays using soluble, recombinant forms of the proteins. Inhibition of the classical pathway (CP) was best achieved with homologous DAF, although human DAF also inhibited rat complement, rat DAF also inhibited human complement and mouse DAF inhibited complement from all species. Human DAF was the best inhibitor of alternative pathway (AP)-mediated attack, inhibiting complement from all species. Mouse DAF inhibited mouse and rat AP, whilst rat DAF inhibited only rat AP. These data indicate that human and rodent analogues of DAF are not species restricted and highlights interesting differences in the capacity to regulate AP and CP. This has implications in broader fields of research, such as xenotransplantation, where cross-species regulation of complement is of paramount importance.
Collapse
Affiliation(s)
- C L Harris
- Complement Biology Group, Department of Medical Biochemistry, University of Wales College of Medicine, Heath Park, Cardiff, UK
| | | | | |
Collapse
|
19
|
Eisenschenk FC, Houle JJ, Hoffmann EM. Mechanism of serum resistance among Brucella abortus isolates. Vet Microbiol 1999; 68:235-44. [PMID: 10510042 DOI: 10.1016/s0378-1135(99)00075-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
It was shown in this study that complement-resistant Brucella abortus used were unable to activate complement in the absence of specific antibody. Complement-resistant isolates possessed O-antigen, but complement-sensitive organisms used are O-antigen deficient. Since B. abortus LPS does not activate the alternative pathway of complement, we concluded that activation of bovine complement must be due to some other mechanism. In this study, it was shown that bovine C1 binds to the outer membrane proteins of B. abortus. Isolated outer membrane proteins of both smooth (O-antigen positive) and rough (O-antigen negative) B. abortus used bind to C1q. However, only rough isolates were killed by complement. All of the O-antigen positive B. abortus isolates were complement-resistant. We propose that O-antigen shields outer membrane proteins and blocks C1q binding.
Collapse
Affiliation(s)
- F C Eisenschenk
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville 32611, USA
| | | | | |
Collapse
|
20
|
Abstract
Because of its strong potential for generating inflammation and causing tissue destruction the complement system has to be kept strictly under control. Cells of the host need special protection against the cytolytic complement system. This paper will describe how inappropriate activation of complement in the fluid phase is prevented and how viable human blood cells defend themselves against being destroyed and cleared away by the complement system. Since disturbances in complement regulation occasionally result in disease a brief reference will be made to two of the syndromes caused by complement regulator deficiency, hereditary angioedema (HAE) and paroxysmal nocturnal hemoglobinuria (PNH).
Collapse
Affiliation(s)
- S Meri
- Department of Bacteriology and Immunology, Haartman Institute, Helsinki University Central Hospital, Finland.
| | | |
Collapse
|
21
|
Watkins NJ, Braidley P, Bray CJ, Savill CM, White DJ. Coating of human decay accelerating factor (hDAF) onto medical devices to improve biocompatibility. IMMUNOPHARMACOLOGY 1997; 38:111-8. [PMID: 9476122 DOI: 10.1016/s0162-3109(97)00068-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In passing blood through an artificial circulatory system, the blood is exposed to surfaces that result in activation of the complement system. The consequences of the activation of complement can be extremely serious for the patient ranging from mild discomfort to respiratory distress and even anaphylaxis. An entirely novel approach was to express recombinant GPI anchored human decay accelerating factor (hDAF) using the baculovirus system and then coat the recombinant protein onto the surfaces of these materials to reduce complement activation. Expression of hDAF in Sf9 cells was shown by ELISA, FACS analysis, and Western blot. Functional activity was tested by CH50 assay. For the coating experiments a small scale model of a cardiovascular bypass circuit constructed from COBE tubing was used. hDAF was either coated onto the circuit using adsorption or covalently linked via the photoreactive crosslinker, p-azidobenzoyl hydrazide. After coating, heparinised human blood was pumped around the circuit and samples were collected into EDTA collection tubes at different time points. Complement activation was measured using a Quidel C3a-des-arg EIA. The photolinked circuits gave a reduction in C3a production of 20-50%, compared to 10-20% seen with an absorbed hDAF circuit. Furthermore, the inhibition of complement was seen over the whole time scale of the photolinked circuit, 60-90 min, whilst in the adsorbed circuit inhibition was not seen to a significant degree after 60 min. The time scale of a standard cardiac bypass is 45-90 min, therefore, the photolinked circuit results are encouraging, as significant inhibition of complement activation is seen within this time frame.
Collapse
Affiliation(s)
- N J Watkins
- Imutran Ltd. (A Novartis Pharma AG Company), Cambridge, UK
| | | | | | | | | |
Collapse
|
22
|
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired, clonal disorder of hematopoietic cells caused by somatic mutation in the X-linked PIGA gene encoding a protein involved in the synthesis of the glycosylphosphatidylinositol (GPI) anchor by which many proteins are attached to the membrane of cells. About 15 proteins have been found to be lacking or markedly deficient on the abnormal blood cells. These defects result in a clinical syndrome that includes intravascular hemolysis mediated by complement, unusual venous thromboses, deficits of hematopoiesis, and other manifestations. Therapy is presently directed mainly at the consequences of the disorder rather than its basic causes and includes replacement of iron, folic acid, and whole blood; hormonal modulation (prednisone, androgens); anticoagulation; and bone marrow transplantation. PNH is a chronic disease with more than half of adult patients surviving 15 years or more; prognosis is less good in children.
Collapse
Affiliation(s)
- W F Rosse
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
23
|
Atkinson JP. Impact of the discovery of membrane inhibitors of complement. RESEARCH IN IMMUNOLOGY 1996; 147:95-100. [PMID: 8792467 DOI: 10.1016/0923-2494(96)87180-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- J P Atkinson
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| |
Collapse
|
24
|
Davies A. Policing the membrane: cell surface proteins which regulate complement. RESEARCH IN IMMUNOLOGY 1996; 147:82-7. [PMID: 8792465 DOI: 10.1016/0923-2494(96)87178-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- A Davies
- MIP Unit, MRC Centre, Cambridge, UK
| |
Collapse
|
25
|
Abstract
The complement system has developed a remarkably simple but elegant manner of regulating itself. It has faced and successfully dealt with how to facilitate activation on a microbe while preventing the same on host tissue. It solved this problem primarily by creating a series of secreted and membrane-regulatory proteins that prevent two highly undesirable events: activation in the fluid phase (no target) and on host tissue (inappropriate target). Also, if not checked, even on an appropriate target, the system would go to exhaustion and have nothing left for the next microbe. Therefore, the complement enzymes have an intrinsic instability and the fluid-phase control proteins play a major role in limiting activation in time. The symmetry of the regulatory process between fluid phase and membrane inhibitors at the C4/C3 step of amplification and convertase formation as well as at the MAC steps are particularly striking features of the self/nonself discrimination system. The use of glycolipid anchored proteins on membranes to decay enzymes and block membrane insertion events is unlikely to be by chance. Finally, it is economical for the cofactor regulatory activity to produce derivatives of C3b that now specifically engage additional receptors. Likewise, C1-Inh leads to C1q remaining on the immune complex to interact with the C1q receptor. Thus the complement system is designed to allow rapid, efficient, unimpeded activation on an appropriate foreign target while regulatory proteins intervene to prevent three undesirable consequences of complement activation: excessive activation on a single target, fluid phase activation, and activation on self.
Collapse
Affiliation(s)
- M K Liszewski
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
26
|
Morgan BP, Meri S. Membrane proteins that protect against complement lysis. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1994; 15:369-96. [PMID: 8153873 DOI: 10.1007/bf01837366] [Citation(s) in RCA: 174] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- B P Morgan
- Department of Medical Biochemistry, University of Wales College of Medicine, Heath Park, Cardiff, UK
| | | |
Collapse
|
27
|
Kusakari C, Nose M, Takasaka T, Yuasa R, Kato M, Miyazono K, Fujita T, Kyogoku M. Immunopathological features of palatine tonsil characteristic of IgA nephropathy: IgA1 localization in follicular dendritic cells. Clin Exp Immunol 1994; 95:42-8. [PMID: 7507015 PMCID: PMC1534624 DOI: 10.1111/j.1365-2249.1994.tb06012.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
IgA nephropathy (IgAN) is generally thought to be mediated by the glomerular deposition of circulating immune complexes containing IgA as the major antibody component. Upper respiratory infections and tonsillitis often precede IgAN, and in some cases tonsillectomy is effective for the treatment of IgAN. Thus, the tonsil seems to be a unique organ causing initial and/or progressive events to generate nephritogenic immune complexes in IgAN. In this study we focused on the analysis of immunopathological features of the palatine tonsil characteristic of IgAN patients by using an immunohistochemical technique. The IgA1 subclass was demonstrated in follicular dendritic cells (FDC) of the tonsil of IgAN patients, but not in FDC of non-IgAN controls. On the other hand, IgA2, IgG, IgM and C3 did not show any differences in distribution between the two groups. Moreover, the expression of decay-accelerating factor (DAF), an inhibitor of homologous complement activation, and transforming growth factor-beta 1 (TGF-beta 1), an inducer of antibody-producing cells to IgA class switching, in FDC and interdigitating dendritic cells of the tonsil, respectively, which was also clarified in this study for the first time, was found to be identically distributed in the two groups. These findings may support the idea that IgA1, possibly in an immune complex form, is trapped by FDC and plays an important role in the persistent activation of particular B cell repertoires responsible for the onset and/or progression of IgAN.
Collapse
Affiliation(s)
- C Kusakari
- Department of Pathology, Tohoku University School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Nowicki B, Hart A, Coyne KE, Lublin DM, Nowicki S. Short consensus repeat-3 domain of recombinant decay-accelerating factor is recognized by Escherichia coli recombinant Dr adhesin in a model of a cell-cell interaction. J Exp Med 1993; 178:2115-21. [PMID: 7504058 PMCID: PMC2191283 DOI: 10.1084/jem.178.6.2115] [Citation(s) in RCA: 141] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
A bacterial pathogen that is important in both urinary tract and intestinal infections is Escherichia coli which expresses Dr or related adhesins. In this report, we present a model for testing cell-cell interaction, using both molecularly characterized laboratory cells that express recombinant molecules of human decay-accelerating factor (DAF), and recombinant bacterial Dr colonization factors. Dr adhesin ligand was identified as DAF (CD55), a membrane protein that protects autologous tissues from damage due to the complement system. Structure-function studies mapped the adhesin-binding site on the DAF molecule. A single-point substitution in the third short consensus repeat domain, Ser165 to Leu, corresponding to the Dra to Drb allelic polymorphism, caused complete abolition of adhesin binding to DAF.
Collapse
Affiliation(s)
- B Nowicki
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston 77550
| | | | | | | | | |
Collapse
|
29
|
Petty AC, Daniels GL, Anstee DJ, Tippett P. Use of the MAIEA technique to confirm the relationship between the Cromer antigens and decay-accelerating factor and to assign provisionally antigens to the short-consensus repeats. Vox Sang 1993; 65:309-15. [PMID: 7508660 DOI: 10.1111/j.1423-0410.1993.tb02172.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The MAIEA (monoclonal-antibody-specific immobilisation of erythrocyte antigens) assay has recently been developed for the assignment of red cell antigens, recognised by human alloantisera, to particular membrane components of the red cell membrane. This technique detects trimolecular complexes formed by the reaction of a human antibody and a mouse antibody with a particular red cell protein. A positive reaction, in an ELISA-type detection procedure, occurs if the epitopes to the human and mouse antibodies are present on the same membrane component but at different regions. In this report, we show how the MAIEA assay can be used to confirm the relationship between Cromer system antigens and the complement-regulatory protein, decay-accelerating factor (DAF, CD 55). In addition, the location of the antigens along the protein is postulated by using three anti-DAF monoclonal antibodies with specificities to different regions of DAF. Tca and Esa are assigned provisionally to the first short-consensus repeat (SCR), UMC to the second SCR, Dra to the third SCR and Cra, WESa and WESb to the fourth SCR or to the serine/threonine rich region of the DAF protein.
Collapse
Affiliation(s)
- A C Petty
- MRC Blood Group Unit, University College, London, UK
| | | | | | | |
Collapse
|
30
|
Schieren G, Hänsch GM. Membrane-associated proteins regulating the complement system: functions and deficiencies. Int Rev Immunol 1993; 10:87-101. [PMID: 7688019 DOI: 10.3109/08830189309051173] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- G Schieren
- Institut für Immunologie, Univ. Heidelberg, Germany
| | | |
Collapse
|
31
|
Holmes CH, Simpson KL. Complement and pregnancy: new insights into the immunobiology of the fetomaternal relationship. BAILLIERE'S CLINICAL OBSTETRICS AND GYNAECOLOGY 1992; 6:439-60. [PMID: 1446417 DOI: 10.1016/s0950-3552(05)80005-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recent studies have revealed that human trophoblast expresses three membrane-bound proteins which function specifically to regulate the activity of complement. These proteins are already known to be widely distributed in normal adult tissues where they protect host cells from damage resulting from the fortuitous deposition of activated complement components. Their activities are focused at two distinct steps in the complement pathway. Decay accelerating factor (DAF, CD55) and membrane co-factor protein (MCP, CD46) act at the level of the C3 convertase enzymes which activate C3 to C3b. A further protein, CD59, directly regulates the formation and function of the terminal cytolytic membrane attack complex (MAC) by specifically interacting with C8 and C9. These proteins appear to play an important role in the maintenance of normal human pregnancy. DAF, MCP and CD59 are all expressed where trophoblast surfaces are in contact with maternal blood and tissues and expression occurs from at least 6 weeks of gestation. The semi-allogeneic human conceptus therefore appears to be effectively protected from maternal complement-mediated damage arising either from alternative or classical pathway activation or in a bystander fashion following a response to microbial infection in the mother. Complement regulatory protein deficiency disorders with clinically demonstrable consequences especially in terms of haemolytic disease are known to exist and have proved valuable in establishing the biological role of these proteins in vivo. The demonstration of this new family of immunoregulatory proteins on trophoblast raises important questions about the potential involvement of these products in pregnancy pathologies.
Collapse
Affiliation(s)
- C H Holmes
- University of Bristol, Department of Obstetrics and Gynaecology, St Michael's Hospital, UK
| | | |
Collapse
|
32
|
Hänsch GM. The complement attack phase: control of lysis and non-lethal effects of C5b-9. IMMUNOPHARMACOLOGY 1992; 24:107-17. [PMID: 1473962 DOI: 10.1016/0162-3109(92)90017-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- G M Hänsch
- Institut für Immunologie, Universität Heidelberg, FRG
| |
Collapse
|
33
|
Kuraya M, Yefenof E, Klein G, Klein E. Expression of the complement regulatory proteins CD21, CD55 and CD59 on Burkitt lymphoma lines: their role in sensitivity to human serum-mediated lysis. Eur J Immunol 1992; 22:1871-6. [PMID: 1378022 DOI: 10.1002/eji.1830220729] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
On a panel of nine human B cell lines we showed that the expression of the complement regulatory factors complement receptor type 2 (CR2; CD21), decay-accelerating factor, (DAF; CD55) and homologous restriction factor (HRF20, CD59) is not correlated. All lines expressed DAF, six lines carried detectable amounts of CR2 and three carried HRF20. Upon incubation in human serum, under conditions which allowed the activation of complement through the alternative pathway, the CR2-carrying lines bound C3 fragments and two of them (Ramos and one of its two sublines) were damaged. These two lines had the lowest DAF expression, less than 50% of the cells reacted with the IA10 monoclonal antibody. By modulating the expression of the complement regulatory molecules, the lytic sensitivity of the B cell lines could be altered. Blockade of DAF on the HRF20-, CR2+ lines with the specific monoclonal antibodies increased their sensitivity to lysis by human serum. With the DAF- and HRF20+ cells significant lytic effect was obtained only when they were pretreated with both of the specific antibodies. Interferon-gamma or tumor necrosis factor-alpha treatment elevated the amount of CR2 on the low-CR2 expressor line (Ramos/HR1K) which thereafter bound higher amounts of C3 fragments and was lysed when incubated in human serum. This line had relatively low DAF level and lacked HRF20. The cytokine treatment did not alter the expression of these molecules. The CR2+ Ramos and the CR2- Rael cells were treated with 5-azacytidine which induced HRF20 and increased DAF expression. In parallel with this change Ramos cells became resistant to C-mediated lysis. The experiments with the panel of human B cell lines showed thus that cytolysis through activation of complement in homologous serum can be regulated at several steps by cell surface molecules. While expression of CR2 was required for C3 fixation, DAF and HRF20 inhibited lysis. By independent modulation of the quantities of these molecules, cells acquired or lost their sensitivity.
Collapse
Affiliation(s)
- M Kuraya
- Department of Tumor Biology, Karolinska Institute, Stockholm, Sweden
| | | | | | | |
Collapse
|
34
|
Toney DM, Marciano-Cabral F. Alterations in protein expression and complement resistance of pathogenic Naegleria amoebae. Infect Immun 1992; 60:2784-90. [PMID: 1319405 PMCID: PMC257235 DOI: 10.1128/iai.60.7.2784-2790.1992] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Highly pathogenic strains of Naegleria fowleri activate the alternative complement pathway but are resistant to lysis. In contrast, weakly pathogenic and nonpathogenic Naegleria spp. activate the complement pathway and are readily lysed. The present study was undertaken to determine whether surface components on amoebae accounted for resistance to complement lysis. Enzymatic removal of surface components from highly pathogenic N. fowleri with phosphatidylinositol-specific phospholipase C or with endoglycosidase H increased the susceptibility of these amoebae to complement-mediated lysis. Similar treatment of nonpathogenic amoebae had no effect on susceptibility to complement. Tunicamycin treatment of highly and weakly pathogenic N. fowleri increased susceptibility to lysis by complement in a dose-related manner. Tunicamycin treatment did not alter the susceptibility of nonpathogenic amoebae to complement. Proteins of 234 and 47 kDa were detected in supernatant fluid from phosphatidylinositol-specific phospholipase C-treated highly pathogenic amoebae but not in supernatant fluid from phosphatidylinositol-specific phospholipase C-treated weakly pathogenic amoebae. Electrophoretic analysis of iodinated surface proteins of highly pathogenic N. fowleri revealed species of 89, 60, 44, and 28 kDa. Western immunoblots of lysates from surface-iodinated amoebae were stained with biotinylated concanavalin A or biotinylated Ulex europaeus agglutinin I. Surface proteins, identified in highly pathogenic amoebae by iodination, were shown to be glycoproteins by lectin analysis specific for the detection of mannose and fucose residues.
Collapse
Affiliation(s)
- D M Toney
- Department of Microbiology and Immunology, Virginia Commonwealth University, Medical College of Virginia, Richmond 23298-0678
| | | |
Collapse
|
35
|
Ratnoff WD, Knez JJ, Prince GM, Okada H, Lachmann PJ, Medof ME. Structural properties of the glycoplasmanylinositol anchor phospholipid of the complement membrane attack complex inhibitor CD59. Clin Exp Immunol 1992; 87:415-21. [PMID: 1371955 PMCID: PMC1554345 DOI: 10.1111/j.1365-2249.1992.tb03012.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
CD59, the membrane regulator of autologous C5b-9 channel formation, exhibits variable sensitivity to cleavage by phosphatidylinositol-specific phospholipase C (PI-PLC), an enzyme that releases glyco-inositolphospholipid (GPI)-anchored proteins from cell surfaces. To determine whether the GPI-anchor phospholipid of CD59 is similar to that of decay-accelerating factor (DAF) and whether variation in its structure underlies its variable enzyme susceptibility, the GPI anchors of the two proteins expressed on erythrocytes, polymorphonuclear and mononuclear leucocytes were compared in situ and after purification. Flow cytometric analyses of PI-PLC-treated cells showed parallel cell type specific release of both proteins as a function of enzyme concentration. Non-denaturing PAGE analyses of alkaline/hydroxylamine-treated proteins (affinity-purified from [125I]-surface-labelled cells) provided evidence for (i) comparable proportions of GPI-anchor acylation, and (ii) alkali-resistant rather than alkali-sensitive lipid substituents in erythrocytes. These findings argue that the differential C5b-9 sensitivity that distinguishes paroxysmal nocturnal haemoglobinuria II and III erythrocytes does not derive from expression of CD59 molecules with alternative GPI-anchor phospholipid structures.
Collapse
Affiliation(s)
- W D Ratnoff
- Institute of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Horta MF, Ramalho-Pinto FJ, Fatima M. Role of human decay-accelerating factor in the evasion of Schistosoma mansoni from the complement-mediated killing in vitro. J Exp Med 1991; 174:1399-406. [PMID: 1720809 PMCID: PMC2119036 DOI: 10.1084/jem.174.6.1399] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Decay-accelerating factor (DAF) is a 70-kD membrane glycoprotein that prevents complement (C)-mediated hemolysis by blocking the assembly or accelerating the decay of C3 convertase. Purified DAF is known to incorporate into the membrane of DAF-deficient cells, inhibiting lysis. Since Schistosoma mansoni is a blood-dwelling parasite, we investigated whether DAF can be transferred from human erythrocytes to the worm and protect it against C-mediated killing in vitro. We have found that schistosomula (schla) incubated with normal human erythrocytes (N-HuE), but not with DAF-deficient erythrocytes, become resistant to C damage in vitro. Protected parasites acquire a 70-kD surface protein which can be immunoprecipitated by anti-DAF antibodies. The acquired resistance is abrogated by treatment of N-HuE-incubated parasites with anti-DAF antibody. These results indicate that, in vitro, N-HuE DAF can be transferred to schla, and suggest its participation in preventing their C-mediated killing. This could represent an important strategy of parasites to evade the host's immune response in vivo.
Collapse
Affiliation(s)
- M F Horta
- Department of Biochemistry-Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | |
Collapse
|
38
|
Atkinson JP, Oglesby TJ, White D, Adams EA, Liszewski MK. Separation of self from non-self in the complement system: a role for membrane cofactor protein and decay accelerating factor. Clin Exp Immunol 1991; 86 Suppl 1:27-30. [PMID: 1718640 PMCID: PMC1554035 DOI: 10.1111/j.1365-2249.1991.tb06203.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- J P Atkinson
- Howard Hughes Medical Institute Laboratories, Washington University School of Medicine, St. Louis, MO 63110
| | | | | | | | | |
Collapse
|
39
|
Itoh J, Nose M, Fujita T, Kato M, Ohyama A, Kyogoku M. Expression of decay-accelerating factor is reduced on hyperplastic synovial lining cells in rheumatoid synovitis. Clin Exp Immunol 1991; 83:364-8. [PMID: 1706235 PMCID: PMC1535342 DOI: 10.1111/j.1365-2249.1991.tb05644.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Decay-accelerating factor (DAF), a membrane inhibitor of homologous complement activation, is present in synovial cells lining joint space and detected in synovial fluid. DAF is considered to protect synovial membrane from complement-mediated injury associated with articular inflammation. We studied the immunohistopathological features of DAF molecules in synovial membrane of rheumatoid synovitis using a DAF-specific monoclonal antibody, 1C6. Reacting molecules with the 1C6 antibodies in synovial tissue extracts formed a 70-kD band in Western blot analysis. DAF was strongly detected on the flat synovial lining cells, but weakly on the hyperplastic and multi-layered lining cells in rheumatoid synovitis. The latter cells reacted with anti-Leu-M3 antibodies specific for a cell surface marker of activated macrophages, sometimes accompanied by C3 and IgM deposition on the superficial synovial membrane. These results suggest that active rheumatoid synovitis characteristically with hyperplastic synovial lining cells is out of control by DAF, thereby permitting further complement-mediated injury.
Collapse
Affiliation(s)
- J Itoh
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Okada N, Harada R, Okada H. Erythrocytes of patients with paroxysmal nocturnal haemoglobinuria acquire resistance to complement attack by purified 20-kD homologous restriction factor. Clin Exp Immunol 1990; 80:109-13. [PMID: 1691059 PMCID: PMC1535229 DOI: 10.1111/j.1365-2249.1990.tb06449.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A 20-kD homologous restriction factor (HRF20) which is a membrane inhibitor of the terminal stage of human complement action can be detected by the monoclonal antibody 1F5, and is deficient on abnormal erythrocytes as well as leucocytes from patients with paroxysmal nocturnal haemoglobinuria (PNH). The erythrocytes of PNH patients significantly improved their resistance to homologous complement after adsorption of purified HRF20.
Collapse
Affiliation(s)
- N Okada
- Department of Microbiology, Fukuoka University School of Medicine, Japan
| | | | | |
Collapse
|
41
|
Lublin DM, Atkinson JP. Decay-accelerating factor and membrane cofactor protein. Curr Top Microbiol Immunol 1990; 153:123-45. [PMID: 1688754 DOI: 10.1007/978-3-642-74977-3_7] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- D M Lublin
- Department of Pathology, Washington University School of Medicine, St. Louis, MO 63110
| | | |
Collapse
|
42
|
Whiteman LY, Marciano-Cabral F. Resistance of highly pathogenic Naegleria fowleri amoebae to complement-mediated lysis. Infect Immun 1989; 57:3869-75. [PMID: 2807551 PMCID: PMC259919 DOI: 10.1128/iai.57.12.3869-3875.1989] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Weakly pathogenic and nonpathogenic Naegleria spp. are readily lysed by human and guinea pig complement. Highly pathogenic Naegleria fowleri are resistant to complement-mediated lysis. Electrophoretic analysis of normal human serum (NHS) incubated with pathogenic or nonpathogenic Naegleria spp. demonstrates that amoebae activate the complement cascade, resulting in the production of C3 and C5 complement cleavage products. To determine whether surface constituents play a role in resistance to complement lysis, trophozoites of Naegleria spp. were subjected to enzymatic treatments prior to incubation in NHS. Treatment of trophozoites with papain or trypsin for 1 h, but not with neuraminidase, increased susceptibility of highly pathogenic Naegleria fowleri to complement lysis. Treatment of trophozoites with actinomycin D or cycloheximide during incubation with NHS or pretreatment with various protease inhibitors for 4 h did not increase the susceptibility of N. fowleri amoebae to lysis. Neither a repair process involving de novo protein synthesis nor a complement-inactivating protease appears to account for the increased resistance of N. fowleri amoebae to complement-mediated lysis.
Collapse
Affiliation(s)
- L Y Whiteman
- Department of Microbiology and Immunology, Virginia Commonwealth University, Medical College of Virginia, Richmond, 23298-0678
| | | |
Collapse
|
43
|
Morgan BP. Complement membrane attack on nucleated cells: resistance, recovery and non-lethal effects. Biochem J 1989; 264:1-14. [PMID: 2690818 PMCID: PMC1133540 DOI: 10.1042/bj2640001] [Citation(s) in RCA: 280] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- B P Morgan
- Department of Medical Biochemistry, University of Wales College of Medicine, Heath Park, Cardiff, U.K
| |
Collapse
|
44
|
Okada N, Harada R, Taguchi R, Okada H. Complete deficiency of 20 KDa homologous restriction factor (HRF20) and restoration with purified HRF20. Biochem Biophys Res Commun 1989; 164:468-73. [PMID: 2803313 DOI: 10.1016/0006-291x(89)91743-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
20 KDa homologous restriction factor (HRF20) is a membrane glycoprotein which inhibits formation of membrane attack complexes of homologous complement. Erythrocytes from a patient who is completely deficient in HRF20 were readily hemolyzed by homologous complement activated by sucrose or by acidification as in paroxysmal nocturnal hemoglobinuria (PNH). After incubating PNH erythrocytes (PNH-E) with purified HRF20, the cells were analyzed by flow cytometry using a monoclonal antibody to HRF20 and shown to have the antigen absorbed. These PNH-E acquired resistance to hemolysis by homologous complement suggesting that HRF20 may be successfully used for treatment of these patients.
Collapse
Affiliation(s)
- N Okada
- Department of Microbiology, Fukuoka University School of Medicine, Japan
| | | | | | | |
Collapse
|
45
|
Holguin MH, Fredrick LR, Bernshaw NJ, Wilcox LA, Parker CJ. Isolation and characterization of a membrane protein from normal human erythrocytes that inhibits reactive lysis of the erythrocytes of paroxysmal nocturnal hemoglobinuria. J Clin Invest 1989; 84:7-17. [PMID: 2738160 PMCID: PMC303946 DOI: 10.1172/jci114172] [Citation(s) in RCA: 283] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The observation that type III erythrocytes of paroxysmal nocturnal hemoglobinuria (PNH) are susceptible to hemolysis initiated by activated cobra venom factor complexes (CoFBb), whereas normal erythrocytes are resistant, implies that the PNH III cells are deficient in a membrane constituent that regulates this process. To isolate the inhibitory factor from normal erythrocytes, membrane proteins were first extracted with butanol and then subjected to sequential anion exchange, hydroxylapatite, and hydrophobic chromatography. Analysis by SDS-PAGE and silver stain of the inhibitory fractions showed a single band corresponding to a protein with an apparent Mr of 18 kD. PNH erythrocytes were incubated with incremental concentrations of the radiolabeled protein and then washed. In a dose-dependent fashion, the protein incorporated into the cell membrane and inhibited CoFBb-initiated lysis. This protein inhibitor functioned by restricting the assembly of the membrane attack complex at the level of C7 and C8 incorporation. By using a monospecific antibody to block the function of the inhibitor, it was shown that normal erythrocytes are rendered susceptible to CoFBb-initiated hemolysis. Analysis by Western blot of membrane proteins revealed that PNH III erythrocytes are deficient in the 18-kD protein. By virtue of its molecular weight and inhibitory activity, the 18-kD protein appears to be discrete from other previously described erythrocyte membrane proteins that regulate complement. These studies also indicate that the susceptibility of PNH III erythrocytes to reactive lysis is causally related to a deficiency of the 18-kD membrane inhibitor.
Collapse
Affiliation(s)
- M H Holguin
- Department of Medicine, University of Utah School of Medicine, Salt Lake City 84132
| | | | | | | | | |
Collapse
|
46
|
Abstract
Cromer-related antigens, a series of blood group antigens phenotypically related to each other through various serological characteristics, have recently been shown to be carried on the complement regulatory glycoprotein decay-accelerating factor (DAF). Cromer-related antigens, therefore, represent a number of inherited variants and polymorphisms of DAF. The Inab phenotype, a 'Cromer-null' phenotype in which the red cells lack all Cromer-related antigens, appears to be an inherited DAF deficiency.
Collapse
Affiliation(s)
- G Daniels
- Medical Research Council Blood Group Unit, London, UK
| |
Collapse
|
47
|
Affiliation(s)
- D J Anstee
- Blood Group Reference Laboratory, Radcliffe Infirmary, Oxford, UK
| | | |
Collapse
|
48
|
Affiliation(s)
- D Hourcade
- Howard Hughes Medical Institute Laboratories, Washington University School of Medicine, St. Louis, Missouri 63110
| | | | | |
Collapse
|
49
|
Dierich MP, Schulz TF, Eigentler A, Huemer H, Schwäble W. Structural and functional relationships among receptors and regulators of the complement system. Mol Immunol 1988; 25:1043-51. [PMID: 2975757 DOI: 10.1016/0161-5890(88)90136-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The classical and alternative pathway of complement activation are regulated by a series of fluid phase and cell-bound factors, some of which at the same time serve as receptors for fragments of C3 and C4. These molecules are factor H, CR1 (C3b/C4b receptor), CR2 (C3d/EBV receptor), C4BP (C4b binding protein), DAF (decay accelerating factor), MCP (membrane cofactor protein; earlier designated p45/70), CR3 (iC3b receptor or Mac-1) and CR4 (protein 150/95). Due to structural, genetic and functional features these factors are members of one or several newly recognized large families of proteins: (1) molecules with 60 amino acids long repeats (H, CR1, CR2, C4BP, DAF); (2) proteins with 1,2-diacylglycerol membrane anchoring (DAF); (3) proteins with a heterodimer structure and preference for ligands containing the tripeptide arginine-glycine-asparagine (CR3, CR4). Recognizing the above mentioned regulators and receptors of the complement system as belonging to these protein families opens new perspectives for further genetic and functional research of mutual interest to complement and noncomplement scientists.
Collapse
Affiliation(s)
- M P Dierich
- Institut für Hygiene, University of Innsbruck, Austria
| | | | | | | | | |
Collapse
|
50
|
Cheung NK, Walter EI, Smith-Mensah WH, Ratnoff WD, Tykocinski ML, Medof ME. Decay-accelerating factor protects human tumor cells from complement-mediated cytotoxicity in vitro. J Clin Invest 1988; 81:1122-8. [PMID: 2450893 PMCID: PMC329640 DOI: 10.1172/jci113426] [Citation(s) in RCA: 106] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The disialoganglioside GD2 is expressed on a wide spectrum of human tumor types, including neuroblastomas and melanomas. Upon binding of 3F8, a murine monoclonal antibody (MAb) specific for GD2, neuroblastomas and some melanomas are sensitive to killing by human complement, whereas some melanomas are not. To investigate the mechanism underlying these differences in complement mediated cytotoxicity, complement-insensitive melanoma cell lines were compared with respect to expression of the decay-accelerating factor (DAF), a membrane regulatory protein that protects blood cells from autologous complement attack. While DAF was undetectable among neuroblastomas, it was present in complement-insensitive melanomas. When the function of DAF was blocked by anti-DAF MAb, C3 uptake and complement-mediated lysis of the insensitive melanoma lines were markedly enhanced. F(ab')2 fragments were as effective in enhancing lysis as intact anti-DAF MAb. The DAF-negative and DAF-positive melanoma cell lines were comparably resistant to passive lysis by cobra venom factor-treated serum. The data suggest that in some tumors, DAF activity accounts for their resistance to complement-mediated killing. The ability to render these cells complement-sensitive by blocking DAF function may have implications for immunotherapy.
Collapse
Affiliation(s)
- N K Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York 10021
| | | | | | | | | | | |
Collapse
|