110 results on '"C4BP"'
Search Results
2. Conservation of C4BP-binding sequence patterns in Streptococcus pyogenes M and Enn proteins
- Author
-
Kolesiński, Piotr, McGowan, Matthew, Botteaux, Anne, Smeesters, Pierre R, and Ghosh, Partho
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Emerging Infectious Diseases ,Infectious Diseases ,Infection ,Streptococcus pyogenes ,Complement C4b-Binding Protein ,Antigens ,Bacterial ,Humans ,Bacterial Outer Membrane Proteins ,Carrier Proteins ,Protein Binding ,Amino Acid Sequence ,Bacterial Proteins ,C4BP ,M protein ,cross-reactivity ,immunogen ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Antigenically sequence variable M proteins of the major bacterial pathogen Streptococcus pyogenes (Strep A) are responsible for recruiting human C4b-binding protein (C4BP) to the bacterial surface, which enables Strep A to evade destruction by the immune system. The most sequence divergent portion of M proteins, the hypervariable region (HVR), is responsible for binding C4BP. Structural evidence points to the conservation of two C4BP-binding sequence patterns (M2 and M22) in the HVR of numerous M proteins, with this conservation applicable to vaccine immunogen design. These two patterns, however, only partially explain C4BP binding by Strep A. Here, we identified several M proteins that lack these patterns but still bind C4BP and determined the structures of two, M68 and M87 HVRs, in complex with a C4BP fragment. Mutagenesis of these M proteins led to the identification of amino acids that are crucial for C4BP binding, enabling formulation of new C4BP-binding patterns. Mutagenesis was also carried out on M2 and M22 proteins to refine or generate experimentally grounded C4BP-binding patterns. The M22 pattern was the most prevalent among M proteins, followed by the M87 and M2 patterns, while the M68 pattern was rare. These patterns, except for M68, were also evident in numerous M-like Enn proteins. Binding of C4BP via these patterns to Enn proteins was verified. We conclude that C4BP-binding patterns occur frequently in Strep A strains of differing M types, being present in their M or Enn proteins, or frequently both, providing further impetus for their use as vaccine immunogens.
- Published
- 2024
3. A conserved 3D pattern in a Streptococcus pyogenes M protein immunogen elicits M-type crossreactivity
- Author
-
Wang, Kuei-Chen, Kuliyev, Eziz, Nizet, Victor, and Ghosh, Partho
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,Emerging Infectious Diseases ,Biotechnology ,Prevention ,Immunization ,Clinical Research ,Vaccine Related ,Infection ,Good Health and Well Being ,Humans ,Antigens ,Bacterial ,Bacterial Outer Membrane Proteins ,Carrier Proteins ,Protein Binding ,Streptococcus pyogenes ,Cross Reactions ,C4BP ,M protein ,crossreactivity ,immunogen ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Coiled coil-forming M proteins of the widespread and potentially deadly bacterial pathogen Streptococcus pyogenes (strep A) are immunodominant targets of opsonizing antibodies. However, antigenic sequence variability of M proteins into >220 M types, as defined by their hypervariable regions (HVRs), is considered to limit M proteins as vaccine immunogens because of type specificity in the antibody response. Surprisingly, a multi-HVR immunogen in clinical vaccine trials was shown to elicit M-type crossreactivity. The basis for this crossreactivity is unknown but may be due in part to antibody recognition of a 3D pattern conserved in many M protein HVRs that confers binding to human complement C4b-binding protein (C4BP). To test this hypothesis, we investigated whether a single M protein immunogen carrying the 3D pattern would elicit crossreactivity against other M types carrying the 3D pattern. We found that a 34-amino acid sequence of S. pyogenes M2 protein bearing the 3D pattern retained full C4BP-binding capacity when fused to a coiled coil-stabilizing sequence from the protein GCN4. We show that this immunogen, called M2G, elicited cross-reactive antibodies against a number of M types that carry the 3D pattern but not against those that lack the 3D pattern. We further show that the M2G antiserum-recognized M proteins displayed natively on the strep A surface and promoted the opsonophagocytic killing of strep A strains expressing these M proteins. As C4BP binding is a conserved virulence trait of strep A, we propose that targeting the 3D pattern may prove advantageous in vaccine design.
- Published
- 2023
4. C4b-binding protein inhibits particulate- and crystalline-induced NLRP3 inflammasome activation.
- Author
-
Bierschenk, Damien, Papac-Milicevic, Nikolina, Bresch, Ian P., Kovacic, Valentina, Bettoni, Serena, Dziedzic, Mateusz, Wetsel, Rick A., Eschenburg, Susanne, Binder, Christoph J., Blom, Anna M., and King, Ben C.
- Subjects
URATES ,NLRP3 protein ,INFLAMMASOMES ,BLOOD proteins ,COMPLEMENT inhibition ,ADAPTOR proteins - Abstract
Dysregulated NLRP3 inflammasome activation drives a wide variety of diseases, while endogenous inhibition of this pathway is poorly characterised. The serum protein C4b-binding protein (C4BP) is a well-established inhibitor of complement with emerging functions as an endogenously expressed inhibitor of the NLRP3 inflammasome signalling pathway. Here, we identified that C4BP purified from human plasma is an inhibitor of crystalline- (monosodium urate, MSU) and particulate-induced (silica) NLRP3 inflammasome activation. Using a C4BP mutant panel, we identified that C4BP bound these particles via specific protein domains located on the C4BP a-chain. Plasma-purified C4BP was internalised into MSU- or silica-stimulated human primary macrophages, and inhibited MSU- or silica-induced inflammasome complex assembly and IL-1b cytokine secretion. While internalised C4BP in MSU or silica-stimulated human macrophages was in close proximity to the inflammasome adaptor protein ASC, C4BP had no direct effect on ASC polymerisation in in vitro assays. C4BP was also protective against MSU- and silica-induced lysosomal membrane damage. We further provide evidence for an anti-inflammatory function for C4BP in vivo, as C4bp-/- mice showed an elevated pro-inflammatory state following intraperitoneal delivery of MSU. Therefore, internalised C4BP is an inhibitor of crystal- or particle-induced inflammasome responses in human primary macrophages, while murine C4BP protects against an enhanced inflammatory state in vivo. Our data suggests C4BP has important functions in retaining tissue homeostasis in both human and mice as an endogenous serum inhibitor of particulate-stimulated inflammasome activation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Complement Activation-Independent Attenuation of SARS-CoV-2 Infection by C1q and C4b-Binding Protein.
- Author
-
Beirag, Nazar, Varghese, Praveen M., Neto, Martin Mayora, Al Aiyan, Ahmad, Khan, Haseeb A., Qablan, Moneeb, Shamji, Mohamed H., Sim, Robert B., Temperton, Nigel, and Kishore, Uday
- Subjects
- *
COMPLEMENT receptors , *SARS-CoV-2 , *MEMBRANE proteins , *VIRUS diseases , *COMPLEMENT activation , *CYTOKINE release syndrome - Abstract
The complement system is a key component of the innate immune response to viruses and proinflammatory events. Exaggerated complement activation has been attributed to the induction of a cytokine storm in severe SARS-CoV-2 infection. However, there is also an argument for the protective role of complement proteins, given their local synthesis or activation at the site of viral infection. This study investigated the complement activation-independent role of C1q and C4b-binding protein (C4BP) against SARS-CoV-2 infection. The interactions of C1q, its recombinant globular heads, and C4BP with the SARS-CoV-2 spike and receptor binding domain (RBD) were examined using direct ELISA. In addition, RT-qPCR was used to evaluate the modulatory effect of these complement proteins on the SARS-CoV-2-mediated immune response. Cell binding and luciferase-based viral entry assays were utilised to assess the effects of C1q, its recombinant globular heads, and C4BP on SARS-CoV-2 cell entry. C1q and C4BP bound directly to SARS-CoV-2 pseudotype particles via the RBD domain of the spike protein. C1q via its globular heads and C4BP were found to reduce binding as well as viral transduction of SARS-CoV-2 spike protein expressing lentiviral pseudotypes into transfected A549 cells expressing human ACE2 and TMPRSS2. Furthermore, the treatment of the SARS-CoV-2 spike, envelope, nucleoprotein, and membrane protein expressing alphaviral pseudotypes with C1q, its recombinant globular heads, or C4BP triggered a reduction in mRNA levels of proinflammatory cytokines and chemokines such as IL-1β, IL-8, IL-6, TNF-α, IFN-α, and RANTES (as well as NF-κB) in A549 cells expressing human ACE2 and TMPRSS2. In addition, C1q and C4BP treatment also reduced SARS-CoV-2 pseudotype infection-mediated NF-κB activation in A549 cells expressing human ACE2 and TMPRSS2. C1q and C4BP are synthesised primarily by hepatocytes; however, they are also produced by macrophages, and alveolar type II cells, respectively, locally at the pulmonary site. These findings support the notion that the locally produced C1q and C4BP can be protective against SARS-CoV-2 infection in a complement activation-independent manner, offering immune resistance by inhibiting virus binding to target host cells and attenuating the infection-associated inflammatory response. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. C4b-binding protein inhibits particulate- and crystalline-induced NLRP3 inflammasome activation
- Author
-
Damien Bierschenk, Nikolina Papac-Milicevic, Ian P. Bresch, Valentina Kovacic, Serena Bettoni, Mateusz Dziedzic, Rick A. Wetsel, Susanne Eschenburg, Christoph J. Binder, Anna M. Blom, and Ben C. King
- Subjects
inflammasome ,C4BP ,cytokine ,pyroptosis ,gout ,MSU ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Dysregulated NLRP3 inflammasome activation drives a wide variety of diseases, while endogenous inhibition of this pathway is poorly characterised. The serum protein C4b-binding protein (C4BP) is a well-established inhibitor of complement with emerging functions as an endogenously expressed inhibitor of the NLRP3 inflammasome signalling pathway. Here, we identified that C4BP purified from human plasma is an inhibitor of crystalline- (monosodium urate, MSU) and particulate-induced (silica) NLRP3 inflammasome activation. Using a C4BP mutant panel, we identified that C4BP bound these particles via specific protein domains located on the C4BP α-chain. Plasma-purified C4BP was internalised into MSU- or silica-stimulated human primary macrophages, and inhibited MSU- or silica-induced inflammasome complex assembly and IL-1β cytokine secretion. While internalised C4BP in MSU or silica-stimulated human macrophages was in close proximity to the inflammasome adaptor protein ASC, C4BP had no direct effect on ASC polymerisation in in vitro assays. C4BP was also protective against MSU- and silica-induced lysosomal membrane damage. We further provide evidence for an anti-inflammatory function for C4BP in vivo, as C4bp-/- mice showed an elevated pro-inflammatory state following intraperitoneal delivery of MSU. Therefore, internalised C4BP is an inhibitor of crystal- or particle-induced inflammasome responses in human primary macrophages, while murine C4BP protects against an enhanced inflammatory state in vivo. Our data suggests C4BP has important functions in retaining tissue homeostasis in both human and mice as an endogenous serum inhibitor of particulate-stimulated inflammasome activation.
- Published
- 2023
- Full Text
- View/download PDF
7. Complement in metabolic disease: metaflammation and a two-edged sword.
- Author
-
King, B. C. and Blom, A. M.
- Subjects
- *
METABOLIC disorders , *COMPLEMENT activation , *WESTERN diet , *OBESITY , *SEDENTARY lifestyles - Abstract
We are currently experiencing an enduring global epidemic of obesity and diabetes. It is now understood that chronic low-grade tissue inflammation plays an important role in metabolic disease, brought upon by increased uptake of a so-called Western diet, and a more sedentary lifestyle. Many evolutionarily conserved links exist between metabolism and the immune system, and an imbalance in this system induced by chronic over-nutrition has been termed 'metaflammation'. The complement system is an important and evolutionarily ancient part of innate immunity, but recent work has revealed that complement not only is involved in the recognition of pathogens and induction of inflammation, but also plays important roles in cellular and tissue homeostasis. Complement can therefore contribute both positively and negatively to metabolic control, depending on the nature and anatomical site of its activity. This review will therefore focus on the interactions of complement with mechanisms and tissues relevant for metabolic control, obesity and diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
8. FHR4‐based immunoconjugates direct complement‐dependent cytotoxicity and phagocytosis towards HER2‐positive cancer cells
- Author
-
Carole Seguin‐Devaux, Jean‐Marc Plesseria, Charlène Verschueren, Cécile Masquelier, Gilles Iserentant, Marie Fullana, Mihály Józsi, Jacques H. M. Cohen, and Xavier Dervillez
- Subjects
C4bp ,CDC ,complement resistance ,FHR4 ,MAC ,multimers ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Directing selective complement activation towards tumour cells is an attractive strategy to promote their elimination. In the present work, we have generated heteromultimeric immunoconjugates that selectively activate the complement alternative pathway (AP) on tumour cells. We used the C4b‐binding protein C‐terminal‐α‐/β‐chain scaffold for multimerisation to generate heteromultimeric immunoconjugates displaying (a) a multivalent‐positive regulator of the AP, the human factor H‐related protein 4 (FHR4) with; (b) a multivalent targeting function directed against erbB2 (HER2); and (c) a monovalent enhanced GFP tracking function. Two distinct VHH targeting two different epitopes against HER2 and competing either with trastuzumab or with pertuzumab‐recognising epitopes [VHH(T) or VHH(P)], respectively, were used as HER2 anchoring moieties. Optimised high‐FHR4 valence heteromultimeric immunoconjugates [FHR4/VHH(T) or FHR4/VHH(P)] were selected by sequential cell cloning and a selective multistep His‐Trap purification. Optimised FHR4‐heteromultimeric immunoconjugates successfully overcame FH‐mediated complement inhibition threshold, causing increased C3b deposition on SK‐OV‐3, BT474 and SK‐BR3 tumour cells, and increased formation of lytic membrane attack complex densities and complement‐dependent cytotoxicity (CDC). CDC varies according to the pattern expression and densities of membrane‐anchored complement regulatory proteins on tumour cell surfaces. In addition, opsonised BT474 tumour cells were efficiently phagocytosed by macrophages through complement‐dependent cell‐mediated cytotoxicity. We showed that the degree of FHR4‐multivalency within the multimeric immunoconjugates was the key element to efficiently compete and deregulate FH and FH‐mediated convertase decay locally on tumour cell surface. FHR4 can thus represent a novel therapeutic molecule, when expressed as a multimeric entity and associated with an anchoring system, to locally shift the complement steady‐state towards activation on tumour cell surface.
- Published
- 2019
- Full Text
- View/download PDF
9. A Protective and Pathogenic Role for Complement During Acute Toxoplasma gondii Infection
- Author
-
Patricia M. Sikorski, Alessandra G. Commodaro, and Michael E. Grigg
- Subjects
complement ,Toxoplasma gondii ,C4BP ,factor H ,regulation ,immune evasion ,Microbiology ,QR1-502 - Abstract
The infection competence of the protozoan pathogen Toxoplasma gondii is critically dependent on the parasite’s ability to inactivate the host complement system. Toxoplasma actively resists complement-mediated killing in non-immune serum by recruiting host-derived complement regulatory proteins C4BP and Factor H (FH) to the parasite surface to inactivate surface-bound C3 and limit formation of the C5b-9 membrane attack complex (MAC). While decreased complement activation on the parasite surface certainly protects Toxoplasma from immediate lysis, the biological effector functions of C3 split products C3b and C3a are maintained, which includes opsonization of the parasite for phagocytosis and potent immunomodulatory effects that promote pro-inflammatory responses and alters mucosal defenses during infection, respectively. In this review, we discuss how complement regulation by Toxoplasma controls parasite burden systemically but drives exacerbated immune responses locally in the gut of genetically susceptible C57BL/6J mice. In effect, Toxoplasma has evolved to strike a balance with the complement system, by inactivating complement to protect the parasite from immediate serum killing, it generates sufficient C3 catabolites that signal through their cognate receptors to stimulate protective immunity. This regulation ultimately controls tachyzoite proliferation and promotes host survival, parasite persistence, and transmissibility to new hosts.
- Published
- 2021
- Full Text
- View/download PDF
10. C4b Binding Protein Acts as an Innate Immune Effector Against Influenza A Virus
- Author
-
Praveen M. Varghese, Valarmathy Murugaiah, Nazar Beirag, Nigel Temperton, Haseeb A. Khan, Salman H. Alrokayan, Mohammed N. Al-Ahdal, Beatrice Nal, Futwan A. Al-Mohanna, Robert B. Sim, and Uday Kishore
- Subjects
complement ,C4BP ,influenza A virus ,inflammation ,pseudo-typed lentiviral particles ,Immunologic diseases. Allergy ,RC581-607 - Abstract
C4b Binding Protein (C4BP) is a major fluid phase inhibitor of the classical and lectin pathways of the complement system. Complement inhibition is achieved by binding to and restricting the role of activated complement component C4b. C4BP functions as a co-factor for factor I in proteolytic inactivation of both soluble and cell surface-bound C4b, thus restricting the formation of the C3-convertase, C4b2a. C4BP also accelerates the natural decay/dissociation of the C3 convertase. This makes C4BP a prime target for exploitation by pathogens to escape complement attack, as seen in Streptococcus pyogenes or Flavivirus. Here, we examined whether C4BP can act on its own in a complement independent manner, against pathogens. C4BP bound H1N1 and H3N2 subtypes of Influenza A Virus (IAV) most likely via multiple sites in Complement Control Protein (CCP) 1-2, 4-5, and 7-8 domains of its α-chain. In addition, C4BP CCP1-2 bound H3N2 better than H1N1. C4BP bound three IAV envelope proteins: Haemagglutinin (~70 kDa), Neuraminidase (~55 kDa), and Matrix protein 1 (~25kDa). C4BP suppressed H1N1 subtype infection into the lung epithelial cell line, A549, while it promoted infection by H3N2 subtype. C4BP restricted viral entry for H1N1 but had the opposite effect on H3N2, as evident from experiments using pseudo-typed viral particles. C4BP downregulated mRNA levels of pro-inflammatory IFN-α, IL-12, and NFκB in the case of H1N1, while it promoted a pro-inflammatory immune response by upregulating IFN- α, TNF-α, RANTES, and IL-6 in the case of H3N2. We conclude that C4BP differentially modulates the efficacy of IAV entry, and hence, replication in a target cell in a strain-dependent manner, and acts as an entry inhibitor for H1N1. Thus, CCP containing complement proteins such as factor H and C4BP may have additional defense roles against IAV that do not rely on the regulation of complement activation.
- Published
- 2021
- Full Text
- View/download PDF
11. A Protective and Pathogenic Role for Complement During Acute Toxoplasma gondii Infection.
- Author
-
Sikorski, Patricia M., Commodaro, Alessandra G., and Grigg, Michael E.
- Subjects
TOXOPLASMA gondii ,LABORATORY mice ,COMPLEMENT activation ,PHAGOCYTOSIS ,PROTOZOAN diseases ,TOXOPLASMA ,IMMUNE response - Abstract
The infection competence of the protozoan pathogen Toxoplasma gondii is critically dependent on the parasite's ability to inactivate the host complement system. Toxoplasma actively resists complement-mediated killing in non-immune serum by recruiting host-derived complement regulatory proteins C4BP and Factor H (FH) to the parasite surface to inactivate surface-bound C3 and limit formation of the C5b-9 membrane attack complex (MAC). While decreased complement activation on the parasite surface certainly protects Toxoplasma from immediate lysis, the biological effector functions of C3 split products C3b and C3a are maintained, which includes opsonization of the parasite for phagocytosis and potent immunomodulatory effects that promote pro-inflammatory responses and alters mucosal defenses during infection, respectively. In this review, we discuss how complement regulation by Toxoplasma controls parasite burden systemically but drives exacerbated immune responses locally in the gut of genetically susceptible C57BL/6J mice. In effect, Toxoplasma has evolved to strike a balance with the complement system, by inactivating complement to protect the parasite from immediate serum killing, it generates sufficient C3 catabolites that signal through their cognate receptors to stimulate protective immunity. This regulation ultimately controls tachyzoite proliferation and promotes host survival, parasite persistence, and transmissibility to new hosts. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. C4b Binding Protein Acts as an Innate Immune Effector Against Influenza A Virus.
- Author
-
Varghese, Praveen M., Murugaiah, Valarmathy, Beirag, Nazar, Temperton, Nigel, Khan, Haseeb A., Alrokayan, Salman H., Al-Ahdal, Mohammed N., Nal, Beatrice, Al-Mohanna, Futwan A., Sim, Robert B., and Kishore, Uday
- Subjects
CARRIER proteins ,INFLUENZA A virus ,INFLUENZA A virus, H1N1 subtype ,ECULIZUMAB ,INFLUENZA A virus, H3N2 subtype ,EXTRACELLULAR matrix proteins - Abstract
C4b Binding Protein (C4BP) is a major fluid phase inhibitor of the classical and lectin pathways of the complement system. Complement inhibition is achieved by binding to and restricting the role of activated complement component C4b. C4BP functions as a co-factor for factor I in proteolytic inactivation of both soluble and cell surface-bound C4b, thus restricting the formation of the C3-convertase, C4b2a. C4BP also accelerates the natural decay/dissociation of the C3 convertase. This makes C4BP a prime target for exploitation by pathogens to escape complement attack, as seen in Streptococcus pyogenes or Flavivirus. Here, we examined whether C4BP can act on its own in a complement independent manner, against pathogens. C4BP bound H1N1 and H3N2 subtypes of Influenza A Virus (IAV) most likely via multiple sites in Complement Control Protein (CCP) 1-2, 4-5, and 7-8 domains of its α-chain. In addition, C4BP CCP1-2 bound H3N2 better than H1N1. C4BP bound three IAV envelope proteins: Haemagglutinin (~70 kDa), Neuraminidase (~55 kDa), and Matrix protein 1 (~25kDa). C4BP suppressed H1N1 subtype infection into the lung epithelial cell line, A549, while it promoted infection by H3N2 subtype. C4BP restricted viral entry for H1N1 but had the opposite effect on H3N2, as evident from experiments using pseudo-typed viral particles. C4BP downregulated mRNA levels of pro-inflammatory IFN-α, IL-12, and NFκB in the case of H1N1, while it promoted a pro-inflammatory immune response by upregulating IFN- α, TNF-α, RANTES, and IL-6 in the case of H3N2. We conclude that C4BP differentially modulates the efficacy of IAV entry, and hence, replication in a target cell in a strain-dependent manner, and acts as an entry inhibitor for H1N1. Thus, CCP containing complement proteins such as factor H and C4BP may have additional defense roles against IAV that do not rely on the regulation of complement activation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
13. FHR4‐based immunoconjugates direct complement‐dependent cytotoxicity and phagocytosis towards HER2‐positive cancer cells.
- Author
-
Seguin‐Devaux, Carole, Plesseria, Jean‐Marc, Verschueren, Charlène, Masquelier, Cécile, Iserentant, Gilles, Fullana, Marie, Józsi, Mihály, Cohen, Jacques H. M., and Dervillez, Xavier
- Abstract
Directing selective complement activation towards tumour cells is an attractive strategy to promote their elimination. In the present work, we have generated heteromultimeric immunoconjugates that selectively activate the complement alternative pathway (AP) on tumour cells. We used the C4b‐binding protein C‐terminal‐α‐/β‐chain scaffold for multimerisation to generate heteromultimeric immunoconjugates displaying (a) a multivalent‐positive regulator of the AP, the human factor H‐related protein 4 (FHR4) with; (b) a multivalent targeting function directed against erbB2 (HER2); and (c) a monovalent enhanced GFP tracking function. Two distinct VHH targeting two different epitopes against HER2 and competing either with trastuzumab or with pertuzumab‐recognising epitopes [VHH(T) or VHH(P)], respectively, were used as HER2 anchoring moieties. Optimised high‐FHR4 valence heteromultimeric immunoconjugates [FHR4/VHH(T) or FHR4/VHH(P)] were selected by sequential cell cloning and a selective multistep His‐Trap purification. Optimised FHR4‐heteromultimeric immunoconjugates successfully overcame FH‐mediated complement inhibition threshold, causing increased C3b deposition on SK‐OV‐3, BT474 and SK‐BR3 tumour cells, and increased formation of lytic membrane attack complex densities and complement‐dependent cytotoxicity (CDC). CDC varies according to the pattern expression and densities of membrane‐anchored complement regulatory proteins on tumour cell surfaces. In addition, opsonised BT474 tumour cells were efficiently phagocytosed by macrophages through complement‐dependent cell‐mediated cytotoxicity. We showed that the degree of FHR4‐multivalency within the multimeric immunoconjugates was the key element to efficiently compete and deregulate FH and FH‐mediated convertase decay locally on tumour cell surface. FHR4 can thus represent a novel therapeutic molecule, when expressed as a multimeric entity and associated with an anchoring system, to locally shift the complement steady‐state towards activation on tumour cell surface. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
14. The complement-independent role of C4b-binding protein in Neisseria gonorrhoeae pathogenesis
- Subjects
gonorrhea ,neutrophil ,phagocytosis ,complement ,C4BP - Abstract
Neisseria gonorrhoeae (Gc) is the human-specific pathogen that causes the sexually transmitted infection gonorrhea. Gonorrhea is a significant public health concern with no vaccine, increasing antibiotic resistance, and recurrent infections due to a lack of protective immunity. The hallmark immune response to Gc is an abundant neutrophilic influx. However, these neutrophils are unable to clear the infection, and viable, Opacity-associated (Opa) protein-expressing Gc (Opa+ Gc) are recovered in exudates from infected patients. Although Opa proteins promote colonization, they also engage carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) on the neutrophil surface, which can result in neutrophil activation and antigonococcal activities. How these two opposing outcomes are balanced to favor Opa expression during infection remains enigmatic. In addition to neutrophils, serum is present in inflamed mucosal secretions. In this research, I unexpectedly found that normal human serum enhanced Opa+ Gc survival from primary human neutrophils from healthy donors. I hypothesized that soluble factor(s) in serum were affecting Gc-neutrophil interactions, and sought to determine the identity of the serum factor(s) responsible for enhanced Gc survival from neutrophils. Using mass spectrometry, I identified C4b-binding protein (C4BP), a canonical complement inhibitor protein, as a candidate protein of interest in serum and secretions relevant to gonorrhea. Using C4BP-depleted serum and purified C4BP, I confirmed that C4BP was necessary and sufficient for serum-mediated enhanced survival of Opa+ Gc. Next, I characterized how C4BP modulated Gc-neutrophil interactions. C4BP suppressed Gc-induced neutrophil reactive oxygen species production and inhibited neutrophil association with and phagocytosis of Opa+ Gc. These effects required binding of the multimeric alpha chain form of C4BP to the surface of Gc. Though C4BP is a canonical complement inhibitor protein, the effects of C4BP on Gc-neutrophil interactions were complement-independent. To investigate whether C4BP might be disrupting Opa-CEACAM interactions, I developed a quantitative, imaging flow cytometry method to detect N-CEACAM binding to the surface of Opa+ Gc. Ultimately, using a panel of strains constitutively expressing single Opa proteins with unique CEACAM-binding profiles, I found that the role of C4BP in limiting neutrophil phagocytosis was specific to Gc expressing CEACAM-binding Opa proteins, and did not extend to bacteria that engaged neutrophils in other manners. The results of this work underscore the importance of C4BP to Gc infection, not only as a contributor to serum resistance, but also as a modulator of neutrophil function. In the future, the effect of C4BP during in vivo infections and on therapeutic efficacy should be determined. Additionally, it is not known whether neutrophil signaling downstream of Opa-CEACAM engagement is modulated by C4BP, or if neutrophils process the C4BP bound to the Gc surface. The phenomena described here could extend to the numerous other microbes that bind C4BP or to other complement inhibitor proteins. This research identifies for the first time a complement-independent role for C4BP in enhancing the survival of a pathogenic bacterium from phagocytes, and contributes to our understanding of how Gc persists in neutrophil-rich conditions during infection.
- Published
- 2023
- Full Text
- View/download PDF
15. Chimeric Proteins Containing MAP-1 and Functional Domains of C4b-Binding Protein Reveal Strong Complement Inhibitory Capacities
- Author
-
Cecilie E. Hertz, Rafael Bayarri-Olmos, Nikolaj Kirketerp-Møller, Sander van Putten, Katrine Pilely, Mikkel-Ole Skjoedt, and Peter Garred
- Subjects
complement activation ,lectin pathway ,classical pathway ,MAP-1 ,C4BP ,complement inhibition ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The complement system is a tightly regulated network of proteins involved in defense against pathogens, inflammatory processes, and coordination of the innate and adaptive immune responses. Dysregulation of the complement cascade is associated with many inflammatory disorders. Thus, inhibition of the complement system has emerged as an option for treatment of a range of different inflammatory diseases. MAP-1 is a pattern recognition molecule (PRM)-associated inhibitor of the lectin pathway of the complement system, whereas C4b-binding protein (C4BP) regulates both the classical and lectin pathways. In this study we generated chimeric proteins consisting of MAP-1 and the first five domains of human C4BP (C4BP1−5) in order to develop a targeted inhibitor acting at different levels of the complement cascade. Two different constructs were designed and expressed in CHO cells where MAP-1 was fused with C4BP1−5 in either the C- or N-terminus. The functionality of the chimeric proteins was assessed using different in vitro complement activation assays. Both chimeric proteins displayed the characteristic Ca2+-dependent dimerization and binding to PRMs of native MAP-1, as well as the co-factor activity of native C4BP. In ELISA-based complement activation assays they could effectively inhibit the lectin and classical pathways. Notably, MAP-1:C4BP1−5 was five times more effective than rMAP-1 and rC4BP1−5 applied at the same time, emphasizing the advantage of a single inhibitor containing both functional domains. The MAP-1/C4BP chimeras exert unique complement inhibitory properties and represent a novel therapeutic approach targeting both upstream and central complement activation.
- Published
- 2018
- Full Text
- View/download PDF
16. Gonococcal MtrE and its surface-expressed Loop 2 are immunogenic and elicit bactericidal antibodies.
- Author
-
Wang, Shuyi, Xue, Juan, Lu, Ping, Ni, Chunshan, Cheng, Hao, Han, Rui, and van der Veen, Stijn
- Abstract
Objectives: The rise in multidrug resistant Neisseria gonorrhoeae poses a threat to healthcare, while the development of an effective vaccine has remained elusive due to antigenic and phase variability of surface-expressed proteins. In the current study, we identified a fully conserved surface expressed protein and characterized its suitability as a vaccine antigen.Methods: An in silico approach was used to predict surface-expressed proteins and analyze sequence conservation and phase variability. The most conserved protein and its surface-exposed Loop 2, which was displayed as both a structural and linear epitope on the oligomerization domain of C4b binding protein, were used to immunize mice. Immunogenicity was subsequently analyzed by determination of antibody titers and serum bactericidal activity.Results: MtrE was identified as one of the most conserved surface-expressed proteins. Furthermore, MtrE and both Loop 2-containing fusion proteins elicited high protein-specific antibody titers and particularly the two Loop 2 fusion proteins showed high anti-Loop 2 titers. In addition, antibodies raised against all three proteins were able to recognize MtrE expressed on the surface of N. gonorrhoeae and showed high MtrE-dependent bactericidal activity.Conclusions: Our results show that MtrE and Loop 2 are promising novel conserved surface-expressed antigens for vaccine development against N. gonorrhoeae. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
17. Chimeric Proteins Containing MAP-1 and Functional Domains of C4b-Binding Protein Reveal Strong Complement Inhibitory Capacities.
- Author
-
Hertz, Cecilie E., Bayarri-Olmos, Rafael, Kirketerp-Møller, Nikolaj, van Putten, Sander, Pilely, Katrine, Skjoedt, Mikkel-Ole, and Garred, Peter
- Subjects
CHIMERIC proteins ,COMPLEMENT inhibition ,CARRIER proteins - Abstract
The complement system is a tightly regulated network of proteins involved in defense against pathogens, inflammatory processes, and coordination of the innate and adaptive immune responses. Dysregulation of the complement cascade is associated with many inflammatory disorders. Thus, inhibition of the complement system has emerged as an option for treatment of a range of different inflammatory diseases. MAP-1 is a pattern recognition molecule (PRM)-associated inhibitor of the lectin pathway of the complement system, whereas C4b-binding protein (C4BP) regulates both the classical and lectin pathways. In this study we generated chimeric proteins consisting of MAP-1 and the first five domains of human C4BP (C4BP
1−5 ) in order to develop a targeted inhibitor acting at different levels of the complement cascade. Two different constructs were designed and expressed in CHO cells where MAP-1 was fused with C4BP1−5 in either the C- or N-terminus. The functionality of the chimeric proteins was assessed using different in vitro complement activation assays. Both chimeric proteins displayed the characteristic Ca2+ -dependent dimerization and binding to PRMs of native MAP-1, as well as the co-factor activity of native C4BP. In ELISA-based complement activation assays they could effectively inhibit the lectin and classical pathways. Notably, MAP-1:C4BP1−5 was five times more effective than rMAP-1 and rC4BP1−5 applied at the same time, emphasizing the advantage of a single inhibitor containing both functional domains. The MAP-1/C4BP chimeras exert unique complement inhibitory properties and represent a novel therapeutic approach targeting both upstream and central complement activation. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
18. High protein S activity due to C4b‐binding protein deficiency in a 34‐year‐old Surinamese female with ischemic retinopathy.
- Author
-
Mulder, René, de Vries, Jeroen K., Müskens, Rogier P. H. M., Mulder, André B., and Lukens, Michaël V.
- Subjects
- *
PROTEIN S , *THROMBOSIS risk factors , *GENETIC mutation , *CARRIER proteins , *ANTICOAGULANTS - Abstract
Key Clinical Message: In this study, we present the first case of a 34‐year‐old Surinamese female with ischemic retinopathy and increased free protein S due to C4BP deficiency. Possibly, the low PS/C4BP complex level has increased the risk of arterial thrombosis in our patient. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
19. Vitamin K-Dependent Protein S: Beyond the Protein C Pathway.
- Author
-
Dahlbäck, Björn
- Subjects
- *
VITAMIN K , *PROTEIN C , *PROTEIN S deficiency , *THROMBOSIS risk factors , *LABORATORY mice - Abstract
Protein S is a vitamin K-dependent plasma glycoprotein circulating in plasma at a concentration of around 350 nM. Approximately 60% of protein S in human plasma is bound to the complement regulatory protein C4b-binding protein (C4BP) in a highaffinity, high-molecular-weight complex. Protein S in plasma has multiple anticoagulant properties and heterozygous protein S deficiency is associated with increased risk of venous thrombosis. Homozygous deficiency in man and mice is associated with severe thrombosis in fetal life, defects in the vascular system development, and not compatible with life. Protein S has additional functions beyond being an anticoagulant. It affects the complement regulatory properties of C4BP, and moreover, protein S interacts with tyrosine kinase receptors of the TAM family, which comprises Tyro3, Axl, and Mer. The TAM receptor interaction is important for the ability of protein S to stimulate phagocytosis of apoptotic cells. This review will discuss the multiple functions of protein S, describing its role as cofactor to activated protein C with a subsequent focus on the other functions of protein S. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
20. A novel C3d-containing oligomeric vaccine provides insight into the viability of testing human C3d-based vaccines in mice.
- Author
-
He, Yong-Gang, Pappworth, Isabel Y., Rossbach, Andreas, Paulin, Joshua, Mavimba, Tarirai, Hayes, Christine, Kulik, Liudmila, Holers, V.Michael, Knight, Andrew M., and Marchbank, Kevin J.
- Subjects
- *
VACCINES , *CARRIER proteins , *TETANUS toxin , *COMPLEMENT receptors , *IMMUNOLOGICAL adjuvants - Abstract
The use of C3d, the final degradation product of complement protein C3, as a “natural” adjuvant has been widely examined since the initial documentation of its immunogenicity-enhancing properties as a consequence of binding to complement receptor 2. Subsequently it was demonstrated that these effects are most evident when oligomeric, rather than when monomeric forms of C3d, are linked to various test protein antigens. In this study, we examined the feasibility of enhancing the adjuvant properties of human C3d further by utilizing C4b-binding protein (C4BP) to provide an oligomeric arrayed scaffold fused to the model antigen, tetanus toxin C fragment (TTCF). High molecular weight, C3d-containing oligomeric vaccines were successfully expressed, purified from mammalian cells and used to immunize groups of mice. Surprisingly, anti-TTCF antibody responses measured in these mice were poor. Subsequently we established by in vitro and in vivo analysis that, in the presence of mouse C3, human C3d does not interact with either mouse or even human complement receptor 2. These data confirm the requirement to develop murine versions of C3d based adjuvant compounds to test in mice or that mice would need to be developed that express both human C3 and human CR2 to allow the testing of human C3d based adjuvants in mouse in any capacity. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
21. The human serum protein C4b-binding protein inhibits pancreatic IAPP-induced inflammasome activation.
- Author
-
Kulak, Klaudia, Westermark, Gunilla, Papac-Milicevic, Nikolina, Renström, Erik, Blom, Anna, and King, Ben
- Abstract
Aims/hypothesis: Inflammasome activation and subsequent IL-1β production is a driver of islet pathology in type 2 diabetes. Oligomers, but not mature amyloid fibrils, of human islet amyloid polypeptide (IAPP), which is co-secreted with insulin, trigger NOD-like receptor pyrin domain containing-3 (NLRP3) inflammasome activation. C4b-binding protein (C4BP), present in serum, binds to IAPP and affects transition of IAPP monomers and oligomers to amyloid fibrils. We therefore hypothesised that C4BP inhibits IAPP-mediated inflammasome activation and IL-1β production. Methods: Macrophages were exposed to IAPP in the presence or absence of plasma-purified human C4BP, and inflammasome activation was assessed by IL-1β secretion as detected by ELISA and reporter cell lines. IAPP fibrillation was assessed by thioflavin T assay. Uptake of IAPP-C4BP complexes and their effects on phagolysosomal stability were assessed by flow cytometry and confocal microscopy. The effect of C4BP regulation of IAPP-mediated inflammasome activation on beta cell function was assessed using a clonal rat beta cell line. Immunohistochemistry was used to examine the association of IAPP amyloid deposits and macrophage infiltration in isolated human and mouse pancreatic islets, and expression of C4BP from isolated human pancreatic islets was assessed by quantitative PCR, immunohistochemistry and western blot. Results: C4BP significantly inhibited IAPP-mediated IL-1β secretion from primed macrophages at physiological concentrations in a dose-dependent manner. C4BP bound to and was internalised together with IAPP. C4BP did not affect IAPP uptake into phagolysosomal compartments, although it did inhibit its formation into amyloid fibrils. The loss of macrophage phagolysosomal integrity induced by IAPP incubation was inhibited by co-incubation with C4BP. Supernatant fractions from macrophages activated with IAPP inhibited both insulin secretion and viability of clonal beta cells in an IL-1β-dependent manner but the presence of C4BP during macrophage IAPP incubation rescued beta cell function and viability. In human and mouse islets, the presence of amyloid deposits correlated with higher numbers of infiltrating macrophages. Isolated human islets expressed and secreted C4BP, which increased with addition of IL-1β. Conclusions/interpretation: IAPP deposition is associated with inflammatory cell infiltrates in pancreatic islets. C4BP blocks IAPP-induced inflammasome activation by preventing the loss of macrophage phagolysosomal integrity required for NLRP3 activation. The consequence of this is the preservation of beta cell function and viability. C4BP is secreted directly from human pancreatic islets and this increases in response to inflammatory cytokines. We therefore propose that C4BP acts as an extracellular chaperone protein that limits the proinflammatory effects of IAPP. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
22. Non-traditional roles of complement in type 2 diabetes: Metabolism, insulin secretion and homeostasis.
- Author
-
King, Ben C. and Blom, Anna M.
- Subjects
- *
TYPE 2 diabetes , *COMPLEMENT (Immunology) , *HOMEOSTASIS , *INSULIN resistance , *HYPERGLYCEMIA , *IMMUNOLOGY of inflammation , *ISLANDS of Langerhans , *PHYSIOLOGY - Abstract
Type 2 Diabetes (T2D) is a disease of increasing importance and represents a growing burden on global healthcare and human health. In T2D, loss of effectiveness of insulin signaling in peripheral tissues cannot be compensated for by adequate insulin secretion, leading to hyperglycemia and resultant complications. In recent years, inflammation has been identified as a central component of T2D, both in inducing peripheral insulin resistance as well as in the pancreatic islet, where it contributes to loss of insulin secretion and death of insulin-secreting beta cells. In this review we will focus on non-traditional roles of complement proteins which have been identified in T2D-associated inflammation, beta cell secretory function, and in maintaining homeostasis of the pancreatic islet. Improved understanding of both traditional and novel roles of complement proteins in T2D may lead to new therapeutic approaches for this global disease. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
23. Acquisition of negative complement regulators by the saprophyte Leptospira biflexa expressing LigA or LigB confers enhanced survival in human serum.
- Author
-
Castiblanco-Valencia, Mónica M., Fraga, Tatiana R., Breda, Leandro C.D., Vasconcellos, Sílvio A., Figueira, Cláudio P., Picardeau, Mathieu, Wunder, Elsio, Ko, Albert I., Barbosa, Angela S., and Isaac, Lourdes
- Subjects
- *
SAPROPHYTES , *LEPTOSPIRA , *IMMUNOGLOBULINS , *SERUM , *CARRIER proteins , *PROTEIN-protein interactions , *GENE expression - Abstract
Leptospiral immunoglobulin-like (Lig) proteins are surface exposed molecules present in pathogenic but not in saprophytic Leptospira species. We have previously shown that Lig proteins interact with the soluble complement regulators Factor H (FH), FH like-1 (FHL-1), FH related-1 (FHR-1) and C4b Binding Protein (C4BP). In this study, we used the saprophyte L. biflexa serovar Patoc as a surrogate host to address the specific role of LigA and LigB proteins in leptospiral complement evasion. L. biflexa expressing LigA or LigB was able to acquire FH and C4BP. Bound complement regulators retained their cofactor activities of FI in the proteolytic cleavage of C3b and C4b. Moreover, heterologous expression of ligA and ligB genes in the saprophyte L. biflexa enhanced bacterial survival in human serum. Complement deposition on lig -transformed L. biflexa was assessed by flow cytometry analysis. With regard to MAC deposition, L. biflexa expressing LigA or LigB presented an intermediate profile: MAC deposition levels were greater than those found in the pathogenic L. interrogans , but lower than those observed for L. biflexa wildtype. In conclusion, Lig proteins contribute to in vitro control of complement activation on the leptospiral surface, promoting an increased bacterial survival in human serum. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
24. Ein Apoptose-induzierendes Heptamer, das effizient den Todesrezeptor 5 bündelt.
- Author
-
Valldorf, Bernhard, Fittler, Heiko, Deweid, Lukas, Ebenig, Aileen, Dickgiesser, Stephan, Sellmann, Carolin, Becker, Janine, Zielonka, Stefan, Empting, Martin, Avrutina, Olga, and Kolmar, Harald
- Abstract
Multivalente Liganden von Todesrezeptoren sind vielversprechende tumorspezifische Wirkstoffe, da sie eine apoptotische Kaskade in Krebszellen induzieren. Hier beschreiben wir einen modularen Ansatz für den Aufbau von Konstrukten, die an den Todesrezeptor 5 (“death receptor 5”, DR5) binden. In diesen Konstrukten sind mehrere Kopien gegen DR5 gerichteter Peptide (DR5TP) kovalent mit biomolekularen Gerüsten verknüpft. Mit dieser Strategie gelingt eine effiziente Oligomerisierung der Peptide in verschiedenen räumlichen Orientierungen durch enzymvermittelte Konjugationen oder rekombinante Produktion. Heptamere Konstrukte mit einem kurzen Gerüst (60 – 75 Reste) der C ‐ terminalen Oligomerisierungdomäne des humanen C4b ‐ Bindeproteins zeigten eine bemerkenswert hohe proapoptotische Aktivität (EC50=3 nm), wenn DR5TP an den Carboxyterminus ligiert wurde. Unsere Daten stützen die Ansicht, dass der Interligandabstand, die relative räumliche Orientierung und die Kopienzahl der rezeptorbindenden Module wesentliche Kriterien für die Rezeptoraktivierung und Zelltötung sind. Gegen den Todesrezeptor 5 (DR5) gerichtete Peptide wurden kovalent mit biomolekularen Gerüsten verknüpft, um multivalente Liganden zu erhalten, die spezifisch eine apoptotische Kaskade in Krebszellen induzieren. Anzahl und räumliche Orientierung der Kopien entscheiden über ihr Vermögen zur Rezeptoraktivierung. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
25. An Apoptosis-Inducing Peptidic Heptad That Efficiently Clusters Death Receptor 5.
- Author
-
Valldorf, Bernhard, Fittler, Heiko, Deweid, Lukas, Ebenig, Aileen, Dickgiesser, Stephan, Sellmann, Carolin, Becker, Janine, Zielonka, Stefan, Empting, Martin, Avrutina, Olga, and Kolmar, Harald
- Subjects
- *
PEPTIDES , *LIGANDS (Chemistry) , *CANCER cell culture , *OLIGOMERIZATION , *CARRIER proteins - Abstract
Multivalent ligands of death receptors hold particular promise as tumor cell-specific therapeutic agents because they induce an apoptotic cascade in cancerous cells. Herein, we present a modular approach to generate death receptor 5 (DR5) binding constructs comprising multiple copies of DR5 targeting peptide (DR5TP) covalently bound to biomolecular scaffolds of peptidic nature. This strategy allows for efficient oligomerization of synthetic DR5TP-derived peptides in different spatial orientations using a set of enzyme-promoted conjugations or recombinant production. Heptameric constructs based on a short (60-75 residues) scaffold of a C-terminal oligomerization domain of human C4b binding protein showed remarkable proapoptotic activity (EC50=3 n m) when DR5TP was ligated to its carboxy terminus. Our data support the notion that inter-ligand distance, relative spatial orientation and copy number of receptor-binding modules are key prerequisites for receptor activation and cell killing. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
26. The recombinant LIC10508 is a plasma fibronectin, plasminogen, fibrinogen and C4BP-binding protein of Leptospira interrogans.
- Author
-
Siqueira, Gabriela H., Teixeira, Aline F., Fernandes, Luis G., de Souza, GiseleO., Kirchgatter, Karin, Romero, Eliete C., Vasconcellos, Silvio A., Vieira, Monica L., and Nascimento, Ana LuciaT.O.
- Subjects
- *
FIBRONECTINS , *PLASMINOGEN , *FIBRINOGEN , *LEPTOSPIRA interrogans , *CARRIER proteins , *BLOOD testing - Abstract
Leptospirosis is a zoonosis caused by pathogenic Leptospira spp. In this study, we report that the recombinant proteins LIC10507, LIC10508 and LIC10509 are recognized by confirmed leptospirosis serum samples at both phases of the disease. The recombinant rLIC10508 and rLIC10507 are plasminogen (PLG)-binding proteins, capable of generating plasmin in the presence of a PLG activator. The proteins bind to PLG in a dose-dependent and saturable manner, fulfilling host-ligand interaction. Furthermore, rLIC10508 interacts with fibrinogen (Fg), plasma fibronectin and C4b binding protein (C4BP). The binding of rLIC10508 to Fg decreases the fibrin clotting in a thrombin-catalyzed reaction. The incubation with 4 μM of protein promoted 40% inhibition upon clotting formation. C4BP bound to rLIC10508 retained its cofactor activity for factor I promoting the cleavage of C4b protein, which may reduce the membrane attack complex formation. Although these proteins have high amino acid sequence similarity, rLIC10508 is the most talented of the three, a behavior that might be explained by its unique putative 3D structure, whereas structures of rLIC10507 and rLIC10509 are very similar. Plasmin generation (rLIC10507 and rLIC10508), together with decreasing fibrin clot formation (rLIC10508) and impairment of the complement system (rLIC10508) may help the bacteria to overcome host defense, facilitating the infection process. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
27. Epididymal C4b-binding protein is processed and degraded during transit through the duct and is not essential for fertility.
- Author
-
Nonaka, Mayumi I., Zsigmond, Eva, Akihiko Kudo, Hayato Kawakami, Kaoru Yoshida, Manabu Yoshida, Natsuko Kawano, Kenji Miyado, Masaru Nonaka, and Wetsel, Rick A.
- Subjects
- *
EPIDIDYMIS diseases , *BIODEGRADATION , *FERTILITY , *CARRIER proteins , *SPERMATOZOA , *LABORATORY mice - Abstract
C4b-binding protein (C4BP) is known as one of the circulating complement regulators that prevents excessive activation of the host-defense complement system. We have reported previously that C4BP is expressed abundantly in the rodent epididymis, one of the male reproductive organs connecting the testis and vas deferens, where immature spermatozoa acquire their motility and fertilizing ability during their transit through the duct. Epididymal C4BP (EpC4BP) is synthesized androgen-dependently by the epithelial cells, secreted into the lumen, and bound to the outer membrane of the passing spermatozoa. In this study, we found that EpC4BP is secreted as a large oligomer, similar to the serum C4BP, but is digested during the epididymal transit and is almost lost from both the luminal fluid and the sperm surface in the vas deferens. Such a processing pattern is not known in serum C4BP, suggesting that EpC4BP and serum C4BP might have different functional mechanisms, and that there is a novel function of EpC4BP in reproduction. In addition, the disappearance of EpC4BP from the sperm surface prior to ejaculation suggests that EpC4BP works only in the epididymis and would not work in the female reproductive tract to protect spermatozoa from complement attack. Next, we generated C4BP-deficient (C4BP-/-) mice to examine the possible role of EpC4BP in reproduction. However, the C4BP-/- mice were fertile and no significant differences were observed between the C4BP-/- and wild-type mouse spermatozoa in terms of morphology, motility, and rate of the spontaneous acrosome reaction. These results suggest that EpC4BP is involved in male reproduction, but not essential for sperm maturation. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
28. Protein sieving characteristics of sub-20-nm pore size filters at varying ionic strength during nanofiltration of Coagulation Factor IX.
- Author
-
Winkler, Clint J., Jorba, Nuria, Shitanishi, Kenneth T., and Herring, Steven W.
- Subjects
- *
MOLECULAR sieves , *PORE size (Materials) , *FILTERS & filtration , *NANOFILTRATION , *IONIC liquids , *BLOOD coagulation factor IX , *THERAPEUTIC use of proteins - Abstract
Abstract: Nanofiltration assures that protein therapeutics are free of adventitious agents such as viruses. Nanofilter pores must allow passage of protein drugs but be small enough to retain viruses. Five nanofilters have been evaluated to identify those that can be used interchangeably to yield a high purity Coagulation Factor IX product. When product preparations prior to nanofiltration were analyzed using electrophoresis, Western blot, liquid chromatography – tandem mass spectrometry and size exclusion HPLC, factor IX, inter – α – trypsin inhibitor and C4b binding protein (C4BP) were observed. C4BP was removed from product by all five nanofilters when nanofiltration was performed at physiological ionic strength. However, at high ionic strength, C4BP was removed by only two nanofilters. HPLC indicated that the Stokes radius of C4BP was larger at low ionic strength than at high ionic strength. The results suggest that C4BP exists in an open conformation at physiological ionic strength and is removed by nanofiltration whereas, at high ionic strength, the protein collapses to an extent that allows passage through some nanofilters. Manufacturers should be aware that protein contaminants in other nanofiltered protein drugs could behave similarly and conditions of nanofiltration must be evaluated to ensure consistent product purity. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
29. Lsa30, a novel adhesin of Leptospira interrogans binds human plasminogen and the complement regulator C4bp
- Author
-
Souza, Natalie M., Vieira, Monica L., Alves, Ivy J., de Morais, Zenaide M., Vasconcellos, Silvio A., and Nascimento, Ana L.T.O.
- Subjects
- *
BACTERIAL adhesins , *LEPTOSPIROSIS , *LEPTOSPIRA interrogans , *PLASMINOGEN , *GENETIC regulation , *ETIOLOGY of diseases , *BACTERIAL proteins , *BACTERIA - Abstract
Abstract: Pathogenic Leptospira is the etiological agent of leptospirosis, a life-threatening disease that affects populations worldwide. Surface proteins have the potential to promote several activities, including adhesion. This work aimed to study the leptospiral coding sequence (CDS) LIC11087, genome annotated as hypothetical outer membrane protein. The LIC11087 gene was cloned and expressed in Escherichia coli BL21 (DE3) strain by using the expression vector pAE. The recombinant protein tagged with N-terminal 6XHis was purified by metal-charged chromatography and characterized by circular dichroism (CD) spectroscopy. The recombinant protein has the ability to mediate attachment to the extracellular matrix (ECM) components, laminin and plasma fibronectin, and was named Lsa30 (Leptospiral surface adhesin of 30 kDa). Lsa30 binds to laminin and to plasma fibronectin in a dose-dependent and saturable manner, with dissociation equilibrium constants (K D ) of 292 ± 24 nm and 157 ± 35 nm, respectively. Moreover, the Lsa30 is a plasminogen (PLG) receptor, capable of generating plasmin, in the presence of activator. This protein may interfere with the complement cascade by interacting with C4bp regulator. The Lsa30 is probably a new surface protein of Leptospira as revealed by immunofluorescence assays with living organisms and the reactivity with antibodies present in serum samples of experimentally infected hamsters. Thus, Lsa30 is a novel versatile protein that may play a role in mediating adhesion and may help pathogenic Leptospira to overcome tissue barriers and to escape the immune system. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
30. C4b-binding protein in Alzheimer's disease: Binding to Aβ1–42 and to dead cells
- Author
-
Trouw, Leendert. A., Nielsen, Henrietta M., Minthon, Lennart, Londos, Elisabet, Landberg, Göran, Veerhuis, Robert, Janciauskiene, Sabina, and Blom, Anna M.
- Subjects
- *
HUNTINGTON disease , *GENETIC disorders , *CEREBROSPINAL fluid , *CARRIER proteins - Abstract
Abstract: In the Alzheimer''s disease (AD) brain, binding of Clq within the Cl complex, the initiating molecule of the classical complement pathway, to apoptotic cells, DNA and amyloid-β (Aβ), the major constituent of senile plaques, can initiate complement activation. However, the extent of activation is determined by the balance between activation and inhibition. Fluid-phase complement inhibitor C4b-binding protein (C4BP) was immunohistochemically detected in Aβ plaques and on apoptotic cells in AD brain. In vitro, C4BP bound apoptotic and necrotic but not viable brain cells (astrocytes, neurons and oligodendrocytes) and limited complement activation on dead brain cells. C4BP also bound Aβ1–42 peptide directly, via the C4BP α-chain, and limited the extent of complement activation by Aβ. C4BP levels in cerebrospinal fluid (CSF) of dementia patients and controls were low compared to levels in plasma and correlated with CSF levels of other inflammation-related factors. In conclusion, C4BP binds to dead brain cells and Aβ peptide in vitro, is present in CSF and possibly protects against excessive complement activation in AD brains. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
31. Binding of complement regulators factor H and C4b binding protein to group A streptococcal strains isolated from tonsillar tissue and blood
- Author
-
Suvilehto, Jari, Jarva, Hanna, Seppänen, Mikko, Siljander, Tuula, Vuopio-Varkila, Jaana, and Meri, Seppo
- Subjects
- *
PHARYNGITIS , *PATHOGENIC microorganisms , *STREPTOCOCCUS , *LYMPHOID tissue - Abstract
Abstract: Group A streptococcus (GAS) is the most common pathogen causing bacterial pharyngitis. We isolated streptococcal strains from tonsils removed from patients with tonsillar disease (n =202) and studied their ability to bind the complement regulators factor H (FH) and C4b binding protein (C4BP) using 125I-labeled proteins. Blood isolates of GAS (n =10) were obtained from patients with bacteraemia. Streptococci were isolated from 21% of the tonsillitis patients. The emm and T types of the GAS strains were determined. Of the 26 GAS strains studied, only six could bind FH and/or C4BP above the threshold levels. The fraction of the offered radioactive protein bound ranged between 6–12% for FH and 19–56% for C4BP. The clinical course of the tonsillar disease was not related to the binding of FH or C4BP by GAS. The binding strains were mostly of the T4M4 or T28M28 type. From the invasive strains (n =10), three bound FH (binding level: 8–11%) and two C4BP (36–39%). The binding correlated only partially to M-protein (emm) type suggesting that the binding was not exclusively due to M-protein. The results indicate that complement regulator binding by GAS is only partially related to pathogenicity and not a universal property of all group A streptococci. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
32. Complement components, regulators and receptors are produced by human monocyte-derived dendritic cells
- Author
-
Reis, Edimara S., Barbuto, José Alexandre M., and Isaac, Lourdes
- Subjects
- *
LYMPHOID tissue , *NATURAL immunity , *DENDRITIC cells , *CARRIER proteins - Abstract
Abstract: Complement and dendritic cells (DCs) are essential components of innate immunity. Both participate in local inflammation and moreover have roles in the initiation of the acquired immunity response and in the maintenance of tolerance. Recent studies have demonstrated the ability of DCs to synthesize C1q, C3, Factor I, Factor B and complement receptors 3 and 4. In this study, we demonstrate that human DCs are a source of other soluble complement proteins including C1q, C4b binding protein (C4BP), C7 and C8. Complement receptors (CR)1 and the CD18 chain (common for CR3 and CR4) were also present on DCs while CR2 was not detected. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
33. The human serum protein C4b-binding protein inhibits pancreatic IAPP-induced inflammasome activation
- Author
-
Erik Renström, Nikolina Papac-Milicevic, Anna M. Blom, Gunilla T. Westermark, Ben C. King, and Klaudia Kulak
- Subjects
0301 basic medicine ,Male ,Medicin och hälsovetenskap ,endocrine system diseases ,Inflammasomes ,Endocrinology, Diabetes and Metabolism ,Interleukin-1beta ,Amylin ,Type 2 diabetes ,Medical and Health Sciences ,Inflammasome ,IAPP ,Insulin ,Cells, Cultured ,geography.geographical_feature_category ,C4b-binding protein ,Complement C4b-Binding Protein ,Diabetes ,Middle Aged ,Islet ,Cell biology ,Islet Amyloid Polypeptide ,Female ,medicine.symptom ,medicine.drug ,medicine.medical_specialty ,endocrine system ,Amyloid ,Blotting, Western ,Complement ,Inflammation ,Biology ,Article ,03 medical and health sciences ,Islets of Langerhans ,Internal medicine ,Cell Line, Tumor ,mental disorders ,NLR Family, Pyrin Domain-Containing 3 Protein ,Internal Medicine ,medicine ,Animals ,Humans ,C4BP ,Beta (finance) ,Pancreas ,Aged ,geography ,medicine.disease ,Rats ,030104 developmental biology ,Endocrinology - Abstract
Aims/hypothesis Inflammasome activation and subsequent IL-1β production is a driver of islet pathology in type 2 diabetes. Oligomers, but not mature amyloid fibrils, of human islet amyloid polypeptide (IAPP), which is co-secreted with insulin, trigger NOD-like receptor pyrin domain containing-3 (NLRP3) inflammasome activation. C4b-binding protein (C4BP), present in serum, binds to IAPP and affects transition of IAPP monomers and oligomers to amyloid fibrils. We therefore hypothesised that C4BP inhibits IAPP-mediated inflammasome activation and IL-1β production. Methods Macrophages were exposed to IAPP in the presence or absence of plasma-purified human C4BP, and inflammasome activation was assessed by IL-1β secretion as detected by ELISA and reporter cell lines. IAPP fibrillation was assessed by thioflavin T assay. Uptake of IAPP–C4BP complexes and their effects on phagolysosomal stability were assessed by flow cytometry and confocal microscopy. The effect of C4BP regulation of IAPP-mediated inflammasome activation on beta cell function was assessed using a clonal rat beta cell line. Immunohistochemistry was used to examine the association of IAPP amyloid deposits and macrophage infiltration in isolated human and mouse pancreatic islets, and expression of C4BP from isolated human pancreatic islets was assessed by quantitative PCR, immunohistochemistry and western blot. Results C4BP significantly inhibited IAPP-mediated IL-1β secretion from primed macrophages at physiological concentrations in a dose-dependent manner. C4BP bound to and was internalised together with IAPP. C4BP did not affect IAPP uptake into phagolysosomal compartments, although it did inhibit its formation into amyloid fibrils. The loss of macrophage phagolysosomal integrity induced by IAPP incubation was inhibited by co-incubation with C4BP. Supernatant fractions from macrophages activated with IAPP inhibited both insulin secretion and viability of clonal beta cells in an IL-1β-dependent manner but the presence of C4BP during macrophage IAPP incubation rescued beta cell function and viability. In human and mouse islets, the presence of amyloid deposits correlated with higher numbers of infiltrating macrophages. Isolated human islets expressed and secreted C4BP, which increased with addition of IL-1β. Conclusions/interpretation IAPP deposition is associated with inflammatory cell infiltrates in pancreatic islets. C4BP blocks IAPP-induced inflammasome activation by preventing the loss of macrophage phagolysosomal integrity required for NLRP3 activation. The consequence of this is the preservation of beta cell function and viability. C4BP is secreted directly from human pancreatic islets and this increases in response to inflammatory cytokines. We therefore propose that C4BP acts as an extracellular chaperone protein that limits the proinflammatory effects of IAPP. Electronic supplementary material The online version of this article (doi:10.1007/s00125-017-4286-3) contains peer-reviewed but unedited supplementary material, which is available to authorised users.
- Published
- 2017
34. Single-step purification of human C4b-binding protein (C4BP) by affinity chromatography on a peptide derived from a streptococcal surface protein
- Author
-
Persson, Jenny and Lindahl, Gunnar
- Subjects
- *
BLOOD plasma , *PROTEINS , *PROKARYOTES , *STAPHYLOCOCCUS aureus - Abstract
Abstract: Many Gram-positive bacteria express surface proteins that bind human plasma proteins. These bacterial proteins, and derivatives of them, are of interest for analysis of bacterial pathogenesis and as immunochemical tools. Well-characterized examples include the IgG-binding reagents staphylococcal protein A and streptococcal protein G, and the recently described streptococcal IgA-binding peptide Sap. Here, we show that a peptide derived from the streptococcal M22 protein can be used for single-step affinity purification of the human complement regulator C4b-binding protein (C4BP). Binding of C4BP was strongly enhanced by dimerization of the peptide via a C-terminal cysteine residue not present in the intact M22 protein. The purified C4BP had the expected binding characteristics, and acted as a cofactor for factor I in the degradation of C4b. Passage of serum through a peptide column under non-saturating conditions resulted in binding of >99.5% of serum C4BP, implying that such a column can be used to deplete serum of C4BP. These data indicate that the C4BP-binding peptide is a versatile tool that can be used for simple and rapid purification of biologically active human C4BP or for removal of C4BP from serum. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
35. C4b Binding Protein (C4BP) acts as an innate immune effector against Influenza A Virus
- Author
-
Varghese, PM, Murugaiah, V, Nazar, B, Temperton, N, Khan, HA, Alrokayan, S, Al-Ahdal, MN, Nal, B, A-Mohanna, F, Sim, RB, and Kishore, U
- Subjects
Inflammation ,InformationSystems_GENERAL ,ComputerApplications_MISCELLANEOUS ,Complement ,C4BP ,pseudo-typed particles ,Influenza - Abstract
International Scientific Partnership Programme
- Published
- 2020
36. Effect of hybrid complement regulatory proteins on xenogeneic cells
- Author
-
Fukuta, Daisuke, Miyagawa, Shuji, Kubo, Tomoko, Matsunami, Katsuyoshi, Shirasu, Akio, Hattori, Hiroyuki, and Shirakura, Ryota
- Subjects
- *
COMPLEMENTATION (Genetics) , *PROTEINS - Abstract
To suppress C3 fragment deposition in the classical pathway complement activation on xenogeneic membranes, decay accelerating factor (DAF) was the most effective molecule among the complement regulatory proteins (CRPs) used in the present study. C3 fragment deposition was closely related to subsequent xenogeneic cell lysis. However, other molecules were also very effective in different ways and include phosphatidylinositol (PI)-anchored short consensus repeat (SCR) 2–4 of membrane cofactor protein (MCP-PI), PI-anchored C1 esterase inhibitor (C1-INH-PI), and PI-anchored SCR8–11 of complement receptor type 1 (CR1-PI). On the other hand, regarding a strategy for downregulating C4 fragment deposition, the use of only C1-INH-PI and PI-anchored SCR1–3 of the C4b-binding protein (C4bp-PI) was found to be effective. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
37. Role of CCP2 of the C4b-binding protein β-chain in protein S binding evaluated by mutagenesis and monoclonal antibodies.
- Author
-
Webb, Joanna H., Villoutreix, Bruno O., Dahlbäck, Björn, and Blom, Anna M.
- Subjects
- *
COMPLEMENT (Immunology) , *PROTEIN S , *PROTEIN binding - Abstract
Complement regulator C4b-binding protein (C4BP) and the anticoagulant vitamin K-dependent protein S form a high affinity complex in human plasma. C4BP is composed of seven α-chains and a unique β-chain, each chain comprising repeating complement control protein (CCP) modules. The binding site for protein S mainly involves the first of the three β-chain CCPs (CCP1). However, recently it has been suggested that CCP2 of the β-chain also contributes to the binding of protein S. To elucidate the structural background for the involvement of CCP2 in the protein S binding, several recombinant β-chain CCP1-2 variants having mutations in CCP2 were expressed and tested for protein S binding. Mutations were chosen based on analysis of a homology model of the β-chain and included R60A/R101A, D66A, L105A, F114A/I116A and H108A. All mutant proteins bound equally well as recombinant wild type to protein S. Several monoclonal antibodies against the β-chain CCP2 were raised and their influence on protein S binding characterized. Taken together, the results suggest that the role of CCP2 in protein S binding is to orient and stabilize CCP1 rather than to be directly part of the binding site. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
38. Decline in the expression of C4 binding protein alpha-chain gene during ageing of the rat liver.
- Author
-
Lavery, Lindsay W. and Goyns, Malcolm H.
- Subjects
CELLS ,GENETIC polymorphisms ,POPULATION genetics ,DENSITOMETRY ,OPTICAL measurements ,AGING - Abstract
It appears that consistent changes in the levels of activity of a small cohort of genes (probably less than 1% of all active genes) occur in all mammalian cells during ageing. We have studied this phenomenon in rat liver using an optimised form of differential display. During this investigation we observed one gene which exhibited a decline in expression in livers from young adult (6 months) to aged adult (24 months) animals. The differential expression of this gene was confirmed by single strand conformational polymorphism (SSCP) gel analysis and Northern blotting. Densitometry of the latter indicated that there was a decline of 35% in its expression with age. Characterisation of the isolated PCR fragment demonstrated it to code for the alpha subchain of the complement 4 binding protein (C4BP). The C4BP is a key regulatory protein of the complement system and this observation therefore indicates that a decline in the efficiency of the complement system may be an important factor in the overall decline in immune function that has been observed during ageing. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
39. Consumption of C4b-binding protein (C4BP) during in vivo activation of the classical complement pathway.
- Author
-
BERGAMASCHINI, MIEDICO, CICARDI, COPPOLA, FAIONI, AGOSTONI, and Bergamaschini, Luigi
- Subjects
- *
GLYCOPROTEINS , *COMPLEMENT activation - Abstract
C4BP has a central role in regulating the classical complement (C′) pathway, but it is still uncertain whether or not it is consumed during in vivo complement activation. Attempts to demonstrate changes in C4BP plasma levels in systemic lupus erythematosus and essential mixed cryoglobulinaemia have failed, probably due to up-regulation of this protein during the inflammatory reaction. We have studied one patient with severe post-transfusion complement-mediated anaphylaxis (CMA), and 67 patients with hereditary C1 inhibitor deficiency (hereditary angioedema (HAE)). The first of these two conditions is characterized by the absence of systemic inflammatory reaction and the second by acute and chronic activation of the C′ classical pathway. C4BP, C4BP–C4b complex, and soluble terminal C′ complex (sC5b-9) were measured in the patients' plasmas by ELISA techniques and C3a and C4a by radioimmunoassays. In CMA, 15 min after the transfusion, there was a massive C′ activation, with increases in C4a, C3a, sC5b-9, C4BP–C4b complexes and decreases in C4, C3 and C4BP. All parameters reverted to preinfusion values within 24 h. Depletion of C4 was correlated with that of C4BP. In patients with HAE, the median value of C4BP (83% range 54–165) was significantly lower (P < 0.0001) than in normal controls (99% range 70–159), with no difference between patients in remission or during acute attacks. C4BP–C4b complexes could not be detected in HAE patients. The results of this study indicate that C4BP is consumed in vivo during acute, and possibly during chronic activation of the C′ classical pathway, and that this protein, after interaction with C4b, not longer circulates in plasma. [ABSTRACT FROM AUTHOR]
- Published
- 1999
- Full Text
- View/download PDF
40. FHR4-based immunoconjugates direct complement-dependent cytotoxicity and phagocytosis towards HER2-positive cancer cells
- Author
-
Marie Fullana, Xavier Dervillez, Mihály Józsi, Charlène Verschueren, Gilles Iserentant, Cécile Masquelier, Jacques H. M. Cohen, Carole Seguin-Devaux, and Jean-Marc Plesseria
- Subjects
0301 basic medicine ,Cancer Research ,Immunoconjugates ,Receptor, ErbB-2 ,Cell ,Complement Membrane Attack Complex ,lcsh:RC254-282 ,Epitope ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Antibodies, Bispecific ,Genetics ,medicine ,Humans ,C4bp ,Cytotoxicity ,Complement Activation ,Research Articles ,FHR4 ,MAC ,Chemistry ,multimers ,General Medicine ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Complement-dependent cytotoxicity ,Complement system ,Cell biology ,Apolipoproteins ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Alternative complement pathway ,Molecular Medicine ,complement resistance ,Complement membrane attack complex ,CDC ,Research Article - Abstract
Directing selective complement activation towards tumour cells is an attractive strategy to promote their elimination. In the present work, we have generated heteromultimeric immunoconjugates that selectively activate the complement alternative pathway (AP) on tumour cells. We used the C4b‐binding protein C‐terminal‐α‐/β‐chain scaffold for multimerisation to generate heteromultimeric immunoconjugates displaying (a) a multivalent‐positive regulator of the AP, the human factor H‐related protein 4 (FHR4) with; (b) a multivalent targeting function directed against erbB2 (HER2); and (c) a monovalent enhanced GFP tracking function. Two distinct VHH targeting two different epitopes against HER2 and competing either with trastuzumab or with pertuzumab‐recognising epitopes [VHH(T) or VHH(P)], respectively, were used as HER2 anchoring moieties. Optimised high‐FHR4 valence heteromultimeric immunoconjugates [FHR4/VHH(T) or FHR4/VHH(P)] were selected by sequential cell cloning and a selective multistep His‐Trap purification. Optimised FHR4‐heteromultimeric immunoconjugates successfully overcame FH‐mediated complement inhibition threshold, causing increased C3b deposition on SK‐OV‐3, BT474 and SK‐BR3 tumour cells, and increased formation of lytic membrane attack complex densities and complement‐dependent cytotoxicity (CDC). CDC varies according to the pattern expression and densities of membrane‐anchored complement regulatory proteins on tumour cell surfaces. In addition, opsonised BT474 tumour cells were efficiently phagocytosed by macrophages through complement‐dependent cell‐mediated cytotoxicity. We showed that the degree of FHR4‐multivalency within the multimeric immunoconjugates was the key element to efficiently compete and deregulate FH and FH‐mediated convertase decay locally on tumour cell surface. FHR4 can thus represent a novel therapeutic molecule, when expressed as a multimeric entity and associated with an anchoring system, to locally shift the complement steady‐state towards activation on tumour cell surface., We propose a new approach of complement‐mediated destructive tumour cell targeting by generating immunoconjugates harbouring multimeric (a) factor H‐related protein 4 (FHR4)‐complement effector functions with; (b) VHH anti‐HER2 targeting functions. The number of FHR4 valences dictates the efficacy of the multimers to selectively and locally activate complement alternative pathway on HER2‐tumour cell surface, leading to complement‐dependent cytotoxicity and complement‐dependent cell‐mediated phagocytosis.
- Published
- 2019
41. Chimeric Proteins Containing MAP-1 and Functional Domains of C4b-Binding Protein Reveal Strong Complement Inhibitory Capacities
- Author
-
Peter Garred, Sander van Putten, Nikolaj Kirketerp-Møller, Rafael Bayarri-Olmos, Mikkel-Ole Skjoedt, Katrine Pilely, and Cecilie Elkjær Hertz
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,Recombinant Fusion Proteins ,Immunology ,Enzyme-Linked Immunosorbent Assay ,CHO Cells ,lectin pathway ,03 medical and health sciences ,Classical complement pathway ,Cricetulus ,Immune system ,complement inhibition ,Animals ,Humans ,Immunology and Allergy ,C4BP ,classical pathway ,Molecular Biology ,chimeric protein ,Original Research ,Adaptor Proteins, Signal Transducing ,complement activation ,biology ,C4b-binding protein ,Chemistry ,Complement C4b-Binding Protein ,Chinese hamster ovary cell ,Lectin ,Complement Pathway, Mannose-Binding Lectin ,Fusion protein ,Complement (complexity) ,Cell biology ,Complement system ,030104 developmental biology ,Lectin pathway ,biology.protein ,Apoptosis Regulatory Proteins ,lcsh:RC581-607 ,MAP-1 - Abstract
The complement system is a tightly regulated network of proteins involved in defense against pathogens, inflammatory processes, and coordination of the innate and adaptive immune responses. Dysregulation of the complement cascade is associated with many inflammatory disorders. Thus, inhibition of the complement system has emerged as an option for treatment of a range of different inflammatory diseases. MAP-1 is a pattern recognition molecule (PRM)-associated inhibitor of the lectin pathway of the complement system, whereas C4b-binding protein (C4BP) regulates both the classical and lectin pathways. In this study we generated chimeric proteins consisting of MAP-1 and the first five domains of human C4BP (C4BP1-5) in order to develop a targeted inhibitor acting at different levels of the complement cascade. Two different constructs were designed and expressed in CHO cells where MAP-1 was fused with C4BP1-5 in either the C- or N-terminus. The functionality of the chimeric proteins was assessed using different in vitro complement activation assays. Both chimeric proteins displayed the characteristic Ca2+-dependent dimerization and binding to PRMs of native MAP-1, as well as the co-factor activity of native C4BP. In ELISA-based complement activation assays they could effectively inhibit the lectin and classical pathways. Notably, MAP-1:C4BP1-5 was five times more effective than rMAP-1 and rC4BP1-5 applied at the same time, emphasizing the advantage of a single inhibitor containing both functional domains. The MAP-1/C4BP chimeras exert unique complement inhibitory properties and represent a novel therapeutic approach targeting both upstream and central complement activation.
- Published
- 2018
- Full Text
- View/download PDF
42. A novel C3d-containing oligomeric vaccine provides insight into the viability of testing human C3d-based vaccines in mice
- Author
-
Yong-Gang, He, Isabel Y, Pappworth, Andreas, Rossbach, Joshua, Paulin, Tarirai, Mavimba, Christine, Hayes, Liudmila, Kulik, V Michael, Holers, Andrew M, Knight, and Kevin J, Marchbank
- Subjects
chemical and pharmacologic phenomena ,Antibodies ,Article ,RT-PCR, reverse transcriptase polymerase chain reaction ,Cell Line ,C3d ,SLE, systemic lupus erythematosus ,Mice ,Adjuvants, Immunologic ,Tetanus Toxin ,CR, complement receptor ,Animals ,Humans ,C4BP ,SRBC, sheep red blood cell ,Complement receptor ,FDC, follicular dendritic cell ,FO, follicular ,Adjuvant ,Mice, Knockout ,B-Lymphocytes ,Vaccines, Synthetic ,B cell ,IC, immune complex ,Complement C4b-Binding Protein ,Vaccination ,Peptide Fragments ,Mice, Inbred C57BL ,Complement C3d ,SCR, short consensus repeat ,Models, Animal ,BM, bone marrow ,Receptors, Complement 3d ,Protein Multimerization ,MZ, marginal zone ,GC, germinal centre - Abstract
The use of C3d, the final degradation product of complement protein C3, as a “natural” adjuvant has been widely examined since the initial documentation of its immunogenicity-enhancing properties as a consequence of binding to complement receptor 2. Subsequently it was demonstrated that these effects are most evident when oligomeric, rather than when monomeric forms of C3d, are linked to various test protein antigens. In this study, we examined the feasibility of enhancing the adjuvant properties of human C3d further by utilizing C4b-binding protein (C4BP) to provide an oligomeric arrayed scaffold fused to the model antigen, tetanus toxin C fragment (TTCF). High molecular weight, C3d-containing oligomeric vaccines were successfully expressed, purified from mammalian cells and used to immunize groups of mice. Surprisingly, anti-TTCF antibody responses measured in these mice were poor. Subsequently we established by in vitro and in vivo analysis that, in the presence of mouse C3, human C3d does not interact with either mouse or even human complement receptor 2. These data confirm the requirement to develop murine versions of C3d based adjuvant compounds to test in mice or that mice would need to be developed that express both human C3 and human CR2 to allow the testing of human C3d based adjuvants in mouse in any capacity.
- Published
- 2017
43. Reduction in protein S activity during normal pregnancy.
- Author
-
KURASAWA, Gotaro, KOTANI, Kazuhiko, ITO, Yuji, SAIGA, Kyoko, and IIJIMA, Kenji
- Subjects
- *
PROTEIN S , *PROTEIN C , *BLOOD proteins , *DELIVERY (Obstetrics) , *PREGNANCY , *PREGNANT women - Abstract
We investigated the serial changes in blood protein S (PS) and related proteins in 11 normal pregnant women. The PS activity decreased significantly in the third trimester and reached minimum levels (23.3%) one hour after delivery. Although the PS activity was reduced markedly below the normal limits, all the women delivered safely. The mechanisms that cause the reduction in PS activity and the clinically dangerous conditions involving PS activity during pregnancy warrant further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
44. Complement Factor H interferes with Mycobacterium bovis BCG entry into macrophages and modulates the pro-inflammatory cytokine response
- Author
-
Abdul-Aziz, Munirah, Tsolaki, Anthony G., Kouser, Lubna, Carroll, Maria V., Al-Ahdal, Mohammed N., Sim, Robert B., and Kishore, Uday
- Subjects
Phagocytosis ,Macrophage ,Immunology ,Factor H ,Complement ,Immunology and Allergy ,Tuberculosis ,BCG ,C4BP ,Hematology ,Cytokine ,Mycobacterium - Abstract
Mycobacterium tuberculosis is an accomplished intracellular pathogen, particularly within the macrophage and this is of the utmost importance in the host-pathogen stand-off observed in the granuloma during latent tuberculosis. Contact with innate immune molecules is one of the primary interactions that can occur with the pathogen M. tuberculosis once inhaled. Complement proteins may play a role in facilitating M. tuberculosis interactions with macrophages. Here, we demonstrate that factor H, a complement regulatory protein that down-regulates complement alternative pathway activation, binds directly to the model organism M. bovis BCG. Binding of factor H reaches saturation at 5–10μg of factor H/ml, well below the plasma level. C4 binding protein (C4BP) competed with factor H for binding to mycobacteria. Factor H was also found to inhibit uptake of M. bovis BCG by THP-1 macrophage cells in a dose-dependent manner. Real-time qPCR analysis showed stark differential responses of pro- and anti-inflammatory cytokines during the early stages of phagocytosis, as evident from elevated levels of TNF-α, IL-1β and IL-6, and a concomitant decrease in IL-10, TGF-β and IL-12 levels, when THP-1:BCG interaction took place in the presence of factor H. Our results suggest that factor H can interfere with mycobacterial entry into macrophages and modulate inflammatory cytokine responses, particularly during the initial stages of infection, thus affecting the extracellular survival of the pathogen. Our results offer novel insights into complement activation-independent functions of factor H during the host-pathogen interaction in tuberculosis.
- Published
- 2016
45. Serum profiling of leptospirosis patients to investigate proteomic alterations
- Author
-
Sandipan Ray, Kishore Gollapalli, Tulip Jhaveri, Snigdha Dhali, Rajneesh Srivastava, Santosh R Taur, Sanjeeva Srivastava, Urmila M Thatte, Vineet Vaibhav, Rapole Srikanth, and Nithya J Gogtay
- Subjects
Serum ,Male ,Proteomics ,Proteome ,Biophysics ,Activation ,Acute phase signaling ,Pathogenesis ,Disease ,Biology ,Biochemistry ,Article ,Coagulation Cascade ,medicine ,Humans ,C4bp ,Leptospirosis ,Interrogans ,Mass spectrometry ,Coinfection ,Acute-phase protein ,Proteins ,Blood Proteins ,Binding ,medicine.disease ,Blood proteins ,Malaria ,Gene Expression Regulation ,Infectious disease (medical specialty) ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Immunology ,2D-DIGE ,Female ,Biomarkers - Abstract
Leptospirosis is a zoonotic infectious disease of tropical, subtropical and temperate zones, which is caused by the pathogenic spirochetes of genus Leptospira. Although this zoonosis is generally not considered as fatal, the pathogen can eventually cause severe infection with septic shock, multi-organ failure and lethal pulmonary hemorrhages leading to mortality. In this study, we have performed a proteomic analysis of serum samples from leptospirosis patients (n = 6), febrile controls (falciparum malaria) (n = 8) and healthy subjects (n = 18) to obtain an insight about disease pathogenesis and host immune responses in leptospiral infections. 2DE and 2D-DIGE analysis in combination with MALDI-TOF/TOF MS revealed differential expression of 22 serum proteins in leptospirosis patients compared to the healthy controls. Among the identified differentially expressed proteins, 8 candidates exhibited different trends compared to the febrile controls. Functional analysis suggested the involvement of differentially expressed proteins in vital physiological pathways, including acute phase response, complement and coagulation cascades and hemostasis. This is the first report of analysis of human serum proteome alterations in leptospirosis patients, which revealed several differentially expressed proteins, including α-1-antitrypsin, vitronectin, ceruloplasmin, G-protein signaling regulator, apolipoprotein A-IV, which have not been reported in context of leptospirosis previously. This study will enhance our understanding about leptospirosis pathogenesis and provide a glimpse of host immunological responses. Additionally, a few differentially expressed proteins identified in this study may further be investigated as diagnostic or prognostic serum biomarkers for leptospirosis. This article is part of a Special Issue entitled: Integrated omics., Graphical abstract Highlights ► Alteration in serum proteome in Leptospirosis was investigated. ► Classical 2DE and 2D-DIGE were used for serum proteome profiling. ► Differentially expressed serum proteins were identified using MALDI TOF/TOF MS. ► Study enhanced understanding about leptospirosis pathogenesis and provided a glimpse of host immunological responses.
- Published
- 2012
- Full Text
- View/download PDF
46. Interactions between platelets and complement with implications for the regulation at surfaces
- Author
-
Nilsson, Per H.
- Subjects
ADP ,regulation) ,Cell- och molekylärbiologi ,Immunology ,Biomaterialvetenskap ,Biochemistry and Molecular Biology ,apyrase ,factor H ,biocompatibility ,platelets (activation ,complement system (activation ,chondroitin sulfate-A ,Immunologi ,Biomaterials Science ,C4BP ,C3 ,C1q ,Biokemi och molekylärbiologi ,Cell and Molecular Biology ,biomaterials - Abstract
Disturbances of host integrity have the potential to evoke activation of innate immunologic and hemostatic protection mechanisms in blood. Irrespective of whether the activating stimulus is typically immunogenic or thrombotic, it will generally affect both the complement system and platelets to a certain degree. The theme of this thesis is complement and platelet activity, which is intersected in all five included papers. The initial aim was to study the responses and mechanisms of the complement cascade in relation to platelet activation. The secondary aim was to use an applied approach to regulate platelets and complement on model biomaterial and cell surfaces. Complement activation was found in the fluid phase in response to platelet activation in whole blood. The mechanism was traced to platelet release of stored chondroitin sulfate-A (CS-A) and classical pathway activation via C1q. C3 was detected at the platelet surface, though its binding was independent of complement activation. The inhibitors factor H and C4-binding protein (C4BP) were detected on activated platelets, and their binding was partly dependent on surface-exposed CS-A. Collectively, these results showed that platelet activation induces inflammatory complement activation in the fluid phase. CS-A was shown to be a central molecule in the complement-modulatory functions of platelets by its interaction with C1q, C4BP, and factor H. Platelet activation and surface adherence were successfully attenuated by conjugating an ADP-degrading apyrase on a model biomaterial. Only minor complement regulation was seen, and was therefore targeted specifically on surfaces and cells by co-immobilizing a factor H-binding peptide together with the apyrase. This combined approach led to a synchronized inhibition of both platelet and complement activation at the interface of biomaterials/xenogeneic cells and blood. In conclusion, here presents a novel crosstalk-mechanism for activation of complement when triggering platelets, which highlights the importance of regulating both complement and platelets to lower inflammatory events. In addition, a strategy to enhance the biocompatibility of biomaterials and cells by simultaneously targeting ADP-dependent platelet activation and the alternative complement C3-convertase is proposed.
- Published
- 2012
47. Comparison of two immunoassays for the complement protein C4b-binding protein in health and disease
- Author
-
Coppola, R., Tombesi, S., Cristilli, P., Bergamaschini, L., and Mannucci, P. M.
- Published
- 1995
- Full Text
- View/download PDF
48. Crosstalk Between Activated Platelets and the Complement System
- Author
-
Hamad, Osama A.
- Subjects
platelet-leukocyte complexes ,platelet microparticles ,Immunology in the medical area ,factor H ,TRAP ,Immunologi inom det medicinska området ,platelets ,C4BP ,Compstatin ,complement ,C3 ,activated platelets ,C1q ,chondroitin sulfate - Abstract
Several studies have shown that complement and thrombotic events co-exist. Platelets have been suspected to act as the bridge between the two cascade systems. To study the platelet-induced complement activation we developed a system in which platelets were activated by thrombin receptor activating peptide (TRAP) in platelet rich plasma (PRP) or whole blood anti-coagulated using the specific thrombin inhibitor, lepirudin. TRAP-activated platelets induced a fluid-phase complement activation measured as generation of C3a and sC5b-9, triggered by released chondroitin sulphate-A (CS-A) which interacted with C1q and activated the complement system through the classical pathway. Complement components C1q, C3, C4 and C9 were also shown to bind to TRAP-activated platelets but this binding did not seem to be due to a complement activation since blocking of complement activation at the C1q or C3 levels did not affect the binding of the complement proteins. The C3 which bound to activated platelets consisted of C3(H2O), indicating that bound C3 was not proteolytically activated. Binding of C1q was partially dependent on CS-A exposure on activated platelets. The abolished complement activation on the surface of activated platelets was suggested to be dependent on the involvement of several complement inhibitors. We confirmed the binding of C1INH and factor H to activated platelets. To this list we have added another potent complement inhibitor, C4BP. The binding of factor H and C4BP was shown to be dependent on exposure of CS-A on activated platelets. The physiological relevance of these reactions was reflected in an elevated expression of CD11b on leukocytes, and increased generation of platelet-leukocyte complexes. The platelets were involved in these events by at least two different mechanisms; generation of C5a which activated leukocytes and binding of C3(H2O)/iC3(H2O), a ligand to the intergrin CD11b/CD18 on their surface. These mechanisms add further to the understanding of how platelets interact with the complement system and will help us to understand the role of the complement system in cardiovascular disease and thrombotic conditions. Platelet Mediated Complement Activation
- Published
- 2010
49. Identifying functional adenovirus-host interactions using tandem mass spectrometry
- Author
-
Anuj, Gaggar, Dmitry, Shayakhmetov, and André, Lieber
- Subjects
Coxsackie and adenovirus receptor ,Base Sequence ,viruses ,CHO Cells ,Recombinant Proteins ,Article ,Adenoviridae ,Mice ,Viral Proteins ,Cricetulus ,Serotype 5 ,Tandem Mass Spectrometry ,Cricetinae ,Animals ,Humans ,Receptors, Virus ,Electrophoresis, Polyacrylamide Gel ,C4BP ,CD46 ,serotype 35 ,DNA Primers ,fiber - Abstract
We describe a systematic, high-throughput approach to identify proteins involved in functional adenovirus (Ad)-host interactions in vitro and in vivo. We were particularly interested in identifying cellular proteins that interact with fiber knob, which is the moiety within the Ad capsid responsible for high-affinity attachment of virus to cellular receptors. We used recombinant fiber knob domains from members of group C and B Ads to purify virus interacting proteins from cell membrane lysates and from human and mouse plasma. Using tandem mass spectrometry, we identified a number of candidate Ad-interacting proteins, including functional cellular receptors and previously unknown interacting partners such as complement component C4-binding protein and other blood proteins that presumably are involved in Ad infection after intravenous virus application. The ability of these proteins to bind to Ad was further confirmed using in vitro protein binding assays as well as infection competition assays. The approach of using a structural protein can be universally applied for a variety of viral and nonviral pathogens and can reveal host cell factors critical in viral infection, immune evasion, and tissue specificity. This information is also a prerequisite to assess in vivo safety and efficacy of Ad-based gene transfer vectors.
- Published
- 2007
50. Low-density lipoprotein receptor-related protein : Novel aspects revealed
- Author
-
Spijkers, Patricia Petronella Elisabeth Maria and University Utrecht
- Subjects
Geneeskunde ,lymphocytes ,adhesion ,β2-integrins ,LRP ,receptor ,expression ,lipids (amino acids, peptides, and proteins) ,alzheimer’s disease ,C4BP ,clearance ,monocytes - Abstract
LDL receptor-related protein (LRP) is a multifunctional cell surface receptor that is expressed in many cell types. It is able to bind numerous structurally unrelated ligands. This thesis describes four novel functions of LRP in different cell types. β2-integrins are heterodimeric proteins present at the cell surface of leukocytes. They are involved in various processes, like leukocyte adhesion to the vascular wall. We reported that LRP directly bound to the α-subunit of β2-integrins. Upon activation, β2-integrins form clusters in rafts of monocytes to increase the avidity for its ligands. LRP was co-immunoprecipitated with β2-integrins in the raft fraction of both naïve and stimulated monocytes. In addition, LRP colocalized with β2-integrins in the clusters. Downregulation of LRP disrupted the β2-integrin clusters, which resulted in a decreased adhesion of monocytes to activated endothelium. These data indicate that LRP is required for optimal β2-integrin function. These results prompted us to explore the functions of LRP in other leukcoytes. Though, cellular expression patterns in lymphocytes have not been fully investigated yet. Our study showed that LRP is present in CD4+ and CD8+ T-cells, natural killer (NK) cells and B-cells. We observed a differential localization of LRP in these cells. Whereas it was present at the cell surface of NK cells and B-cells, the total LRP pool was localized intracellularly in both T-cell subtypes. In these cells, LRP could be transported to the cell surface upon a reaction with allogenic monocytes. These data point to a novel role for LRP in immunologically based diseases, like graft-versus-host-disease. In brain, LRP is a key component in the pathogenesis of Alzheimer’s Disease (AD). It is involved in the uptake of amyloid β (Aβ) precursor protein and in the further processing of it, eventually leading to secretion of β peptides. High β concentrations can lead to the development of AD. The present study showed that β could directly bind to LRP at the cell surface brain endocthelial cells (BECs), with the Aβ40 isoform having a higher affinity than Aβ42. LRP mediated the internalization of β peptides, which were subsequently transported to the blood across the blood-brain barrier. The presence of high concentrations of Aβ decreased LRP levels in BECs, caused by a direction of LRP into the proteasomal degradation pathway. Interestingly, brain LRP levels in mice and patients suffering from AD were also decreased, suggesting LRP to be a new therapeutic target in the challenge of AD. LRP was also involved in the binding and uptake of C4b-binding protein (C4BP), but this uptake resulted in the degradation of C4BP. Mutant C4BP with replacements of their positively charged residues were less efficiently degraded. These residues were also able to bind heparin. We found that heparan sulfate proteoglycans mediated the first step of cellular C4BP binding, which was followed by LRP-mediated degradation. C4BP can also bind anticoagulant factor protein S. Biacore experiments showed that protein S-complexed C4BP was still able to bind to LRP. Future research may be emphasized on how one single receptor can exert many, distinct functions.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.