51
|
Klos A, Wende E, Wareham KJ, Monk PN. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXVII. Complement peptide C5a, C4a, and C3a receptors. Pharmacol Rev 2013; 65:500-43. [PMID: 23383423 DOI: 10.1124/pr.111.005223] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The activation of the complement cascade, a cornerstone of the innate immune response, produces a number of small (74-77 amino acid) fragments, originally termed anaphylatoxins, that are potent chemoattractants and secretagogues that act on a wide variety of cell types. These fragments, C5a, C4a, and C3a, participate at all levels of the immune response and are also involved in other processes such as neural development and organ regeneration. Their primary function, however, is in inflammation, so they are important targets for the development of antiinflammatory therapies. Only three receptors for complement peptides have been found, but there are no satisfactory antagonists as yet, despite intensive investigation. In humans, there is a single receptor for C3a (C3a receptor), no known receptor for C4a, and two receptors for C5a (C5a₁ receptor and C5a₂ receptor). The most recently characterized receptor, the C5a₂ receptor (previously known as C5L2 or GPR77), has been regarded as a passive binding protein, but signaling activities are now ascribed to it, so we propose that it be formally identified as a receptor and be given a name to reflect this. Here, we describe the complex biology of the complement peptides, introduce a new suggested nomenclature, and review our current knowledge of receptor pharmacology.
Collapse
Affiliation(s)
- Andreas Klos
- Department for Medical Microbiology, Medical School Hannover, Hannover, Germany
| | | | | | | |
Collapse
|
52
|
Moghimi SM, Farhangrazi ZS. Nanomedicine and the complement paradigm. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:458-60. [DOI: 10.1016/j.nano.2013.02.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 02/21/2013] [Indexed: 10/27/2022]
|
53
|
Ricklin D, Lambris JD. Complement in immune and inflammatory disorders: therapeutic interventions. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:3839-47. [PMID: 23564578 PMCID: PMC3623010 DOI: 10.4049/jimmunol.1203200] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
With the awareness that immune-inflammatory cross-talk is at the heart of many disorders, the desire for novel immunomodulatory strategies in the therapy of such diseases has grown dramatically. As a prime initiator and important modulator of immunological and inflammatory processes, the complement system has emerged as an attractive target for early and upstream intervention in inflammatory diseases and has moved into the spotlight of drug discovery. Although prevalent conditions such as age-related macular degeneration have attracted the most attention, the diverse array of complement-mediated pathologies, with distinct underlying mechanisms, demands a multifaceted arsenal of therapeutic strategies. Fortunately, efforts in recent years have not only introduced the first complement inhibitors to the clinic but also filled the pipelines with promising candidates. With a focus on immunomodulatory strategies, in this review we discuss complement-directed therapeutic concepts and highlight promising candidate molecules.
Collapse
Affiliation(s)
- Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
54
|
Qu H, Ricklin D, Bai H, Chen H, Reis ES, Maciejewski M, Tzekou A, DeAngelis RA, Resuello RRG, Lupu F, Barlow PN, Lambris JD. New analogs of the clinical complement inhibitor compstatin with subnanomolar affinity and enhanced pharmacokinetic properties. Immunobiology 2013; 218:496-505. [PMID: 22795972 PMCID: PMC3518557 DOI: 10.1016/j.imbio.2012.06.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 06/12/2012] [Indexed: 02/04/2023]
Abstract
Therapeutic modulation of the complement system has become increasingly important in line with the growing recognition of the role of complement in numerous diseases. Compstatin, a peptidic inhibitor that acts at the central level of the complement cascade, is currently in clinical evaluation but routes to improve its efficacy have not yet been fully explored. Here, we report improvements in both the inhibitory potency and pharmacokinetic parameters of compstatin that broaden its clinical applications. Selective modification of the compstatin N-terminus with non-proteinogenic amino acids resulted in the first analogue with subnanomolar binding affinity (KD=0.5nM) and other similarly potent derivatives with improved solubility in clinically relevant solvents. Detailed structure-activity relationship studies based on biophysical and computational methods revealed key structural determinants for the observed improvements. Importantly, pharmacokinetic evaluation in non-human primates revealed target-driven elimination kinetics with plasma half-life values exceeding expectations for peptidic drugs (close to 12h). This successful optimization strategy is expected to pave the way for systemic administration of compstatin in a range of clinical conditions.
Collapse
Affiliation(s)
- Hongchang Qu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Hongjun Bai
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Hui Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Edimara S. Reis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Mateusz Maciejewski
- School of Chemistry and School of Biological Sciences, University of Edinburgh, United Kingdom
| | - Apostolia Tzekou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Robert A. DeAngelis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | | | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, USA
| | - Paul N. Barlow
- School of Chemistry and School of Biological Sciences, University of Edinburgh, United Kingdom
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
55
|
Ricklin D. Manipulating the mediator: modulation of the alternative complement pathway C3 convertase in health, disease and therapy. Immunobiology 2013; 217:1057-66. [PMID: 22964231 DOI: 10.1016/j.imbio.2012.07.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 07/17/2012] [Accepted: 07/17/2012] [Indexed: 10/27/2022]
Abstract
The complement network is increasingly recognized as an important triage system that is able to differentiate between healthy host cells, microbial intruders, cellular debris and immune complexes, and tailor its actions accordingly. At the center of this triage mechanism is the alternative pathway C3 convertase (C3bBb), a potent enzymatic protein complex capable of rapidly converting the inert yet abundant component C3 into powerful effector fragments (C3a and C3b), thereby amplifying the initial response on unprotected surfaces and inducing a variety of effector functions. A fascinating molecular mechanism of convertase assembly and intrinsic regulation, as well as the interplay with a panel of cell surface-bound and soluble inhibitors are essential for directing complement attack to intruders and protecting healthy host cells. While efficiently keeping immune surveillance and homeostasis on track, the reliance on an intricate cascade of interaction and conversion steps also renders the C3 convertase vulnerable to derail. On the one hand, tissue damage, accumulation of debris, or polymorphisms in complement genes may unfavorably shift the balance between activation and regulation, thereby contributing to a variety of clinical conditions. On the other hand, pathogens developed powerful evasion strategies to avoid complement attack by targeting the convertase. Finally, we increasingly challenge our bodies with foreign materials such as biomaterial implants or drug delivery vehicles that may induce adverse effects that are at least partially caused by complement activation and amplification via the alternative pathway. The involvement of the C3 convertase in a range of pathological conditions put this complex into the spotlight of complement-targeted drug discovery efforts. Fortunately, the physiological regulation and microbial evasion approaches provide a rich source of inspiration for the development of powerful treatment options. This review provides insight into the current knowledge about the molecular mechanisms that drive C3 convertase activity, reveals common and divergent strategies of convertase inhibition employed by host and pathogens, and how this inhibitory arsenal can be tapped for developing therapeutic options to treat complement-related diseases.
Collapse
Affiliation(s)
- Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia 19104, USA.
| |
Collapse
|
56
|
Targeted complement inhibition as a promising strategy for preventing inflammatory complications in hemodialysis. Immunobiology 2013; 217:1097-105. [PMID: 22964235 DOI: 10.1016/j.imbio.2012.07.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 07/17/2012] [Indexed: 01/10/2023]
Abstract
Hemodialysis is the most common method used to remove waste and hazardous products of metabolism in patients suffering from renal failure. Hundreds of thousands of people with end-stage renal disease undergo hemodialysis treatment in the United States each year. Strikingly, the 5-year survival rate for all dialysis patients is only 35%. Most of the patients succumb to cardiovascular disease that is exacerbated by the chronic induction of inflammation caused by contact of the blood with the dialysis membrane. The complement system, a strong mediator of pro-inflammatory networks, is a key contributor to such biomaterial-induced inflammation. Though only evaluated in experimental ex vivo settings, specific targeting of complement activation during hemodialysis has uncovered valuable information that points toward the therapeutic use of complement inhibitors as a means to control the unwelcomed inflammatory responses and consequent pathologies in hemodialysis patients.
Collapse
|
57
|
Qing XY, Zhang CH, Li LL, Ji P, Ma S, Wan HL, Wang ZR, Zou J, Yang SY. Retrieving novel C5aR antagonists using a hybrid ligand-based virtual screening protocol based on SVM classification and pharmacophore models. J Biomol Struct Dyn 2013; 31:215-23. [DOI: 10.1080/07391102.2012.698245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
58
|
Zhu LY, Nie L, Zhu G, Xiang LX, Shao JZ. Advances in research of fish immune-relevant genes: a comparative overview of innate and adaptive immunity in teleosts. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 39:39-62. [PMID: 22504163 DOI: 10.1016/j.dci.2012.04.001] [Citation(s) in RCA: 318] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 03/18/2012] [Accepted: 04/05/2012] [Indexed: 05/31/2023]
Abstract
Fish is considered to be an important model in comparative immunology studies because it is a representative population of lower vertebrates serving as an essential link to early vertebrate evolution. Fish immune-relevant genes have received considerable attention due to its role in improving understanding of both fish immunology and the evolution of immune systems. In this review, we discuss the current understanding of teleost immune-relevant genes for both innate and adaptive immunity, including pattern recognition receptors, antimicrobial peptides, complement molecules, lectins, interferons and signaling factors, inflammatory cytokines, chemokines, adaptive immunity relevant cytokines and negative regulators, major histocompatibility complexes, immunoglobulins, and costimulatory molecules. The implications of these factors on the evolutionary history of immune systems were discussed and a perspective outline of innate and adaptive immunity of teleost fish was described. This review may provide clues on the evolution of the essential defense system in vertebrates.
Collapse
Affiliation(s)
- Lv-yun Zhu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | | | | | | | | |
Collapse
|
59
|
Gál P, Dobó J, Beinrohr L, Pál G, Závodszky P. Inhibition of the Serine Proteases of the Complement System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:23-40. [DOI: 10.1007/978-1-4614-4118-2_2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
60
|
Progress and Trends in Complement Therapeutics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:1-22. [PMID: 22990692 DOI: 10.1007/978-1-4614-4118-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The past few years have proven to be a highly successful and exciting period for the field of complement-directed drug discovery and development. Driven by promising experiences with the first marketed complement drugs, increased knowledge about the involvement of complement in health and disease, and improvements in structural and analytical techniques as well as animal models of disease, the field has seen a surge in creative approaches to therapeutically intervene at various stages of the cascade. An impressive panel of compounds that show promise in clinical trials is meanwhile being lined up in the pipelines of both small biotechnology and big pharmaceutical companies. Yet with this new focus on complement-targeted therapeutics, important questions concerning target selection, point and length of intervention, safety, and drug delivery emerge. In view of the diversity of the clinical disorders involving abnormal complement activity or regulation, which include both acute and chronic diseases and affect a wide range of organs, diverse yet specifically tailored therapeutic approaches may be needed to shift complement back into balance. This chapter highlights the key changes in the field that shape our current perception of complement-targeted drugs and provides a brief overview of recent strategies and emerging trends. Selected examples of complement-related diseases and inhibitor classes are highlighted to illustrate the diversity and creativity in field.
Collapse
|
61
|
Gao HW, Zhang MM, Liu YL, Xu QM, Yang SL. Anticomplement activity of ginsenosides from Panax ginseng. J Funct Foods 2013. [DOI: 10.1016/j.jff.2012.09.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
62
|
Yang J, Ahn HN, Chang M, Narasimhan P, Chan PH, Song YS. Complement component 3 inhibition by an antioxidant is neuroprotective after cerebral ischemia and reperfusion in mice. J Neurochem 2012. [PMID: 23199288 DOI: 10.1111/jnc.12111] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Oxidative stress after stroke is associated with the inflammatory system activation in the brain. The complement cascade, especially the degradation products of complement component 3, is a key inflammatory mediator of cerebral ischemia. We have shown that pro-inflammatory complement component 3 is increased by oxidative stress after ischemic stroke in mice using DNA array. In this study, we investigated whether up-regulation of complement component 3 is directly related to oxidative stress after transient focal cerebral ischemia in mice and oxygen-glucose deprivation in brain cells. Persistent up-regulation of complement component 3 expression was reduced in copper/zinc-superoxide dismutase transgenic mice, and manganese-superoxide dismutase knock-out mice showed highly increased complement component 3 levels after transient focal cerebral ischemia. Antioxidant N-tert-butyl-α-phenylnitrone treatment suppressed complement component 3 expression after transient focal cerebral ischemia. Accumulation of complement component 3 in neurons and microglia was decreased by N-tert-butyl-α-phenylnitrone, which reduced infarct volume and impaired neurological deficiency after cerebral ischemia and reperfusion in mice. Small interfering RNA specific for complement component 3 transfection showed a significant increase in brain cells viability after oxygen-glucose deprivation. Our study suggests that the neuroprotective effect of antioxidants through complement component 3 suppression is a new strategy for potential therapeutic approaches in stroke.
Collapse
Affiliation(s)
- Jiwon Yang
- Department of Pharmacology, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
63
|
Abe T, Hosur KB, Hajishengallis E, Reis ES, Ricklin D, Lambris JD, Hajishengallis G. Local complement-targeted intervention in periodontitis: proof-of-concept using a C5a receptor (CD88) antagonist. THE JOURNAL OF IMMUNOLOGY 2012; 189:5442-8. [PMID: 23089394 DOI: 10.4049/jimmunol.1202339] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
When excessively activated or deregulated, complement becomes a major link between infection and inflammatory pathology including periodontitis. This oral inflammatory disease is associated with a dysbiotic microbiota, leads to the destruction of bone and other tooth-supporting structures, and exerts an adverse impact on systemic health. We have previously shown that mice deficient either in complement C5a receptor (C5aR; CD88) or TLR2 are highly and similarly resistant to periodontitis, suggesting that a cross-talk between the two receptors may be involved in the disease process. In this paper, we show that C5aR and TLR2 indeed synergize for maximal inflammatory responses in the periodontal tissue and uncover a novel pharmacological target to abrogate periodontitis. Using two different mouse models of periodontitis, we show that local treatments with a C5aR antagonist inhibited periodontal inflammation through downregulation of TNF, IL-1β, IL-6, and IL-17 and further protected against bone loss, regardless of the presence of TLR2. These findings not only reveal a crucial cooperation between C5aR and TLR2 in periodontal inflammation but also provide proof-of-concept for local targeting of C5aR as a powerful candidate for the treatment of human periodontitis.
Collapse
Affiliation(s)
- Toshiharu Abe
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
64
|
Reis ES, Chen H, Sfyroera G, Monk PN, Köhl J, Ricklin D, Lambris JD. C5a receptor-dependent cell activation by physiological concentrations of desarginated C5a: insights from a novel label-free cellular assay. THE JOURNAL OF IMMUNOLOGY 2012; 189:4797-805. [PMID: 23041570 DOI: 10.4049/jimmunol.1200834] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The complement anaphylatoxins C3a, C5a, and desarginated C5a (C5a(desArg)) play critical roles in the induction of inflammation and the modulation of innate and acquired immune responses after binding to their G protein-coupled receptors, C3a receptor and C5a receptor (C5aR). The role of C5a(desArg) in inducing cell activation has been often neglected, because the affinity of C5a(desArg) for C5aR has been reported to be much lower than that of C5a. We have used a novel label-free cellular assay to reassess the potential of C5a(desArg) to induce activation of transfected and primary immune cells. Our results indicate that physiological levels of C5a(desArg) induce significant levels of cell activation that are even higher than those achieved by stimulating cells with analogous concentrations of C5a. Such activation was strictly dependent on C5aR, because it was completely abrogated by PMX-53, a C5aR antagonist. Pharmacological inhibition of specific G proteins located downstream of C5aR indicated differential involvement of G(α) proteins upon C5aR engagement by C5a or C5a(desArg). Further, mass spectrometric characterization of plasma-derived C5a and C5a(desArg) provided important insight into the posttranslational modification pattern of these anaphylatoxins, which includes glycosylation at Asn(64) and partial cysteinylation at Cys(27). Although the context-specific physiological contribution of C5a(desArg) has to be further explored, our data suggest that C5a(desArg) acts as a key molecule in the triggering of local inflammation as well as the maintenance of blood surveillance and homeostatic status.
Collapse
Affiliation(s)
- Edimara S Reis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
65
|
Abstract
The complement cascade is a major contributor to the innate immune response. It has now been well accepted that complement plays a critical role in hyperacute rejection and acute antibody-mediated rejection of transplanted organ. There is also increasing evidence that complement proteins contribute to the pathogenesis of organ ischemia-reperfusion injury, and even to cell-mediated rejection. Furthermore, the chemoattractants C3a and C5a and the terminal membrane attack complex that are generated by complement activation can directly or indirectly mediate tissue injury and trigger adaptive immune responses. Here, we review recent findings concerning the role of complement in graft ischemia-reperfusion injury, antibody-mediated rejection and accommodation, and cell-mediated rejection. We also discuss the current status of complement intervention therapies in clinical transplantation and describe potential new therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | |
Collapse
|
66
|
Vlam L, van den Berg LH, Cats EA, Piepers S, van der Pol WL. Immune pathogenesis and treatment of multifocal motor neuropathy. J Clin Immunol 2012; 33 Suppl 1:S38-42. [PMID: 22941513 DOI: 10.1007/s10875-012-9779-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 08/22/2012] [Indexed: 11/24/2022]
Abstract
Multifocal motor neuropathy (MMN) is a rare, probably immune-mediated chronic disorder characterized by asymmetric distal limb weakness and conduction block. The exact pathogenesis of MMN is still unclear, but IgM anti-GM1 antibodies, which can be detected in sera from approximately half of all MMN patients, are thought to play an important role. Treatment with intravenous immunoglobulin (IVIG) is effective in the vast majority of patients, but, despite IVIG maintenance treatment, many patients experience a slowly progressive decline in muscle strength. In this review we will summarize the results from studies on pathogenesis. We will discuss current treatment strategies of MMN and how insight into MMN pathogenesis may translate into novel therapies in the future.
Collapse
Affiliation(s)
- Lotte Vlam
- Rudolf Magnus Institute of Neuroscience, Department of Neurology G 03.228, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands
| | | | | | | | | |
Collapse
|
67
|
Larghi EL, Operto MA, Torres R, Kaufman TS. Synthesis and classical pathway Complement inhibitory activity of C7-functionalized filifolinol derivatives, inspired in K-76 COOH. Eur J Med Chem 2012; 55:74-84. [DOI: 10.1016/j.ejmech.2012.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 06/29/2012] [Accepted: 07/02/2012] [Indexed: 11/27/2022]
|
68
|
Kong CJ, Huang ZA, Chen J, Shi YH, Lu XJ. [Molecular cloning, sequence analysis and expression of ayu complement component C9 gene]. DONG WU XUE YAN JIU = ZOOLOGICAL RESEARCH 2012; 33:151-7. [PMID: 22467389 DOI: 10.3724/sp.j.1141.2012.02151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
C9, a component of the membrane attack complex, participates in the final stage of the complement cascade which lyses foreign organisms by disrupting the integrity of their cell membranes. In the present study, a full-length ayu C9 (aC9) cDNA was cloned which contains 2,125 nucleotides and encodes a protein of 592 amino acids. A signal peptide was deposited in the N-terminal 22 residues. The deduced amino acid sequence of aC9 showed 56.8% identity to the C9 of rainbow trout, and 40.9% to 53.8% identity to the C9 of other teleosts. RT-PCR analysis demonstrated that the mRNA of aC9 was expressed in the liver, spleen, intestine, gill and muscle of healthy ayu fish with the highest level in the liver. Quantitative RT-PCR analysis showed that aC9 transcripts were significantly up-regulated in the liver at 4 h post Listonella anguillarum infection, peaked at 16 h post injection. Western blotting analysis revealed that serum aC9 significantly increased in Listonella anguillarum infected ayu fish. Our results suggested that aC9 may play an important role in fish immune response of anti-bacteria.
Collapse
|
69
|
Microglia, Alzheimer's disease, and complement. Int J Alzheimers Dis 2012; 2012:983640. [PMID: 22957298 PMCID: PMC3432348 DOI: 10.1155/2012/983640] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 05/01/2012] [Accepted: 05/07/2012] [Indexed: 02/02/2023] Open
Abstract
Microglia, the immune cell of the brain, are implicated in cascades leading to neuronal loss and cognitive decline in Alzheimer's disease (AD). Recent genome-wide association studies have indicated a number of risk factors for the development of late-onset AD. Two of these risk factors are an altered immune response and polymorphisms in complement receptor 1. In view of these findings, we discuss how complement signalling in the AD brain and microglial responses in AD intersect. Dysregulation of the complement cascade, either by changes in receptor expression, enhanced activation of different complement pathways or imbalances between complement factor production and complement cascade inhibitors may all contribute to the involvement of complement in AD. Altered complement signalling may reduce the ability of microglia to phagocytose apoptotic cells and clear amyloid beta peptides, modulate the expression by microglia of complement components and receptors, promote complement factor production by plaque-associated cytokines derived from activated microglia and astrocytes, and disrupt complement inhibitor production. The evidence presented here indicates that microglia in AD are influenced by complement factors to adopt protective or harmful phenotypes and the challenge ahead lies in understanding how this can be manipulated to therapeutic advantage to treat late onset AD.
Collapse
|
70
|
Shishido SN, Varahan S, Yuan K, Li X, Fleming SD. Humoral innate immune response and disease. Clin Immunol 2012; 144:142-58. [PMID: 22771788 PMCID: PMC3576926 DOI: 10.1016/j.clim.2012.06.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 06/05/2012] [Accepted: 06/09/2012] [Indexed: 12/27/2022]
Abstract
The humoral innate immune response consists of multiple components, including the naturally occurring antibodies (NAb), pentraxins and the complement and contact cascades. As soluble, plasma components, these innate proteins provide key elements in the prevention and control of disease. However, pathogens and cells with altered self proteins utilize multiple humoral components to evade destruction and promote pathogy. Many studies have examined the relationship between humoral immunity and autoimmune disorders. This review focuses on the interactions between the humoral components and their role in promoting the pathogenesis of bacterial and viral infections and chronic diseases such as atherosclerosis and cancer. Understanding the beneficial and detrimental aspects of the individual components and the interactions between proteins which regulate the innate and adaptive response will provide therapeutic targets for subsequent studies.
Collapse
Affiliation(s)
- Stephanie N Shishido
- Department of Diagnostic Medicine and Pathology, Kansas State University, Manhattan, KS 66506, USA
| | | | | | | | | |
Collapse
|
71
|
Zhang M, Yang XY, Tang W, Groeneveld TWL, He PL, Zhu FH, Li J, Lu W, Blom AM, Zuo JP, Nan FJ. Discovery and Structural Modification of 1-Phenyl-3-(1-phenylethyl)urea Derivatives as Inhibitors of Complement. ACS Med Chem Lett 2012; 3:317-21. [PMID: 24900471 PMCID: PMC4025749 DOI: 10.1021/ml300005w] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 02/26/2012] [Indexed: 11/30/2022] Open
Abstract
A series of 1-phenyl-3-(1-phenylethyl)urea derivatives were identified as novel and potent complement inhibitors through structural modification of the original compound from high-throughput screening. Various analogues (7 and 13-15) were synthesized and identified as complement inhibitors, with the introduction of a five- or six-carbon chain (7c, 7d, 7k, 7l, and 7o) greatly improving their activity. Optimized compound 7l has an excellent inhibition activity with IC50 values as low as 13 nM. We demonstrated that the compound 7l inhibited C9 deposition through the classical, the lectin, and the alternative pathways but had no influence on C3 and C4 depositions.
Collapse
Affiliation(s)
- Mei Zhang
- Department of Chemistry and Institute of Medicinal
Chemistry, East China Normal University, 3663 North Zhong Shan Road, Shanghai 200062, People's Republic
of China
- Chinese National
Center for Drug Screening, State Key Laboratory of Drug Research,
Shanghai Institute of Materia Medica, Chinese Academy
of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech
Park, Shanghai 201203, People's Republic of China
| | - Xiao-Ying Yang
- Laboratory of Immunopharmacology, State Key
Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai
201203, People's Republic of China
| | - Wei Tang
- Laboratory of Immunopharmacology, State Key
Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai
201203, People's Republic of China
| | - Tom W. L. Groeneveld
- Department of Laboratory Medicine, Section of Medical Protein Chemistry, Lund University, S-205 02 Malmö, Sweden
| | - Pei-Lan He
- Laboratory of Immunopharmacology, State Key
Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai
201203, People's Republic of China
| | - Feng-Hua Zhu
- Laboratory of Immunopharmacology, State Key
Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai
201203, People's Republic of China
| | - Jia Li
- Chinese National
Center for Drug Screening, State Key Laboratory of Drug Research,
Shanghai Institute of Materia Medica, Chinese Academy
of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech
Park, Shanghai 201203, People's Republic of China
| | - Wei Lu
- Department of Chemistry and Institute of Medicinal
Chemistry, East China Normal University, 3663 North Zhong Shan Road, Shanghai 200062, People's Republic
of China
| | - Anna M. Blom
- Department of Laboratory Medicine, Section of Medical Protein Chemistry, Lund University, S-205 02 Malmö, Sweden
| | - Jian-Ping Zuo
- Laboratory of Immunopharmacology, State Key
Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai
201203, People's Republic of China
| | - Fa-Jun Nan
- Chinese National
Center for Drug Screening, State Key Laboratory of Drug Research,
Shanghai Institute of Materia Medica, Chinese Academy
of Sciences, 189 Guoshoujing Road, Zhangjiang Hi-Tech
Park, Shanghai 201203, People's Republic of China
| |
Collapse
|
72
|
Du D, Cheng Z, Chen D. A New Unusual Δ 11(12)-Oleane Triterpene and Anti-Complementary Triterpenes from Prunella Vulgaris Spikes. Nat Prod Commun 2012. [DOI: 10.1177/1934578x1200700422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Anti-complementary activity-guided fractionation of the ethanolic extract of Prunella vulgaris spikes led to the isolation of a new, unusual Δ11(12) triterpene, 3β,13β-dihydroxyolic-11-ene-28-oic acid (1), along with thirteen known triterpenes (2-14). The structure of the new compound was established by 1D and 2D NMR spectroscopic analysis. All the isolates were evaluated for their anti-complementary activity against the classical pathway (CP) and alternative pathway (AP). Eight triterpenes (1-8) showed anti-complementary activity against CP and AP, with CH50 and AP50 values of 0.15-0.37 mg/mL, and 0.29-0.53 mg/mL, respectively. Mechanism study using complement-depleted sera showed that oleanolic acid (2) acted selectively on C1q, C5, and C9 components, while 2α-hydroxy oleanolic acid (3) interacted with C1q, C3, C5 and C9, and 2α,3α-dihydroxyolic-12-ene-28-oic acid (4) blocked C1q, C2, C3, C5 and C9.
Collapse
Affiliation(s)
- Dongsheng Du
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Zhihong Cheng
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Daofeng Chen
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| |
Collapse
|
73
|
Zaal A, Lissenberg-Thunnissen SN, van Schijndel G, Wouters D, van Ham SM, ten Brinke A. Crosstalk between Toll like receptors and C5a receptor in human monocyte derived DCs suppress inflammatory cytokine production. Immunobiology 2012; 218:175-80. [PMID: 22559913 DOI: 10.1016/j.imbio.2012.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/21/2012] [Accepted: 02/26/2012] [Indexed: 01/25/2023]
Abstract
The complement anaphylatoxin, C5a has been implicated in regulation of adaptive immune responses through modulation of APC function as shown mainly in studies in mice. C5a was shown to enhance cytokine production in immature DCs, but the effect of C5a on DC function during DC activation has not been elucidated in human. In this study we investigated the effect of C5a on human monocyte derived DCs when simultaneously stimulated with TLR ligands. While C5a indeed enhanced cytokine production of immature DCs, the addition of C5a inhibited production of IL-12, IL-23 and TNFα induced by various TLR ligands such as LPS, R848 and Pam(3)CSK(4). The inhibitory effect of C5a on LPS induced IL-6 production was less pronounced and LPS induced IL-10 was not affected at all. This indicates that C5aR signaling has a differential effect on human DC differentiation depending on the crosstalk with other receptors. Furthermore we found that C5a affects the LPS induced cytokines in a small time frame, and requires almost concurrent signaling of C5a receptor and TLR4. These data emphasize the complexity of DC regulation by anaphylatoxins. While complement activation may provide proinflammatory signals to immature DCs in the absence of pathogens, the same products may serve to downmodulate or deviate immune responses upon combat against infections. These context depending effects of anaphylatoxins on immune responses may have important implications for the emerging use of complement inhibitors in clinical practice.
Collapse
Affiliation(s)
- Anouk Zaal
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
74
|
Neher MD, Weckbach S, Flierl MA, Huber-Lang MS, Stahel PF. Molecular mechanisms of inflammation and tissue injury after major trauma--is complement the "bad guy"? J Biomed Sci 2011; 18:90. [PMID: 22129197 PMCID: PMC3247859 DOI: 10.1186/1423-0127-18-90] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 11/30/2011] [Indexed: 02/07/2023] Open
Abstract
Trauma represents the leading cause of death among young people in industrialized countries. Recent clinical and experimental studies have brought increasing evidence for activation of the innate immune system in contributing to the pathogenesis of trauma-induced sequelae and adverse outcome. As the "first line of defense", the complement system represents a potent effector arm of innate immunity, and has been implicated in mediating the early posttraumatic inflammatory response. Despite its generic beneficial functions, including pathogen elimination and immediate response to danger signals, complement activation may exert detrimental effects after trauma, in terms of mounting an "innocent bystander" attack on host tissue. Posttraumatic ischemia/reperfusion injuries represent the classic entity of complement-mediated tissue damage, adding to the "antigenic load" by exacerbation of local and systemic inflammation and release of toxic mediators. These pathophysiological sequelae have been shown to sustain the systemic inflammatory response syndrome after major trauma, and can ultimately contribute to remote organ injury and death. Numerous experimental models have been designed in recent years with the aim of mimicking the inflammatory reaction after trauma and to allow the testing of new pharmacological approaches, including the emergent concept of site-targeted complement inhibition. The present review provides an overview on the current understanding of the cellular and molecular mechanisms of complement activation after major trauma, with an emphasis of emerging therapeutic concepts which may provide the rationale for a "bench-to-bedside" approach in the design of future pharmacological strategies.
Collapse
Affiliation(s)
- Miriam D Neher
- Department of Orthopaedic Surgery, University of Colorado Denver, School of Medicine, Denver Health Medical Center, 777 Bannock Street, Denver, CO 80204, USA
| | | | | | | | | |
Collapse
|
75
|
Ekdahl KN, Lambris JD, Elwing H, Ricklin D, Nilsson PH, Teramura Y, Nicholls IA, Nilsson B. Innate immunity activation on biomaterial surfaces: a mechanistic model and coping strategies. Adv Drug Deliv Rev 2011; 63:1042-50. [PMID: 21771620 PMCID: PMC3166435 DOI: 10.1016/j.addr.2011.06.012] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 06/13/2011] [Accepted: 06/25/2011] [Indexed: 01/28/2023]
Abstract
When an artificial biomaterial (e.g., a stent or implantable pump) is exposed to blood, plasma proteins immediately adhere to the surface, creating a new interface between the biomaterial and the blood. The recognition proteins within the complement and contact activation/coagulation cascade systems of the blood will be bound to, or inserted into, this protein film and generate different mediators that will activate polymorphonuclear leukocytes and monocytes, as well as platelets. Under clinical conditions, the ultimate outcome of these processes may be thrombotic and inflammatory reactions, and consequently the composition and conformation of the proteins in the initial layer formed on the surface will to a large extent determine the outcome of a treatment involving the biomaterial, affecting both the functionality of the material and the patient's life quality. This review presents models of biomaterial-induced activation processes and describes various strategies to attenuate potential adverse reactions by conjugating bioactive molecules to surfaces or by introducing nanostructures.
Collapse
Affiliation(s)
- Kristina N Ekdahl
- Dept of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory C5:3, Uppsala University, SE-751 85 Uppsala, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Small vessels, big trouble in the kidneys and beyond: hematopoietic stem cell transplantation–associated thrombotic microangiopathy. Blood 2011; 118:1452-62. [DOI: 10.1182/blood-2011-02-321315] [Citation(s) in RCA: 235] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Abstract
Transplantation-associated thrombotic microangiopathy (TA-TMA) is a challenging diagnosis after hematopoietic stem cell transplantation. Although endothelial injury represents the final common pathway of disease, the exact pathophysiology of TA-TMA remains unclear. Potential causes include infections, chemotherapy, radiation, and calcineurin inhibitors. Recent literature addresses the roles of cytokines, graft-versus-host disease, the coagulation cascade, and complement in the pathogenesis of TA-TMA. Current diagnostic criteria are unsatisfactory, because patients who have received a transplant can have multiple other reasons for the laboratory abnormalities currently used to diagnose TA-TMA. Moreover, our lack of understanding of the exact mechanism of disease limits the development and evaluation of potential treatments. Short- and long-term renal complications contribute to TA-TMA's overall poor prognosis. In light of these challenges, future research must validate novel markers of disease to aid in early diagnosis, guide current and future treatments, prevent long-term morbidity, and improve outcomes. We focus on TA-TMA as a distinct complication of hematopoietic stem cell transplantation, emphasizing the central role of the kidney in this disease.
Collapse
|
77
|
Oikonomopoulou K, Ricklin D, Ward PA, Lambris JD. Interactions between coagulation and complement--their role in inflammation. Semin Immunopathol 2011; 34:151-65. [PMID: 21811895 DOI: 10.1007/s00281-011-0280-x] [Citation(s) in RCA: 312] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 07/21/2011] [Indexed: 12/11/2022]
Abstract
The parallel expression of activation products of the coagulation, fibrinolysis, and complement systems has long been observed in both clinical and experimental settings. Several interconnections between the individual components of these cascades have also been described, and the list of shared regulators is expanding. The co-existence and interplay of hemostatic and inflammatory mediators in the same microenvironment typically ensures a successful host immune defense in compromised barrier settings. However, dysregulation of the cascade activities or functions of inhibitors in one or both systems can result in clinical manifestations of disease, such as sepsis, systemic lupus erythematosus, or ischemia-reperfusion injury, with critical thrombotic and/or inflammatory complications. An appreciation of the precise relationship between complement activation and thrombosis may facilitate the development of novel therapeutics, as well as improve the clinical management of patients with thrombotic conditions that are characterized by complement-associated inflammatory responses.
Collapse
Affiliation(s)
- Katerina Oikonomopoulou
- Department of Pathology & Laboratory Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6100, USA
| | | | | | | |
Collapse
|
78
|
Hartung HP, Lehmann HC, Kieseier BC, Hughes RAC. Novel treatment for immune neuropathies on the horizon. J Peripher Nerv Syst 2011; 16:75-83. [DOI: 10.1111/j.1529-8027.2011.00334.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
79
|
Subramanian H, Kashem SW, Collington SJ, Qu H, Lambris JD, Ali H. PMX-53 as a dual CD88 antagonist and an agonist for Mas-related gene 2 (MrgX2) in human mast cells. Mol Pharmacol 2011; 79:1005-13. [PMID: 21441599 DOI: 10.1124/mol.111.071472] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Human mast cells express the G protein coupled receptor (GPCR) for C5a (CD88). Previous studies indicated that C5a could cause mast cell degranulation, at least in part, via a mechanism similar to that proposed for basic neuropeptides such as substance P, possibly involving Mas-related gene 2 (MrgX2). We therefore sought to more clearly define the receptor specificity for C5a-induced mast cell degranulation. We found that LAD2, a human mast cell line, and CD34(+) cell-derived primary mast cells express functional MrgX1 and MrgX2 but the immature human mast cell line HMC-1 does not. A potent CD88 antagonist, PMX-53 (10 nM) inhibited C5a-induced Ca(2+) mobilization in HMC-1 cells, but at higher concentrations (≥30 nM) it caused degranulation in LAD2 mast cells, CD34(+) cell-derived mast cells, and RBL-2H3 cells stably expressing MrgX2. PMX-53 did not, however, activate RBL-2H3 cells expressing MrgX1. Although C5a induced degranulation in LAD2 and CD34(+) cell-derived mast cells, it did not activate RBL-2H3 cells expressing MrgX1 or MrgX2. Replacement of Trp with Ala and Arg with dArg abolished the ability of PMX-53 to inhibit C5a-induced Ca(2+) mobilization in HMC-1 cells and to cause degranulation in RBL-2H3 cells expressing MrgX2. These findings demonstrate that C5a does not use MrgX1 or MrgX2 for mast cell degranulation. Moreover, it reveals the novel finding that PMX-53 functions as a potent CD88 antagonist and a low-affinity agonist for MrgX2. Furthermore, Trp and Arg residues are required for the ability of PMX53 to act as both a CD88 antagonist and a MrgX2 agonist.
Collapse
Affiliation(s)
- Hariharan Subramanian
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104-6030, USA
| | | | | | | | | | | |
Collapse
|
80
|
Thurman JM, Renner B. Dynamic control of the complement system by modulated expression of regulatory proteins. J Transl Med 2011; 91:4-11. [PMID: 20921948 PMCID: PMC3109904 DOI: 10.1038/labinvest.2010.173] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The complement system serves many biological functions, including the eradication of invasive pathogens and the removal of damaged cells and immune-complexes. Uncontrolled complement activation causes injury to host cells, however, so adequate regulation of the system is essential. Control of the complement system is maintained by a group of cell surface and circulating proteins referred to as complement regulatory proteins. The expression of the cell surface complement regulatory proteins varies from tissue to tissue. Furthermore, specific cell types can upregulate or downregulate the expression of these proteins in response to a variety of signals or insults. Altered regulation of the complement regulatory proteins can have important effects on local complement activation. In some circumstances this can be beneficial, such as in the setting of certain infections. In other circumstances, however, this can be a cause of complement-mediated injury of the tissue. A full understanding of the mechanisms by which the complement system is modulated at the local level can have important implications for how we diagnose and treat a wide range of inflammatory diseases.
Collapse
|
81
|
Qu H, Magotti P, Ricklin D, Wu EL, Kourtzelis I, Wu YQ, Kaznessis YN, Lambris JD. Novel analogues of the therapeutic complement inhibitor compstatin with significantly improved affinity and potency. Mol Immunol 2010; 48:481-9. [PMID: 21067811 DOI: 10.1016/j.molimm.2010.10.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 10/10/2010] [Indexed: 11/26/2022]
Abstract
Compstatin is a 13-residue disulfide-bridged peptide that inhibits a key step in the activation of the human complement system. Compstatin and its derivatives have shown great promise for the treatment of many clinical disorders associated with unbalanced complement activity. To obtain more potent compstatin analogues, we have now performed an N-methylation scan of the peptide backbone and amino acid substitutions at position 13. One analogue (Ac-I[CVW(Me)QDW-Sar-AHRC](NMe)I-NH(2)) displayed a 1000-fold increase in both potency (IC(50) = 62 nM) and binding affinity for C3b (K(D) = 2.3 nM) over that of the original compstatin. Biophysical analysis using surface plasmon resonance and isothermal titration calorimetry suggests that the improved binding originates from more favorable free conformation and stronger hydrophobic interactions. This study provides a series of significantly improved drug leads for therapeutic applications in complement-related diseases, and offers new insights into the structure-activity relationships of compstatin analogues.
Collapse
Affiliation(s)
- Hongchang Qu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Abstract
The complement system consists of a tightly regulated network of proteins that play an important role in host defense and inflammation. Complement activation results in opsonization of pathogens and their removal by phagocytes, as well as cell lysis. Inappropriate complement activation and complement deficiencies are the underlying cause of the pathophysiology of many diseases such as systemic lupus erythematosus and asthma. This review represents an overview of the complement system in an effort to understand the beneficial as well as harmful roles it plays during inflammatory responses.
Collapse
Affiliation(s)
- J Vidya Sarma
- Department of Pathology, The University of Michigan Medical School, 1301 Catherine Rd., Box 5602, Ann Arbor, MI 48109-5602, USA
| | | |
Collapse
|
83
|
Kocsis A, Kékesi KA, Szász R, Végh BM, Balczer J, Dobó J, Závodszky P, Gál P, Pál G. Selective inhibition of the lectin pathway of complement with phage display selected peptides against mannose-binding lectin-associated serine protease (MASP)-1 and -2: significant contribution of MASP-1 to lectin pathway activation. THE JOURNAL OF IMMUNOLOGY 2010; 185:4169-78. [PMID: 20817870 DOI: 10.4049/jimmunol.1001819] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The complement system, an essential part of the innate immune system, can be activated through three distinct routes: the classical, the alternative, and the lectin pathways. The contribution of individual activation pathways to different biological processes can be assessed by using pathway-selective inhibitors. In this paper, we report lectin pathway-specific short peptide inhibitors developed by phage display against mannose-binding lectin-associated serine proteases (MASPs), MASP-1 and MASP-2. On the basis of the selected peptide sequences, two 14-mer peptides, designated as sunflower MASP inhibitor (SFMI)-1 and SFMI-2, were produced and characterized. SFMI-1 inhibits both MASP-1 and MASP-2 with a K(I) of 65 and 1030 nM, respectively, whereas SFMI-2 inhibits only MASP-2 with a K(I) of 180 nM. Both peptides block the lectin pathway activation completely while leaving the classical and the alternative routes intact and fully functional, demonstrating that of all complement proteases only MASP-1 and/or MASP-2 are inhibited by these peptides. In a C4 deposition inhibitor assay using preactivated MASP-2, SFMI-2 is 10-fold more effective than SFMI-1 in accordance with the fact that SFMI-2 is a more potent inhibitor of MASP-2. Surprisingly, however, out of the two peptides, SFMI-1 is much more effective in preventing C3 and C4 deposition when normal human serum containing zymogen MASPs is used. This suggests that MASP-1 has a crucial role in the initiation steps of lectin pathway activation most probably by activating MASP-2. Because the lectin pathway has been implicated in several life-threatening pathological states, these inhibitors should be considered as lead compounds toward developing lectin pathway blocking therapeutics.
Collapse
Affiliation(s)
- Andrea Kocsis
- Institute of Enzymology, Biological Research Center, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Ferreira VP, Pangburn MK, Cortés C. Complement control protein factor H: the good, the bad, and the inadequate. Mol Immunol 2010; 47:2187-97. [PMID: 20580090 DOI: 10.1016/j.molimm.2010.05.007] [Citation(s) in RCA: 301] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The complement system is an essential component of the innate immune system that participates in elimination of pathogens and altered host cells and comprises an essential link between the innate and adaptive immune system. Soluble and membrane-bound complement regulators protect cells and tissues from unintended complement-mediated injury. Complement factor H is a soluble complement regulator essential for controlling the alternative pathway in blood and on cell surfaces. Normal recognition of self-cell markers (i.e. polyanions) and C3b/C3d fragments is necessary for factor H function. Inadequate recognition of host cell surfaces by factor H due to mutations and polymorphisms have been associated with complement-mediated tissue damage and disease. On the other hand, unwanted recognition of pathogens and altered self-cells (i.e. cancer) by factor H is used as an immune evasion strategy. This review will focus on the current knowledge related to these versatile recognition properties of factor H.
Collapse
Affiliation(s)
- Viviana P Ferreira
- Department of Medical Microbiology and Immunology, College of Medicine, University of Toledo, Toledo, OH 43614, United States.
| | | | | |
Collapse
|
85
|
Fisette A, Cianflone K. The ASP and C5L2 pathway: another bridge between inflammation and metabolic homeostasis. ACTA ACUST UNITED AC 2010. [DOI: 10.2217/clp.10.21] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
86
|
Quan GH, Oh SR, Kim JH, Lee HK, Kinghorn AD, Chin YW. Xanthone constituents of the fruits of garcinia mangostana
with anticomplement activity. Phytother Res 2010; 24:1575-7. [DOI: 10.1002/ptr.3177] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
87
|
Abstract
Thrombosis is a common complication of end-stage renal disease, particularly in patients on hemodialysis. Although substantial progress has been made in preventing thrombotic complications in various other groups of patients, the mechanisms of thrombosis during hemodialysis require clarification. In this report, we demonstrate that complement activation triggered by hemodialysis biomaterials, and the subsequent generation of the complement anaphylatoxin C5a, results in the expression of functionally active tissue factor (TF) in peripheral blood neutrophils. Because TF is a key initiator of coagulation in vivo, we postulate that the recurring complement activation that occurs during long-term hemodialysis contributes to thrombosis in dialyzed end-stage renal disease patients. Furthermore, we found that complement contributed to the induction of granulocyte colony-stimulating factor, which has been implicated in the pathogenesis of thrombosis in patients treated with the recombinant form of this molecule. Importantly, the inhibition of complement activation attenuated the TF expression and granulocyte colony-stimulating factor induction in blood passing through a hemodialysis circuit, suggesting that the complement system could become a new therapeutic target for preventing thrombosis in patients with chronic renal failure who are maintained on hemodialysis.
Collapse
|