1
|
Zhang T, Li Z, He A, Zhou W, Zhu X, Song Y. Clinical Significance and Potential Function of Complement Factor D in Acute Myeloid Leukemia. Cureus 2024; 16:e67260. [PMID: 39310420 PMCID: PMC11414840 DOI: 10.7759/cureus.67260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematologic malignancy characterized by aggressive proliferation and a poor prognosis. The objective of this study is to elucidate the specific role of complement factor D (CFD) in AML, with the aim of identifying robust prognostic markers for the disease. METHODS We performed a systematic investigation on clinical significance and potential function of CFD in AML by using the R Programming Language with The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), The Human Protein Atlas (HPA), The University of ALabama at Birmingham CANcer data analysis Portal (UALCAN), Gene Expression Profiling Interactive Analysis (GEPIA), Kaplan-Meier plotter, Cancer Cell Line Encyclopedia (CCLE) database, and Comprehensive Analysis on Multi-Omics of Immunotherapy in Pan-cancer (CAMOIP) database. The expression of CFD in AML patients was verified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). RESULTS The expression of CFD was the highest in AML cells than in other tumor cell lines. The expression of CFD was also higher in AML patients than in the matched normal group. Compared with the low expression of the CFD group, high expression of CFD predicted better overall survival (OS) and lower tumor mutational burden (TMB) in AML patients. Moreover, a nomogram model based on CFD was successfully constructed to predict the OS of AML patients. Notably, the expression of CFD was associated with drug sensitivity and monocyte cell infiltration. CONCLUSION CFD could serve as a potential OS prognostic biomarker and guide clinical treatment for AML.
Collapse
Affiliation(s)
- Taigang Zhang
- Department of Clinical Laboratory, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, CHN
| | - Zhaozhong Li
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, CHN
| | - Aoyu He
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, CHN
| | - Wenjuan Zhou
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, CHN
| | - Xianjin Zhu
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, CHN
| | - Yanfang Song
- Department of Clinical Laboratory, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, CHN
| |
Collapse
|
2
|
Lenz M, Schönbauer R, Stojkovic S, Lichtenauer M, Paar V, Gatterer C, Schukro C, Emich M, Fritzer-Szekeres M, Strametz-Juranek J, Sponder M. Long-term physical activity modulates adipsin and ANGPTL4 serum levels, a potential link to exercise-induced metabolic changes. Panminerva Med 2023; 65:292-302. [PMID: 34309331 DOI: 10.23736/s0031-0808.21.04382-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Within the presented prospective study, we aimed to illuminate the effect of long-term physical exercise on serum levels of adipsin (complement factor D) and angiopoietin-like 4 (ANGPTL4). Although past studies already outlined the effects of acute exercise, our trial design aimed to depict the development under long-term physical activity conditions. METHODS Ninety-eight participants were included in the study and were asked to perform eight months of moderate physical activity for at least 150 minutes/week and/or vigorous-intensity exercise for at least 75 minutes/week. According to initial performance and performance gain throughout the study period, four groups were formed and subsequently compared. Blood sampling for the determination of routine laboratory parameters was done at baseline, after 2, 6, and 8 months. Additionally, adipsin and ANGPTL4 serum levels were concurrently quantified using commercially available ELISA kits. RESULTS The study cohort consisted of 61.2% male participants with an average age of 49.3±6.7 years. Adipsin and ANGPTL4 were found to be strongly increased by long-term physical exercise. Participants displaying a performance gain of >2.9% throughout the study showed significantly increased serum levels of both biomarkers. CONCLUSIONS Serum levels of adipsin and ANGPTL4 were closely tied to the individual performance gain of the participating probands. An association of adipsin levels, initial performance, and serum triglycerides was found at baseline. Interestingly, this interrelationship was not detectable after eight months of physical training. This finding might indicate adipsin's involvement in linking triglyceride-balance to individual performance and energy demands in a homeostatic state.
Collapse
Affiliation(s)
- Max Lenz
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria -
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria -
| | - Robert Schönbauer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Stefan Stojkovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Michael Lichtenauer
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Vera Paar
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Constantin Gatterer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christoph Schukro
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Michael Emich
- Austrian Federal Ministry of Defence, Austrian Armed Forces, Vienna, Austria
| | - Monika Fritzer-Szekeres
- Department of Medical-Chemical Laboratory Analysis, Medical University of Vienna, Vienna, Austria
| | | | - Michael Sponder
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Dani R, Oroszlán G, Martinusz R, Farkas B, Dobos B, Vadas E, Závodszky P, Gál P, Dobó J. Quantification of the zymogenicity and the substrate-induced activity enhancement of complement factor D. Front Immunol 2023; 14:1197023. [PMID: 37283768 PMCID: PMC10239819 DOI: 10.3389/fimmu.2023.1197023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/04/2023] [Indexed: 06/08/2023] Open
Abstract
Complement factor D (FD) is a serine protease present predominantly in the active form in circulation. It is synthesized as a zymogen (pro-FD), but it is continuously converted to FD by circulating active MASP-3. FD is a unique, self-inhibited protease. It has an extremely low activity toward free factor B (FB), while it is a highly efficient enzyme toward FB complexed with C3b (C3bB). The structural basis of this phenomenon is known; however, the rate enhancement was not yet quantified. It has also been unknown whether pro-FD has any enzymatic activity. In this study, we aimed to measure the activity of human FD and pro-FD toward uncomplexed FB and C3bB in order to quantitatively characterize the substrate-induced activity enhancement and zymogenicity of FD. Pro-FD was stabilized in the proenzyme form by replacing Arg25 (precursor numbering) with Gln (pro-FD-R/Q). Activated MASP-1 and MASP-3 catalytic fragments were also included in the study for comparison. We found that the complex formation with C3b enhanced the cleavage rate of FB by FD approximately 20 million-fold. C3bB was also a better substrate for MASP-1, approximately 100-fold, than free FB, showing that binding to C3b renders the scissile Arg-Lys bond in FB to become more accessible for proteolysis. Though easily measurable, this cleavage by MASP-1 is not relevant physiologically. Our approach provides quantitative data for the two-step mechanism characterized by the enhanced susceptibility of FB for cleavage upon complex formation with C3b and the substrate-induced activity enhancement of FD upon its binding to C3bB. Earlier MASP-3 was also implicated as a potential FB activator; however, MASP-3 does not cleave C3bB (or FB) at an appreciable rate. Finally, pro-FD cleaves C3bB at a rate that could be physiologically significant. The zymogenicity of FD is approximately 800, i.e., the cleavage rate of C3bB by pro-FD-R/Q was found to be approximately 800-fold lower than that by FD. Moreover, pro-FD-R/Q at approximately 50-fold of the physiological FD concentration could restore half-maximal AP activity of FD-depleted human serum on zymosan. The observed zymogen activity of pro-FD might be relevant in MASP-3 deficiency cases or during therapeutic MASP-3 inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
4
|
Pouille CL, Ouaza S, Roels E, Behra J, Tourret M, Molinié R, Fontaine JX, Mathiron D, Gagneul D, Taminiau B, Daube G, Ravallec R, Rambaud C, Hilbert JL, Cudennec B, Lucau-Danila A. Chicory: Understanding the Effects and Effectors of This Functional Food. Nutrients 2022; 14:957. [PMID: 35267932 PMCID: PMC8912540 DOI: 10.3390/nu14050957] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Industrial chicory has been the subject of numerous studies, most of which provide clinical observations on its health effects. Whether it is the roasted root, the flour obtained from the roots or the different classes of molecules that enter into the composition of this plant, understanding the molecular mechanisms of action on the human organism remains incomplete. In this study, we were interested in three molecules or classes of molecules present in chicory root: fructose, chlorogenic acids, and sesquiterpene lactones. We conducted experiments on the murine model and performed a nutrigenomic analysis, a metabolic hormone assay and a gut microbiota analysis, associated with in vitro observations for different responses. We have highlighted a large number of effects of all these classes of molecules that suggest a pro-apoptotic activity, an anti-inflammatory, antimicrobial, antioxidant, hypolipidemic and hypoglycemic effect and also an important role in appetite regulation. A significant prebiotic activity was also identified. Fructose seems to be the most involved in these activities, contributing to approximately 83% of recorded responses, but the other classes of tested molecules have shown a specific role for these different effects, with an estimated contribution of 23-24%.
Collapse
Affiliation(s)
- Céline L. Pouille
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Souad Ouaza
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Elise Roels
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Josette Behra
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
| | - Melissa Tourret
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
| | - Roland Molinié
- UMR Transfontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—BIOlogie des Plantes et Innovation (BIOPI), 80025 Amiens, France; (R.M.); (J.-X.F.)
| | - Jean-Xavier Fontaine
- UMR Transfontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—BIOlogie des Plantes et Innovation (BIOPI), 80025 Amiens, France; (R.M.); (J.-X.F.)
| | - David Mathiron
- Plateforme Analytique UFR des Sciences, UPJV, Bâtiment Serres-Transfert Rue Dallery-Passage du Sourire d’Avril, 80039 Amiens, France;
| | - David Gagneul
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Bernard Taminiau
- Department of Food Sciences–Microbiology, FARAH, University of Liege, 4000 Liege, Belgium; (B.T.); (G.D.)
| | - Georges Daube
- Department of Food Sciences–Microbiology, FARAH, University of Liege, 4000 Liege, Belgium; (B.T.); (G.D.)
| | - Rozenn Ravallec
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
| | - Caroline Rambaud
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Jean-Louis Hilbert
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Benoit Cudennec
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
| | - Anca Lucau-Danila
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| |
Collapse
|
5
|
Complement component C3: A structural perspective and potential therapeutic implications. Semin Immunol 2022; 59:101627. [PMID: 35760703 PMCID: PMC9842190 DOI: 10.1016/j.smim.2022.101627] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 01/18/2023]
Abstract
As the most abundant component of the complement system, C3 and its proteolytic derivatives serve essential roles in the function of all three complement pathways. Central to this is a network of protein-protein interactions made possible by the sequential proteolysis and far-reaching structural changes that accompany C3 activation. Beginning with the crystal structures of C3, C3b, and C3c nearly twenty years ago, the physical transformations underlying C3 function that had long been suspected were finally revealed. In the years that followed, a compendium of crystallographic information on C3 derivatives bound to various enzymes, regulators, receptors, and inhibitors generated new levels of insight into the structure and function of the C3 molecule. This Review provides a concise classification, summary, and interpretation of the more than 50 unique crystal structure determinations for human C3. It also highlights other salient features of C3 structure that were made possible through solution-based methods, including Hydrogen/Deuterium Exchange and Small Angle X-ray Scattering. At this pivotal time when the first C3-targeted therapeutics begin to see use in the clinic, some perspectives are also offered on how this continually growing body of structural information might be leveraged for future development of next-generation C3 inhibitors.
Collapse
|
6
|
Mizuno M, Khaledian B, Maeda M, Hayashi T, Mizuno S, Munetsuna E, Watanabe T, Kono S, Okada S, Suzuki M, Takao S, Minami H, Asai N, Sugiyama F, Takahashi S, Shimono Y. Adipsin-Dependent Secretion of Hepatocyte Growth Factor Regulates the Adipocyte-Cancer Stem Cell Interaction. Cancers (Basel) 2021; 13:4238. [PMID: 34439392 PMCID: PMC8393397 DOI: 10.3390/cancers13164238] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 01/18/2023] Open
Abstract
Adipose tissue is a component of the tumor microenvironment and is involved in tumor progression. We have previously shown that adipokine adipsin (CFD) functions as an enhancer of tumor proliferation and cancer stem cell (CSC) properties in breast cancers. We established the Cfd-knockout (KO) mice and the mammary adipose tissue-derived stem cells (mADSCs) from them. Cfd-KO in mADSCs significantly reduced their ability to enhance tumorsphere formation of breast cancer patient-derived xenograft (PDX) cells, which was restored by the addition of Cfd in the culture medium. Hepatocyte growth factor (HGF) was expressed and secreted from mADSCs in a Cfd-dependent manner. HGF rescued the reduced ability of Cfd-KO mADSCs to promote tumorsphere formation in vitro and tumor formation in vivo by breast cancer PDX cells. These results suggest that HGF is a downstream effector of Cfd in mADSCs that enhances the CSC properties in breast cancers.
Collapse
Affiliation(s)
- Masahiro Mizuno
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake 4701192, Japan or (M.M.); (B.K.); (M.M.); (T.H.); (E.M.); (T.W.)
| | - Behnoush Khaledian
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake 4701192, Japan or (M.M.); (B.K.); (M.M.); (T.H.); (E.M.); (T.W.)
| | - Masao Maeda
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake 4701192, Japan or (M.M.); (B.K.); (M.M.); (T.H.); (E.M.); (T.W.)
- Department of Pathology, Fujita Health University School of Medicine, Toyoake 4701192, Japan;
| | - Takanori Hayashi
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake 4701192, Japan or (M.M.); (B.K.); (M.M.); (T.H.); (E.M.); (T.W.)
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba 3058575, Japan; (S.M.); (F.S.); (S.T.)
| | - Eiji Munetsuna
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake 4701192, Japan or (M.M.); (B.K.); (M.M.); (T.H.); (E.M.); (T.W.)
| | - Takashi Watanabe
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake 4701192, Japan or (M.M.); (B.K.); (M.M.); (T.H.); (E.M.); (T.W.)
| | - Seishi Kono
- Division of Breast and Endocrine Surgery, Kobe University Graduate School of Medicine, Kobe 6500017, Japan; (S.K.); (S.T.)
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 8600811, Japan;
| | - Motoshi Suzuki
- Department of Molecular Oncology, Fujita Health University School of Medicine, Toyoake 4701192, Japan;
| | - Shintaro Takao
- Division of Breast and Endocrine Surgery, Kobe University Graduate School of Medicine, Kobe 6500017, Japan; (S.K.); (S.T.)
| | - Hironobu Minami
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe 6500017, Japan;
| | - Naoya Asai
- Department of Pathology, Fujita Health University School of Medicine, Toyoake 4701192, Japan;
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba 3058575, Japan; (S.M.); (F.S.); (S.T.)
| | - Satoru Takahashi
- Laboratory Animal Resource Center, Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba 3058575, Japan; (S.M.); (F.S.); (S.T.)
| | - Yohei Shimono
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake 4701192, Japan or (M.M.); (B.K.); (M.M.); (T.H.); (E.M.); (T.W.)
| |
Collapse
|
7
|
Wiles JA, Galvan MD, Podos SD, Geffner M, Huang M. Discovery and Development of the Oral Complement Factor D Inhibitor Danicopan (ACH-4471). Curr Med Chem 2020; 27:4165-4180. [PMID: 31573880 DOI: 10.2174/0929867326666191001130342] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 03/28/2019] [Accepted: 04/04/2019] [Indexed: 12/29/2022]
Abstract
Complement plays a vital role in our innate immune defense against invasive microorganisms. Excessive complement activation or insufficient control of activation on host cells, however, is associated with several chronic disorders. Essential to the activation and amplification of the Alternative Pathway (AP) of complement, Complement Factor D (CFD) is a specific serine protease that cleaves its unique substrate, Complement Factor B (CFB) in complex with an activated form of complement component 3 (C3), to generate the AP C3 convertases C3(H2O)Bb and C3bBb. These convertases comprise a central component in eliciting effector responses following AP activation, and they also enable a powerful amplification loop for both the Classical Pathway (CP) and Lectin Pathway (LP) of complement. Because CFD is not required for the activation of either the CP or LP, selective CFD inhibition presents a favorable therapeutic approach to modulating complement activity that leaves intact the effector functions following CP and LP activation and thus poses a lower risk of bacterial infection than other complement-directed approaches. This review provides an update on inhibitors of CFD, which have evolved from irreversible small molecules that demonstrate poor selectivity to reversible small molecules and monoclonal antibodies that demonstrate exceptional selectivity and potency. The reversible small-molecule inhibitor danicopan.
Collapse
Affiliation(s)
- Jason A Wiles
- Drug Discovery, Achillion Pharmaceuticals, New Haven, CT 06511, United States
| | - Manuel D Galvan
- Drug Discovery, Achillion Pharmaceuticals, New Haven, CT 06511, United States
| | - Steven D Podos
- Drug Discovery, Achillion Pharmaceuticals, New Haven, CT 06511, United States
| | - Michael Geffner
- Drug Development, Achillion Pharmaceuticals, New Haven, CT 06511, United States
| | - Mingjun Huang
- Drug Discovery, Achillion Pharmaceuticals, New Haven, CT 06511, United States
| |
Collapse
|
8
|
Dobó J, Kocsis A, Gál P. Be on Target: Strategies of Targeting Alternative and Lectin Pathway Components in Complement-Mediated Diseases. Front Immunol 2018; 9:1851. [PMID: 30135690 PMCID: PMC6092519 DOI: 10.3389/fimmu.2018.01851] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/26/2018] [Indexed: 12/20/2022] Open
Abstract
The complement system has moved into the focus of drug development efforts in the last decade, since its inappropriate or uncontrolled activation has been recognized in many diseases. Some of them are primarily complement-mediated rare diseases, such as paroxysmal nocturnal hemoglobinuria, C3 glomerulonephritis, and atypical hemolytic uremic syndrome. Complement also plays a role in various multifactorial diseases that affect millions of people worldwide, such as ischemia reperfusion injury (myocardial infarction, stroke), age-related macular degeneration, and several neurodegenerative disorders. In this review, we summarize the potential advantages of targeting various complement proteins with special emphasis on the components of the lectin (LP) and the alternative pathways (AP). The serine proteases (MASP-1/2/3, factor D, factor B), which are responsible for the activation of the cascade, are straightforward targets of inhibition, but the pattern recognition molecules (mannose-binding lectin, other collectins, and ficolins), the regulatory components (factor H, factor I, properdin), and C3 are also subjects of drug development. Recent discoveries about cross-talks between the LP and AP offer new approaches for clinical intervention. Mannan-binding lectin-associated serine proteases (MASPs) are not just responsible for LP activation, but they are also indispensable for efficient AP activation. Activated MASP-3 has recently been shown to be the enzyme that continuously supplies factor D (FD) for the AP by cleaving pro-factor D (pro-FD). In this aspect, MASP-3 emerges as a novel feasible target for the regulation of AP activity. MASP-1 was shown to be required for AP activity on various surfaces, first of all on LPS of Gram-negative bacteria.
Collapse
Affiliation(s)
- József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andrea Kocsis
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
9
|
Sng CCT, O'Byrne S, Prigozhin DM, Bauer MR, Harvey JC, Ruhle M, Challis BG, Lear S, Roberts LD, Workman S, Janowitz T, Magiera L, Doffinger R, Buckland MS, Jodrell DJ, Semple RK, Wilson TJ, Modis Y, Thaventhiran JED. A type III complement factor D deficiency: Structural insights for inhibition of the alternative pathway. J Allergy Clin Immunol 2018; 142:311-314.e6. [PMID: 29522842 PMCID: PMC6034011 DOI: 10.1016/j.jaci.2018.01.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 01/24/2023]
Affiliation(s)
| | - Sorcha O'Byrne
- Department of Clinical Immunology, Cambridge University Hospitals National Health Service Trust, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Daniil M Prigozhin
- Molecular Immunity Unit, Department of Medicine, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Matthias R Bauer
- Division of Structural Studies, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Jennifer C Harvey
- Department of Immunology, Royal Free London National Health Service Foundation Trust, London, United Kingdom
| | - Michelle Ruhle
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Ben G Challis
- Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Sara Lear
- Department of Clinical Immunology, Cambridge University Hospitals National Health Service Trust, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds Institute of Genetics, Health and Therapeutics (LIGHT) Laboratories, University of Leeds, Leeds, United Kingdom
| | - Sarita Workman
- Department of Immunology, Royal Free London National Health Service Foundation Trust, London, United Kingdom
| | - Tobias Janowitz
- Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Lukasz Magiera
- Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Rainer Doffinger
- Department of Clinical Immunology, Cambridge University Hospitals National Health Service Trust, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Matthew S Buckland
- Department of Immunology, Royal Free London National Health Service Foundation Trust, London, United Kingdom
| | - Duncan J Jodrell
- Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Robert K Semple
- Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom; University of Edinburgh Centre for Cardiovascular Sciences, Queen's Medical Research Institute, Little France Crescent, Edinburgh, United Kingdom
| | | | - Yorgo Modis
- Molecular Immunity Unit, Department of Medicine, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - James E D Thaventhiran
- Cancer Research UK Cambridge Institute, Cambridge, United Kingdom; Department of Clinical Immunology, Cambridge University Hospitals National Health Service Trust, Addenbrooke's Hospital, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom; MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom.
| |
Collapse
|
10
|
Vulpetti A, Ostermann N, Randl S, Yoon T, Mac Sweeney A, Cumin F, Lorthiois E, Rüdisser S, Erbel P, Maibaum J. Discovery and Design of First Benzylamine-Based Ligands Binding to an Unlocked Conformation of the Complement Factor D. ACS Med Chem Lett 2018; 9:490-495. [PMID: 29795765 DOI: 10.1021/acsmedchemlett.8b00104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022] Open
Abstract
Complement Factor D, a serine protease of the S1 family and key component of the alternative pathway amplification loop, represents a promising target for the treatment of several prevalent and rare diseases linked to the innate immune system. Previously reported FD inhibitors have been shown to bind to the FD active site in its self-inhibited conformation characterized by the presence of a salt bridge at the bottom of the S1 pocket between Asp189 and Arg218. We report herein a new set of small-molecule FD ligands that harbor a basic S1 binding moiety directly binding to the carboxylate of Asp189, thereby displacing the Asp189-Arg218 ionic interaction and significantly changing the conformation of the self-inhibitory loop.
Collapse
Affiliation(s)
- Anna Vulpetti
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Nils Ostermann
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Stefan Randl
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Taeyoung Yoon
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Aengus Mac Sweeney
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Frederic Cumin
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Edwige Lorthiois
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Simon Rüdisser
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Paul Erbel
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Jürgen Maibaum
- Novartis Pharma AG, Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| |
Collapse
|
11
|
Koike R, Amemiya T, Horii T, Ota M. Structural changes of homodimers in the PDB. J Struct Biol 2017; 202:42-50. [PMID: 29233747 DOI: 10.1016/j.jsb.2017.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/30/2017] [Accepted: 12/08/2017] [Indexed: 01/25/2023]
Abstract
Protein complexes are involved in various biological phenomena. These complexes are intrinsically flexible, and structural changes are essential to their functions. To perform a large-scale automated analysis of the structural changes of complexes, we combined two original methods. An application, SCPC, compares two structures of protein complexes and decides the match of binding mode. Another application, Motion Tree, identifies rigid-body motions in various sizes and magnitude from the two structural complexes with the same binding mode. This approach was applied to all available homodimers in the Protein Data Bank (PDB). We defined two complex-specific motions: interface motion and subunit-spanning motion. In the former, each subunit of a complex constitutes a rigid body, and the relative movement between subunits occurs at the interface. In the latter, structural parts from distinct subunits constitute a rigid body, providing the relative movement spanning subunits. All structural changes were classified and examined. It was revealed that the complex-specific motions were common in the homodimers, detected in around 40% of families. The dimeric interfaces were likely to be small and flat for interface motion, while large and rugged for subunit-spanning motion. Interface motion was accompanied by a drastic change in contacts at the interface, while the change in the subunit-spanning motion was moderate. These results indicate that the interface properties of homodimers correlated with the type of complex-specific motion. The study demonstrates that the pipeline of SCPC and Motion Tree is useful for the massive analysis of structural change of protein complexes.
Collapse
Affiliation(s)
- Ryotaro Koike
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Takayuki Amemiya
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Tatsuya Horii
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Motonori Ota
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| |
Collapse
|
12
|
Harrison RA. The properdin pathway: an "alternative activation pathway" or a "critical amplification loop" for C3 and C5 activation? Semin Immunopathol 2017; 40:15-35. [PMID: 29167939 DOI: 10.1007/s00281-017-0661-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/30/2017] [Indexed: 11/28/2022]
Abstract
This review is not intended to cover in detail all aspects of the discovery and evolution of our understanding of the "alternative pathway" of complement activation, there are many excellent reviews that do this (see Fearon (CRC Crit Rev Immunol 1:1-32, 1979), Pangburn and Müller-Eberhard (Springer Semin Immunopathol 7:163-192, 1984)), but instead to give sufficient background for current concepts to be put in context. The prevailing textbook view, of components having a primary role as an alternative "pathway" for C3 activation, is challenged, with an argument developed for the primary role of the system being that of providing a surface-dependent amplification loop for both C3 and C5 activation. Whatever the mechanism by which the initial C3b molecule is generated, deposition onto a surface has the potential to target that surface for elimination. Elimination or escape from initial targeting is determined by a sophisticated and highly regulated amplification loop for C3 activation. This viewpoint of the system is then briefly developed to provide a context for therapeutic treatment of disease caused, at least in part, by dysregulated amplification of C3 activation, and to highlight some of the challenges that such therapies will face and need to address.
Collapse
Affiliation(s)
- Richard A Harrison
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
13
|
Yorulmaz Avsar S, Jackman JA, Kim MC, Yoon BK, Hunziker W, Cho NJ. Immobilization Strategies for Functional Complement Convertase Assembly at Lipid Membrane Interfaces. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:7332-7342. [PMID: 28683197 DOI: 10.1021/acs.langmuir.7b01465] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The self-assembly formation of complement convertases-essential biomacromolecular complexes that amplify innate immune responses-is triggered by protein adsorption. Herein, a supported lipid bilayer platform was utilized to investigate the effects of covalent and noncovalent tethering strategies on the self-assembly of alternative pathway C3 convertase components, starting with C3b protein adsorption followed bythe addition of factors B and D. Quartz crystal microbalance-dissipation (QCM-D) experiments measured the real-time kinetics of convertase assembly onto supported lipid bilayers. The results demonstrate that the nature of C3b immobilization onto supported lipid bilayers is a key factor governing convertase assembly. The covalent attachment of C3b to maleimide-functionalized supported lipid bilayers promoted the self-assembly of functional C3 convertase in the membrane-associated state and further enabled successful evaluation of a clinically relevant complement inhibitor, compstatin. By contrast, noncovalent attachment of C3b to negatively charged supported lipid bilayers also permitted C3b protein uptake, albeit membrane-associated C3b did not support convertase assembly in this case. Taken together, the findings in this work demonstrate that the attachment scheme for immobilizing C3b protein at lipid membrane interfaces is critical for downstream C3 convertase assembly, thereby offering guidance for the design and evaluation of membrane-associated biomacromolecular complexes.
Collapse
Affiliation(s)
- Saziye Yorulmaz Avsar
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research , Singapore 138673, Singapore
| | - Joshua A Jackman
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
| | - Min Chul Kim
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
| | - Bo Kyeong Yoon
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
| | - Walter Hunziker
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research , Singapore 138673, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore 117599, Singapore
- Singapore Eye Research Institute , Singapore 169856, Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, Singapore 639798 Singapore
- Centre for Biomimetic Sensor Science, Nanyang Technological University , 50 Nanyang Drive, Singapore 637553, Singapore
- School of Chemical and Biomedical Engineering, Nanyang Technological University , 62 Nanyang Drive, Singapore 637459, Singapore
| |
Collapse
|
14
|
Yuan X, Gavriilaki E, Thanassi JA, Yang G, Baines AC, Podos SD, Huang Y, Huang M, Brodsky RA. Small-molecule factor D inhibitors selectively block the alternative pathway of complement in paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome. Haematologica 2016; 102:466-475. [PMID: 27810992 PMCID: PMC5394948 DOI: 10.3324/haematol.2016.153312] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/24/2016] [Indexed: 12/13/2022] Open
Abstract
Paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome are diseases of excess activation of the alternative pathway of complement that are treated with eculizumab, a humanized monoclonal antibody against the terminal complement component C5. Eculizumab must be administered intravenously, and moreover some patients with paroxysmal nocturnal hemoglobinuria on eculizumab have symptomatic extravascular hemolysis, indicating an unmet need for additional therapeutic approaches. We report the activity of two novel small-molecule inhibitors of the alternative pathway component Factor D using in vitro correlates of both paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome. Both compounds bind human Factor D with high affinity and effectively inhibit its proteolytic activity against purified Factor B in complex with C3b. When tested using the traditional Ham test with cells from paroxysmal nocturnal hemoglobinuria patients, the Factor D inhibitors significantly reduced complement-mediated hemolysis at concentrations as low as 0.01 μM. Additionally the compound ACH-4471 significantly decreased C3 fragment deposition on paroxysmal nocturnal hemoglobinuria erythrocytes, indicating a reduced potential relative to eculizumab for extravascular hemolysis. Using the recently described modified Ham test with serum from patients with atypical hemolytic uremic syndrome, the compounds reduced the alternative pathway-mediated killing of PIGA-null reagent cells, thus establishing their potential utility for this disease of alternative pathway of complement dysregulation and validating the modified Ham test as a system for pre-clinical drug development for atypical hemolytic uremic syndrome. Finally, ACH-4471 blocked alternative pathway activity when administered orally to cynomolgus monkeys. In conclusion, the small-molecule Factor D inhibitors show potential as oral therapeutics for human diseases driven by the alternative pathway of complement, including paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome.
Collapse
Affiliation(s)
- Xuan Yuan
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eleni Gavriilaki
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Andrea C Baines
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | - Robert A Brodsky
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Ricklin D, Reis ES, Mastellos DC, Gros P, Lambris JD. Complement component C3 - The "Swiss Army Knife" of innate immunity and host defense. Immunol Rev 2016; 274:33-58. [PMID: 27782325 PMCID: PMC5427221 DOI: 10.1111/imr.12500] [Citation(s) in RCA: 273] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As a preformed defense system, complement faces a delicate challenge in providing an immediate, forceful response to pathogens even at first encounter, while sparing host cells in the process. For this purpose, it engages a tightly regulated network of plasma proteins, cell surface receptors, and regulators. Complement component C3 plays a particularly versatile role in this process by keeping the cascade alert, acting as a point of convergence of activation pathways, fueling the amplification of the complement response, exerting direct effector functions, and helping to coordinate downstream immune responses. In recent years, it has become evident that nature engages the power of C3 not only to clear pathogens but also for a variety of homeostatic processes ranging from tissue regeneration and synapse pruning to clearing debris and controlling tumor cell progression. At the same time, its central position in immune surveillance makes C3 a target for microbial immune evasion and, if improperly engaged, a trigger point for various clinical conditions. In our review, we look at the versatile roles and evolutionary journey of C3, discuss new insights into the molecular basis for C3 function, provide examples of disease involvement, and summarize the emerging potential of C3 as a therapeutic target.
Collapse
Affiliation(s)
- Daniel Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Edimara S Reis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dimitrios C Mastellos
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- National Center for Scientific Research 'Demokritos', Athens, Greece
| | - Piet Gros
- Utrecht University, Utrecht, The Netherlands
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Palamini M, Canciani A, Forneris F. Identifying and Visualizing Macromolecular Flexibility in Structural Biology. Front Mol Biosci 2016; 3:47. [PMID: 27668215 PMCID: PMC5016524 DOI: 10.3389/fmolb.2016.00047] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/22/2016] [Indexed: 12/29/2022] Open
Abstract
Structural biology comprises a variety of tools to obtain atomic resolution data for the investigation of macromolecules. Conventional structural methodologies including crystallography, NMR and electron microscopy often do not provide sufficient details concerning flexibility and dynamics, even though these aspects are critical for the physiological functions of the systems under investigation. However, the increasing complexity of the molecules studied by structural biology (including large macromolecular assemblies, integral membrane proteins, intrinsically disordered systems, and folding intermediates) continuously demands in-depth analyses of the roles of flexibility and conformational specificity involved in interactions with ligands and inhibitors. The intrinsic difficulties in capturing often subtle but critical molecular motions in biological systems have restrained the investigation of flexible molecules into a small niche of structural biology. Introduction of massive technological developments over the recent years, which include time-resolved studies, solution X-ray scattering, and new detectors for cryo-electron microscopy, have pushed the limits of structural investigation of flexible systems far beyond traditional approaches of NMR analysis. By integrating these modern methods with powerful biophysical and computational approaches such as generation of ensembles of molecular models and selective particle picking in electron microscopy, more feasible investigations of dynamic systems are now possible. Using some prominent examples from recent literature, we review how current structural biology methods can contribute useful data to accurately visualize flexibility in macromolecular structures and understand its important roles in regulation of biological processes.
Collapse
Affiliation(s)
| | | | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Department of Biology and Biotechnology, University of PaviaPavia, Italy
| |
Collapse
|
17
|
Protein conformational plasticity and complex ligand-binding kinetics explored by atomistic simulations and Markov models. Nat Commun 2015; 6:7653. [PMID: 26134632 PMCID: PMC4506540 DOI: 10.1038/ncomms8653] [Citation(s) in RCA: 306] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 05/28/2015] [Indexed: 12/20/2022] Open
Abstract
Understanding the structural mechanisms of protein–ligand binding and their dependence on protein sequence and conformation is of fundamental importance for biomedical research. Here we investigate the interplay of conformational change and ligand-binding kinetics for the serine protease Trypsin and its competitive inhibitor Benzamidine with an extensive set of 150 μs molecular dynamics simulation data, analysed using a Markov state model. Seven metastable conformations with different binding pocket structures are found that interconvert at timescales of tens of microseconds. These conformations differ in their substrate-binding affinities and binding/dissociation rates. For each metastable state, corresponding solved structures of Trypsin mutants or similar serine proteases are contained in the protein data bank. Thus, our wild-type simulations explore a space of conformations that can be individually stabilized by adding ligands or making suitable changes in protein sequence. These findings provide direct evidence of conformational plasticity in receptors. Conformational plasticity influences several aspects of protein function. Here the authors combine extensive MD simulations with Markov state models—using trypsin as model—to reveal new mechanistic details of how conformational plasticity influence ligand-receptors interactions.
Collapse
|
18
|
Ogawa H, Damrongrungruang T, Hori S, Nouno K, Minagawa K, Sato M, Miyazaki H. Effect of periodontal treatment on adipokines in type 2 diabetes. World J Diabetes 2014; 5:924-931. [PMID: 25512798 PMCID: PMC4265882 DOI: 10.4239/wjd.v5.i6.924] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/29/2014] [Accepted: 11/10/2014] [Indexed: 02/05/2023] Open
Abstract
The association between adipokines and inflammatory periodontal diseases has been studied over the last two decades. This review was intended to explore the observation that periodontal therapy may lead to an improvement of adipokines in diabetic patients. In summary, substantial evidence suggests that diabetes is associated with increased prevalence, extent and severity of periodontitis. Numerous mechanisms have been elucidated to explain the impact of diabetes on the periodontium. However, current knowledge concerning the role of major adipokines indicates only some of their associations with the pathogenesis of periodontitis in type 2 diabetes. Conversely, treatment of periodontal disease and reduction of oral inflammation may have positive effects on the diabetic condition, although evidence for this remains somewhat equivocal.
Collapse
|
19
|
Sun Y, Yin S, Feng Y, Li J, Zhou J, Liu C, Zhu G, Guo Z. Molecular basis of the general base catalysis of an α/β-hydrolase catalytic triad. J Biol Chem 2014; 289:15867-79. [PMID: 24737327 DOI: 10.1074/jbc.m113.535641] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The serine-histidine-aspartate triad is well known for its covalent, nucleophilic catalysis in a diverse array of enzymatic transformations. Here we show that its nucleophilicity is shielded and its catalytic role is limited to being a specific general base by an open-closed conformational change in the catalysis of (1R,6R)-2-succinyl-6-hydroxy-2,4-cyclohexadiene-1-carboxylate synthase (or MenH), a typical α/β-hydrolase fold enzyme in the vitamin K biosynthetic pathway. This enzyme is found to adopt an open conformation without a functional triad in its ligand-free form and a closed conformation with a fully functional catalytic triad in the presence of its reaction product. The open-to-closed conformational transition involves movement of half of the α-helical cap domain, which causes extensive structural changes in the α/β-domain and forces the side chain of the triad histidine to adopt an energetically disfavored gauche conformation to form the functional triad. NMR analysis shows that the inactive open conformation without a triad prevails in ligand-free solution and is converted to the closed conformation with a properly formed triad by the reaction product. Mutation of the residues crucial to this open-closed transition either greatly decreases or completely eliminates the enzyme activity, supporting an important catalytic role for the structural change. These findings suggest that the open-closed conformational change tightly couples formation of the catalytic triad to substrate binding to enhance the substrate specificities and simultaneously shield the nucleophilicity of the triad, thus allowing it to expand its catalytic power beyond the nucleophilic catalysis.
Collapse
Affiliation(s)
- Yueru Sun
- From the Department of Chemistry, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region (SAR), China and
| | - Shuhui Yin
- From the Department of Chemistry, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region (SAR), China and
| | - Yitao Feng
- From the Department of Chemistry, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region (SAR), China and
| | - Jie Li
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Jiahai Zhou
- State Key Laboratory of Bio-organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Changdong Liu
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region (SAR), China and Division of Life Sciences, and
| | - Guang Zhu
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region (SAR), China and Division of Life Sciences, and
| | - Zhihong Guo
- From the Department of Chemistry, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region (SAR), China and
| |
Collapse
|
20
|
Forneris F, Burnley BT, Gros P. Ensemble refinement shows conformational flexibility in crystal structures of human complement factor D. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2014; 70:733-43. [PMID: 24598742 PMCID: PMC3949522 DOI: 10.1107/s1399004713032549] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 11/29/2013] [Indexed: 11/16/2022]
Abstract
Human factor D (FD) is a self-inhibited thrombin-like serine proteinase that is critical for amplification of the complement immune response. FD is activated by its substrate through interactions outside the active site. The substrate-binding, or `exosite', region displays a well defined and rigid conformation in FD. In contrast, remarkable flexibility is observed in thrombin and related proteinases, in which Na(+) and ligand binding is implied in allosteric regulation of enzymatic activity through protein dynamics. Here, ensemble refinement (ER) of FD and thrombin crystal structures is used to evaluate structure and dynamics simultaneously. A comparison with previously published NMR data for thrombin supports the ER analysis. The R202A FD variant has enhanced activity towards artificial peptides and simultaneously displays active and inactive conformations of the active site. ER revealed pronounced disorder in the exosite loops for this FD variant, reminiscent of thrombin in the absence of the stabilizing Na(+) ion. These data indicate that FD exhibits conformational dynamics like thrombin, but unlike in thrombin a mechanism has evolved in FD that locks the unbound native state into an ordered inactive conformation via the self-inhibitory loop. Thus, ensemble refinement of X-ray crystal structures may represent an approach alternative to spectroscopy to explore protein dynamics in atomic detail.
Collapse
Affiliation(s)
- Federico Forneris
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - B. Tom Burnley
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Piet Gros
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
21
|
Kromann-Hansen T, Lund IK, Liu Z, Andreasen PA, Høyer-Hansen G, Sørensen HP. Allosteric inactivation of a trypsin-like serine protease by an antibody binding to the 37- and 70-loops. Biochemistry 2013; 52:7114-26. [PMID: 24079451 DOI: 10.1021/bi400491k] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Serine protease catalytic activity is in many cases regulated by conformational changes initiated by binding of physiological modulators to exosites located distantly from the active site. Inhibitory monoclonal antibodies binding to such exosites are potential therapeutics and offer opportunities for elucidating fundamental allosteric mechanisms. The monoclonal antibody mU1 has previously been shown to be able to inhibit the function of murine urokinase-type plasminogen activator in vivo. We have now mapped the epitope of mU1 to the catalytic domain's 37- and 70-loops, situated about 20 Å from the S1 specificity pocket of the active site. Our data suggest that binding of mU1 destabilizes the catalytic domain and results in conformational transition into a state, in which the N-terminal amino group of Ile16 is less efficiently stabilizing the oxyanion hole and in which the active site has a reduced affinity for substrates and inhibitors. Furthermore, we found evidence for functional interactions between residues in uPA's C-terminal catalytic domain and its N-terminal A-chain, as deletion of the A-chain facilitates the mU1-induced conformational distortion. The inactive, distorted state is by several criteria similar to the E* conformation described for other serine proteases. Hence, agents targeting serine protease conformation through binding to exosites in the 37- and 70-loops represent a new class of potential therapeutics.
Collapse
Affiliation(s)
- Tobias Kromann-Hansen
- Danish-Chinese Centre for Proteases and Cancer and ‡Department of Molecular Biology and Genetics, Aarhus University , DK-8000 Aarhus C, Denmark
| | | | | | | | | | | |
Collapse
|
22
|
Lea SM, Johnson S. Putting the structure into complement. Immunobiology 2013; 217:1117-21. [PMID: 22964238 DOI: 10.1016/j.imbio.2012.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 07/13/2012] [Accepted: 07/15/2012] [Indexed: 11/25/2022]
Abstract
In a field where structure has finally begun to have a real impact, a series of new structures over the last two years have further extended our understanding of some of the critical regulatory events of the complement system. Notably, information has begun to flow from larger assemblies of components which allow insight into the often transient assemblies critical to complement regulation at the cell surface. This review will summarise the key structures determined since the last International Complement Workshop and the insights these have given us, before highlighting some questions that still require molecular frameworks to drive understanding.
Collapse
Affiliation(s)
- Susan M Lea
- Sir William Dunn School of Pathology, South Parks Road, Oxford OX1 3RE, UK.
| | | |
Collapse
|
23
|
Kroh HK, Bock PE. Effect of zymogen domains and active site occupation on activation of prothrombin by von Willebrand factor-binding protein. J Biol Chem 2012; 287:39149-57. [PMID: 23012355 DOI: 10.1074/jbc.m112.415562] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Prothrombin is conformationally activated by von Willebrand factor-binding protein (vWbp) from Staphylococcus aureus through insertion of the NH(2)-terminal residues of vWbp into the prothrombin catalytic domain. The rate of prothrombin activation by vWbp(1-263) is controlled by a hysteretic kinetic mechanism initiated by substrate binding. The present study evaluates activation of prothrombin by full-length vWbp(1-474) through activity progress curve analysis. Additional interactions from the COOH-terminal half of vWbp(1-474) strengthened the initial binding of vWbp to prothrombin, resulting in higher activity and an ∼100-fold enhancement in affinity. The affinities of vWbp(1-263) or vWbp(1-474) were compared by equilibrium binding to the prothrombin derivatives prethrombin 1, prethrombin 2, thrombin, meizothrombin, and meizothrombin(des-fragment 1) and their corresponding active site-blocked analogs. Loss of fragment 1 in prethrombin 1 enhanced affinity for both vWbp(1-263) and vWbp(1-474), with a 30-45% increase in Gibbs free energy, implicating a regulatory role for fragment 1 in the activation mechanism. Active site labeling of all prothrombin derivatives with D-Phe-Pro-Arg-chloromethyl ketone, analogous to irreversible binding of a substrate, decreased their K(D) values for vWbp into the subnanomolar range, reflecting the dependence of the activating conformational change on substrate binding. The results suggest a role for prothrombin domains in the pathophysiological activation of prothrombin by vWbp, and may reveal a function for autocatalysis of the vWbp·prothrombin complexes during initiation of blood coagulation.
Collapse
Affiliation(s)
- Heather K Kroh
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
24
|
Liu Z, Kromann-Hansen T, Lund IK, Hosseini M, Jensen KJ, Høyer-Hansen G, Andreasen PA, Sørensen HP. Interconversion of active and inactive conformations of urokinase-type plasminogen activator. Biochemistry 2012; 51:7804-11. [PMID: 22950516 DOI: 10.1021/bi3005957] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The catalytic activity of serine proteases depends on a salt-bridge between the amino group of residue 16 and the side chain of Asp194. The salt-bridge stabilizes the oxyanion hole and the S1 specificity pocket of the protease. Some serine proteases exist in only partially active forms, in which the amino group of residue 16 is exposed to the solvent. Such a partially active state is assumed by a truncated form of the murine urokinase-type plasminogen activator (muPA), consisting of residues 16-243. Here we investigated the allosteric interconversion between partially active states and the fully active state. Both a monoclonal antibody (mU3) and a peptidic inhibitor (mupain-1--16) stabilize the active state. The epitope of mU3 is located in the 37- and 70-loops at a site homologous to exosite I of thrombin. The N-terminus((Ile16)) of muPA((16--243)) was less exposed upon binding of mU3 or mupain-1--16. In contrast, introduction of the mutations F40Y or E137A into muPA((16--243)) increased exposure of the N-terminus((Ile16)) and resulted in large changes in the thermodynamic parameters for mupain-1--16 binding. We conclude that the distorted state of muPA((16--243)) is conformationally ordered upon binding of ligands to the active site and upon binding of mU3 to the 37- and 70-loops. Our study establishes the 37- and 70-loops as a unique site for binding to compounds stabilizing the active state of serine proteases.
Collapse
Affiliation(s)
- Zhuo Liu
- Danish-Chinese Centre for Proteases and Cancer, Aarhus University, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Pozzi N, Vogt AD, Gohara DW, Di Cera E. Conformational selection in trypsin-like proteases. Curr Opin Struct Biol 2012; 22:421-31. [PMID: 22664096 DOI: 10.1016/j.sbi.2012.05.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 05/08/2012] [Accepted: 05/10/2012] [Indexed: 01/30/2023]
Abstract
For over four decades, two competing mechanisms of ligand recognition--conformational selection and induced-fit--have dominated our interpretation of protein allostery. Defining the mechanism broadens our understanding of the system and impacts our ability to design effective drugs and new therapeutics. Recent kinetics studies demonstrate that trypsin-like proteases exist in equilibrium between two forms: one fully accessible to substrate (E) and the other with the active site occluded (E*). Analysis of the structural database confirms existence of the E* and E forms and vouches for the allosteric nature of the trypsin fold. Allostery in terms of conformational selection establishes an important paradigm in the protease field and enables protein engineers to expand the repertoire of proteases as therapeutics.
Collapse
Affiliation(s)
- Nicola Pozzi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, United States
| | | | | | | |
Collapse
|
26
|
Forneris F, Wu J, Gros P. The modular serine proteases of the complement cascade. Curr Opin Struct Biol 2012; 22:333-41. [PMID: 22560446 DOI: 10.1016/j.sbi.2012.04.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 04/05/2012] [Indexed: 11/30/2022]
Abstract
Modular serine proteases are central to the complement cascade of the mammalian humoral immune system. These proteases form protein complexes through multi-domain interactions to achieve their proteolytic activity. We review the structural insights into complement initiation by auto-activation of the hetero-tetrameric proteases of the large danger-recognition protein complexes, amplification and labelling of particles by the formation and activity of C3 convertases, and regulation by convertase dissociation and degradation to prevent 'bystander' damage to healthy host cells and tissues. The data reveal that complex formation and large domain-domain rearrangements underlie the proteolytic reactions of the complement cascade, which enables the host to recognize and clear invading microbes and host debris from its blood and fluids surrounding tissues.
Collapse
Affiliation(s)
- Federico Forneris
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | | | | |
Collapse
|
27
|
Abstract
Protein C is activated by thrombin with a value of k(cat)/K(m) = 0.11mM(-1)s(-1) that increases 1700-fold in the presence of the cofactor thrombomodulin. The molecular origin of this effect triggering an important feedback loop in the coagulation cascade remains elusive. Acidic residues in the activation domain of protein C are thought to electrostatically clash with the active site of thrombin. However, functional and structural data reported here support an alternative scenario. The thrombin precursor prethrombin-2 has R15 at the site of activation in ionic interaction with E14e, D14l, and E18, instead of being exposed to solvent for proteolytic attack. Residues E160, D167, and D172 around the site of activation at R169 of protein C occupy the same positions as E14e, D14l, and E18 in prethrombin-2. Caging of R169 by E160, D167, and D172 is responsible for much of the poor activity of thrombin toward protein C. The E160A/D167A/D172A mutant is activated by thrombin 63-fold faster than wild-type in the absence of thrombomodulin and, over a slower time scale, spontaneously converts to activated protein C. These findings establish a new paradigm for cofactor-assisted reactions in the coagulation cascade.
Collapse
|
28
|
Katschke KJ, Wu P, Ganesan R, Kelley RF, Mathieu MA, Hass PE, Murray J, Kirchhofer D, Wiesmann C, van Lookeren Campagne M. Inhibiting alternative pathway complement activation by targeting the factor D exosite. J Biol Chem 2012; 287:12886-92. [PMID: 22362762 DOI: 10.1074/jbc.m112.345082] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
By virtue of its amplifying property, the alternative complement pathway has been implicated in a number of inflammatory diseases and constitutes an attractive therapeutic target. An anti-factor D Fab fragment (AFD) was generated to inhibit the alternative complement pathway in advanced dry age-related macular degeneration. AFD potently prevented factor D (FD)-mediated proteolytic activation of its macromolecular substrate C3bB, but not proteolysis of a small synthetic substrate, indicating that AFD did not block access of the substrate to the catalytic site. The crystal structures of AFD in complex with human and cynomolgus FD (at 2.4 and 2.3 Å, respectively) revealed the molecular details of the inhibitory mechanism. The structures show that the AFD-binding site includes surface loops of FD that form part of the FD exosite. Thus, AFD inhibits FD proteolytic function by interfering with macromolecular substrate access rather than by inhibiting FD catalysis, providing the molecular basis of AFD-mediated inhibition of a rate-limiting step in the alternative complement pathway.
Collapse
Affiliation(s)
- Kenneth J Katschke
- Department of Immunology, Genentech Incorporated, South San Francisco, California 94080, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pozzi N, Chen Z, Zapata F, Pelc LA, Barranco-Medina S, Di Cera E. Crystal structures of prethrombin-2 reveal alternative conformations under identical solution conditions and the mechanism of zymogen activation. Biochemistry 2011; 50:10195-202. [PMID: 22049947 DOI: 10.1021/bi2015019] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Prethrombin-2 is the immediate zymogen precursor of the clotting enzyme thrombin, which is generated upon cleavage at R15 and separation of the A chain and catalytic B chain. The X-ray structure of prethrombin-2 determined in the free form at 1.9 Å resolution shows the 215-217 segment collapsed into the active site and occluding 49% of the volume available for substrate binding. Remarkably, some of the crystals harvested from the same crystallization well, under identical solution conditions, diffract to 2.2 Å resolution in the same space group but produce a structure in which the 215-217 segment moves >5 Å and occludes 24% of the volume available for substrate binding. The two alternative conformations of prethrombin-2 have the side chain of W215 relocating >9 Å within the active site and are relevant to the allosteric E*-E equilibrium of the mature enzyme. Another unanticipated feature of prethrombin-2 bears on the mechanism of prothrombin activation. R15 is found buried within the protein in ionic interactions with E14e, D14l, and E18, thereby making its exposure to solvent necessary for proteolytic attack and conversion to thrombin. On the basis of this structural observation, we constructed the E14eA/D14lA/E18A triple mutant to reduce the level of electrostatic coupling with R15 and promote zymogen activation. The mutation causes prethrombin-2 to spontaneously convert to thrombin, without the need for the snake venom ecarin or the physiological prothrombinase complex.
Collapse
Affiliation(s)
- Nicola Pozzi
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | | | | | | | | | | |
Collapse
|
30
|
Allostery in trypsin-like proteases suggests new therapeutic strategies. Trends Biotechnol 2011; 29:577-85. [PMID: 21726912 DOI: 10.1016/j.tibtech.2011.06.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 05/19/2011] [Accepted: 06/02/2011] [Indexed: 11/21/2022]
Abstract
Trypsin-like proteases (TLPs) are a large family of enzymes responsible for digestion, blood coagulation, fibrinolysis, development, fertilization, apoptosis and immunity. A current paradigm posits that the irreversible transition from an inactive zymogen to the active protease form enables productive interaction with substrate and catalysis. Analysis of the entire structural database reveals two distinct conformations of the active site: one fully accessible to substrate (E) and the other occluded by the collapse of a specific segment (E*). The allosteric E*-E equilibrium provides a reversible mechanism for activity and regulation in addition to the irreversible zymogen to protease conversion and points to new therapeutic strategies aimed at inhibiting or activating the enzyme. In this review, we discuss relevant examples, with emphasis on the rational engineering of anticoagulant thrombin mutants.
Collapse
|
31
|
Niu W, Chen Z, Gandhi PS, Vogt AD, Pozzi N, Pelc LA, Zapata F, Di Cera E. Crystallographic and kinetic evidence of allostery in a trypsin-like protease. Biochemistry 2011; 50:6301-7. [PMID: 21707111 DOI: 10.1021/bi200878c] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Protein allostery is based on the existence of multiple conformations in equilibrium linked to distinct functional properties. Although evidence of allosteric transitions is relatively easy to identify by functional studies, structural detection of a pre-existing equilibrium between alternative conformations remains challenging even for textbook examples of allosteric proteins. Kinetic studies show that the trypsin-like protease thrombin exists in equilibrium between two conformations where the active site is either collapsed (E*) or accessible to substrate (E). However, structural demonstration that the two conformations exist in the same enzyme construct free of ligands has remained elusive. Here we report the crystal structure of the thrombin mutant N143P in the E form, which complements the recently reported structure in the E* form, and both the E and E* forms of the thrombin mutant Y225P. The side chain of W215 moves 10.9 Å between the two forms, causing a displacement of 6.6 Å of the entire 215-217 segment into the active site that in turn opens or closes access to the primary specificity pocket. Rapid kinetic measurements of p-aminobenzamidine binding to the active site confirm the existence of the E*-E equilibrium in solution for wild-type and the mutants N143P and Y225P. These findings provide unequivocal proof of the allosteric nature of thrombin and lend strong support to the recent proposal that the E*-E equilibrium is a key property of the trypsin fold.
Collapse
Affiliation(s)
- Weiling Niu
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Forneris F, Ricklin D, Wu J, Tzekou A, Wallace RS, Lambris JD, Gros P. Structures of C3b in complex with factors B and D give insight into complement convertase formation. Science 2011; 330:1816-20. [PMID: 21205667 DOI: 10.1126/science.1195821] [Citation(s) in RCA: 208] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Activation of the complement cascade induces inflammatory responses and marks cells for immune clearance. In the central complement-amplification step, a complex consisting of surface-bound C3b and factor B is cleaved by factor D to generate active convertases on targeted surfaces. We present crystal structures of the pro-convertase C3bB at 4 angstrom resolution and its complex with factor D at 3.5 angstrom resolution. Our data show how factor B binding to C3b forms an open "activation" state of C3bB. Factor D specifically binds the open conformation of factor B through a site distant from the catalytic center and is activated by the substrate, which displaces factor D's self-inhibitory loop. This concerted proteolytic mechanism, which is cofactor-dependent and substrate-induced, restricts complement amplification to C3b-tagged target cells.
Collapse
Affiliation(s)
- Federico Forneris
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| | | | | | | | | | | | | |
Collapse
|
33
|
Sirmaci A, Walsh T, Akay H, Spiliopoulos M, Şakalar YB, Hasanefendioğlu-Bayrak A, Duman D, Farooq A, King MC, Tekin M. MASP1 mutations in patients with facial, umbilical, coccygeal, and auditory findings of Carnevale, Malpuech, OSA, and Michels syndromes. Am J Hum Genet 2010; 87:679-86. [PMID: 21035106 DOI: 10.1016/j.ajhg.2010.09.018] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 09/23/2010] [Accepted: 09/29/2010] [Indexed: 10/18/2022] Open
Abstract
Distinctive facial features consisting of hypertelorism, telecanthus, blepharophimosis, blepharoptosis, epicanthus inversus, periumbilical defects, and skeletal anomalies are seen in autosomal-recessive Carnevale, Malpuech, Michels, and oculo-skeletal-abdominal (OSA) syndromes. The gene or genes responsible for these syndromes were heretofore unknown. We report on three individuals from two consanguineous Turkish families with findings characteristic of these syndromes, including facial dysmorphism, periumbilical depression, mixed hearing loss, radioulnar synostosis, and coccygeal appendage. Homozygosity mapping yielded an autozygous region on chromosome 3q27 in both families. In one family, whole exome sequencing revealed a missense mutation, MASP1 c.2059G>A (p.G687R), that cosegregated with the phenotype. In the second family, Sanger sequencing of MASP1 revealed a nonsense mutation, MASP1 c.870G>A (p.W290X), that also cosegregated with the phenotype. Neither mutation was found in 192 Turkish controls or 1200 controls of various other ancestries. MASP1 encodes mannan-binding lectin serine protease 1. The two mutations occur in a MASP1 isoform that has been reported to process IGFBP-5, thereby playing a critical role in insulin growth factor availability during craniofacial and muscle development. These results implicate mutations of MASP1 as the cause of a human malformation syndrome and demonstrate the involvement of MASP1 in facial, umbilical, and ear development during the embryonic period.
Collapse
|
34
|
Molecular mechanisms of complement evasion: learning from staphylococci and meningococci. Nat Rev Microbiol 2010; 8:393-9. [PMID: 20467445 DOI: 10.1038/nrmicro2366] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The complement system is a crucial component of the innate immune response in humans. Recent studies in Staphylococcus aureus and Neisseria meningitidis have revealed how these bacteria escape complement-mediated killing. In addition, new structural data have provided detailed insights into the molecular mechanisms of host defence mediated by the complement system and how bacterial proteins interfere with this process. This information is fundamental to our understanding of bacterial pathogenesis and may facilitate the design of better vaccines.
Collapse
|
35
|
Dobó J, Harmat V, Beinrohr L, Sebestyén E, Závodszky P, Gál P. MASP-1, a promiscuous complement protease: structure of its catalytic region reveals the basis of its broad specificity. THE JOURNAL OF IMMUNOLOGY 2009; 183:1207-14. [PMID: 19564340 DOI: 10.4049/jimmunol.0901141] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mannose-binding lectin (MBL)-associated serine protease (MASP)-1 is an abundant component of the lectin pathway of complement. The related enzyme, MASP-2 is capable of activating the complement cascade alone. Though the concentration of MASP-1 far exceeds that of MASP-2, only a supporting role of MASP-1 has been identified regarding lectin pathway activation. Several non-complement substrates, like fibrinogen and factor XIII, have also been reported. MASP-1 belongs to the C1r/C1s/MASP family of modular serine proteases; however, its serine protease domain is evolutionary different. We have determined the crystal structure of the catalytic region of active MASP-1 and refined it to 2.55 A resolution. Unusual features of the structure are an internal salt bridge (similar to one in factor D) between the S1 Asp189 and Arg224, and a very long 60-loop. The functional and evolutionary differences between MASP-1 and the other members of the C1r/C1s/MASP family are reflected in the crystal structure. Structural comparison of the protease domains revealed that the substrate binding groove of MASP-1 is wide and resembles that of trypsin rather than early complement proteases explaining its relaxed specificity. Also, MASP-1's multifunctional behavior as both a complement and a coagulation enzyme is in accordance with our observation that antithrombin in the presence of heparin is a more potent inhibitor of MASP-1 than C1 inhibitor. Overall, MASP-1 behaves as a promiscuous protease. The structure shows that its substrate binding groove is accessible; however, its reactivity could be modulated by an unusually large 60-loop and an internal salt bridge involving the S1 Asp.
Collapse
Affiliation(s)
- József Dobó
- Institute of Enzymology, Biological Research Center, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | | | | | |
Collapse
|
36
|
Gál P, Dobó J, Závodszky P, Sim RBM. Early complement proteases: C1r, C1s and MASPs. A structural insight into activation and functions. Mol Immunol 2009; 46:2745-52. [PMID: 19477526 DOI: 10.1016/j.molimm.2009.04.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Accepted: 04/28/2009] [Indexed: 10/20/2022]
Abstract
C1r, C1s and the mannose-binding lectin-associated serine proteases (MASPs) are responsible for the initiation of the classical- and lectin pathway activation of the complement system. These enzymes do not act alone, but form supramolecular complexes with pattern recognition molecules such as C1q, MBL, and ficolins. They share the same domain organization but have different substrate specificities and fulfill different physiological functions. In the recent years the rapid progress of structural biology facilitated the understanding of the molecular mechanism of complement activation at atomic level. In this review we summarize our current knowledge about the structure and function of the early complement proteases, delineate the latest models of the multimolecular complexes and present the functional consequences inferred from the structural studies. We also discuss some open questions and debated issues that need to be resolved in the future.
Collapse
Affiliation(s)
- Péter Gál
- Institute of Enzymology, Biological Research Center, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | |
Collapse
|
37
|
Spraggon G, Hornsby M, Shipway A, Tully DC, Bursulaya B, Danahay H, Harris JL, Lesley SA. Active site conformational changes of prostasin provide a new mechanism of protease regulation by divalent cations. Protein Sci 2009; 18:1081-1094. [PMID: 19388054 PMCID: PMC2771310 DOI: 10.1002/pro.118] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 02/26/2009] [Accepted: 02/27/2009] [Indexed: 01/28/2025]
Abstract
Prostasin or human channel-activating protease 1 has been reported to play a critical role in the regulation of extracellular sodium ion transport via its activation of the epithelial cell sodium channel. Here, the structure of the extracellular portion of the membrane associated serine protease has been solved to high resolution in complex with a nonselective d-FFR chloromethyl ketone inhibitor, in an apo form, in a form where the apo crystal has been soaked with the covalent inhibitor camostat and in complex with the protein inhibitor aprotinin. It was also crystallized in the presence of the divalent cation Ca(+2). Comparison of the structures with each other and with other members of the trypsin-like serine protease family reveals unique structural features of prostasin and a large degree of conformational variation within specificity determining loops. Of particular interest is the S1 subsite loop which opens and closes in response to basic residues or divalent ions, directly binding Ca(+2) cations. This induced fit active site provides a new possible mode of regulation of trypsin-like proteases adapted in particular to extracellular regions with variable ionic concentrations such as the outer membrane layer of the epithelial cell.
Collapse
Affiliation(s)
- Glen Spraggon
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Sun PD, Boyington JC. Overview of protein folds in the immune system. ACTA ACUST UNITED AC 2008; Appendix 1:Appendix 1N. [PMID: 18432648 DOI: 10.1002/0471142735.ima01ns44] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The rapid advancement of X-ray crystallography and nuclear magnetic resonance techniques in recent years has resulted in the solution of macromolecular structures at an unprecedented rate. This review aims at providing a comprehensive description of structures and folds related to the function of the immune system. Focus is placed on immunologically relevant proteins such as immunoreceptors and major histocompatibility complexes. Information is also provided regarding protein structure data banks.
Collapse
Affiliation(s)
- P D Sun
- National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| | | |
Collapse
|
39
|
Abstract
This overview provides an illustrated, comprehensive survey of some commonly observed protein‐fold families and structural motifs, chosen for their functional significance. It opens with descriptions and definitions of the various elements of protein structure and associated terminology. Following is an introduction into web‐based structural bioinformatics that includes surveys of interactive web servers for protein fold or domain annotation, protein‐structure databases, protein‐structure‐classification databases, structural alignments of proteins, and molecular graphics programs available for personal computers. The rest of the overview describes selected families of protein folds in terms of their secondary, tertiary, and quaternary structural arrangements, including ribbon‐diagram examples, tables of representative structures with references, and brief explanations pointing out their respective biological and functional significance.
Collapse
Affiliation(s)
- Peter D Sun
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | | | | |
Collapse
|
40
|
Arlaud GJ, Barlow PN, Gaboriaud C, Gros P, Narayana SVL. Deciphering complement mechanisms: the contributions of structural biology. Mol Immunol 2007; 44:3809-22. [PMID: 17768099 DOI: 10.1016/j.molimm.2007.06.147] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2007] [Indexed: 12/25/2022]
Abstract
Since the resolution of the first three-dimensional structure of a complement component in 1980, considerable efforts have been put into the investigation of this system through structural biology techniques, resulting in about a hundred structures deposited in the Protein Data Bank by the beginning of 2007. By revealing its mechanisms at the atomic level, these approaches significantly improve our understanding of complement, opening the way to the rational design of specific inhibitors. This review is co-authored by some of the researchers currently involved in the structural biology of complement and its purpose is to illustrate, through representative examples, how X-ray crystallography and NMR techniques help us decipher the many sophisticated mechanisms that underlie complement functions.
Collapse
Affiliation(s)
- Gérard J Arlaud
- Institut de Biologie Structurale Jean-Pierre Ebel, CEA, CNRS, Université Joseph Fourier, 41 rue Jules Horowitz, F-38027 Grenoble, France.
| | | | | | | | | |
Collapse
|
41
|
Abstract
The complement system is a central component of innate immunity and bridges the innate to the adaptive immune response. However, it can also turn its destructive capabilities against host cells and is involved in numerous diseases and pathological conditions. Modulation of the complement system has been recognized as a promising strategy in drug discovery, and a large number of therapeutic modalities have been developed. However, successful marketing of complement-targeted drugs has proved to be more difficult than initially expected, and many strategies have been discontinued. The US Food and Drug Administration's approval of the first complement-specific drug, an antibody against complement component C5 (eculizumab; Soliris), in March 2007, was a long-awaited breakthrough in the field. Approval of eculizumab validates the complement system as therapeutic target and might facilitate clinical development of other promising drug candidates.
Collapse
Affiliation(s)
- Daniel Ricklin
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
42
|
Kozlov LV, Burdelev OO, Bureeva SV, Kaplun AP. Artificial inhibition of the complement system. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2007; 33:485-510. [DOI: 10.1134/s1068162007050020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
43
|
Segers K, Rosing J, Nicolaes GAF. Structural models of the snake venom factor V activators from Daboia russelli and Daboia lebetina. Proteins 2006; 64:968-84. [PMID: 16807918 DOI: 10.1002/prot.21051] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Blood coagulation factor V (FV) is a multifunctional protein that circulates in human plasma as a precursor molecule which can be activated by thrombin or activated factor X (FXa) in order to express its cofactor activity in prothrombin activation. FV activation is achieved by limited proteolysis after Arg709, Arg1018, and Arg1545 in the FV molecule. The venoms of Daboia russelli and Daboia lebetina contain a serine protease that specifically activates FV by a single cleavage at Arg1545. We have predicted the three-dimensional structure of these enzymes using comparative protein modeling techniques. The plasminogen activator from Agkistrodon acutus, which shows a high degree of homology with the venom FV activators and for which a high-quality crystallographic structure is available, was used as the molecular template. The RVV-V and LVV-V models provide for the first time a detailed and accurate structure of a snake venom FV activator and explain the observed sensitivity or resistance toward a number of serine protease inhibitors. Finally, electrostatic potential calculations show that two positively charged surface patches are present on opposite sides of the active site. We propose that both FV activators achieve their exquisite substrate specificity for the Arg1545 site via interactions between these exosites and FV.
Collapse
Affiliation(s)
- Kenneth Segers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | | | | |
Collapse
|
44
|
Abstract
Proteases play diverse roles in a variety of essential biological processes, both as non-specific catalysts of protein degradation and as highly specific agents that control physiologic events. Here, we review the mechanisms of substrate specificity employed by serine proteases and focus our discussion on coagulation proteases. We dissect the interplay between active site and exosite specificity and how substrate recognition is regulated allosterically by Na+ binding. We also draw attention to a functional polarity that exists in the serine protease fold, which sheds light on the structural linkages between the active site and exosites.
Collapse
Affiliation(s)
- M J Page
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
45
|
Ponnuraj K, Xu Y, Macon K, Moore D, Volanakis JE, Narayana SVL. Structural analysis of engineered Bb fragment of complement factor B: insights into the activation mechanism of the alternative pathway C3-convertase. Mol Cell 2004; 14:17-28. [PMID: 15068800 DOI: 10.1016/s1097-2765(04)00160-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2003] [Revised: 02/04/2004] [Accepted: 02/13/2004] [Indexed: 10/26/2022]
Abstract
The C-terminal fragment, Bb, of factor B combines with C3b to form the pivotal C3-convertase, C3bBb, of alternative complement pathway. Bb consists of a von Willebrand factor type A (vWFA) domain that is structurally similar to the I domains of integrins and a serine protease (SP) domain that is in inactive conformation. The structure of the C3bBb complex would be important in deciphering the activation mechanism of the SP domain. However, C3bBb is labile and not amenable to X-ray diffraction studies. We engineered a disulfide bond in the vWFA domain of Bb homologous to that shown to lock I domains in active conformation. The crystal structures of Bb(C428-C435) and its inhibitor complexes reveal that the adoption of the "active" conformation by the vWFA domain is not sufficient to activate the C3-convertase catalytic apparatus and also provide insights into the possible mode of C3-convertase activation.
Collapse
Affiliation(s)
- Karthe Ponnuraj
- Center for Biophysical Sciences and Engineering, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
46
|
Lorber B. The crystallization of biological macromolecules under microgravity: a way to more accurate three-dimensional structures? BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1599:1-8. [PMID: 12479400 DOI: 10.1016/s1570-9639(02)00403-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The crystallization of proteins and other biological particles (including nucleic acids, nucleo-protein complexes and large assemblies such as nucleosomes, ribosomal subunits or viruses) in a microgravity environment can produce crystals having lesser defects than crystals prepared under normal gravity on earth. Such microgravity-grown crystals can diffract X-rays to a higher resolution and have a lower mosaic spread. The inferred electron density maps can be richer in details owing to which more accurate three-dimensional structure models can be built. Major results reported in this field of research are reviewed. Novel ones obtained with the Advanced Protein Crystallization Facility are presented. For structural biology, practical applications and implications associated with crystallization and crystallography onboard the International Space Station are discussed.
Collapse
Affiliation(s)
- Bernard Lorber
- Département Mécanismes et Macromolécules de la Synthèse Protéique et Cristallogénèse' UPR 9002, Institut de Biologie Moléculaire et Cellulaire du CNRS, 15 rue René Descartes, F-67084 Strasbourg, France.
| |
Collapse
|
47
|
Metzler DE, Metzler CM, Sauke DJ. Biochemical Defense Mechanisms. Biochemistry 2001. [DOI: 10.1016/b978-012492543-4/50034-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
48
|
Hinshelwood J, Perkins SJ. Conformational changes during the assembly of factor B from its domains by (1)H NMR spectroscopy and molecular modelling: their relevance to the regulation of factor B activity. J Mol Biol 2000; 301:1267-85. [PMID: 10966820 DOI: 10.1006/jmbi.2000.4044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Factor B is a key component of the alternative pathway of complement and is cleaved by factor D into the Ba and Bb fragments in the presence of activated C3 (C3b or C3(H(2)O)). The Ba fragment contains three short consensus/complement repeat domains, while the Bb fragment contains a von Willebrand factor type A (vWF-A) domain and a serine protease (SP) domain, all three of which are implicated in multisite contacts with C3. The upfield-shifted signals in the (1)H NMR spectra of factor B, the Ba and Bb fragments, and the vWF-A and SP domains were used as sensitive conformational probes of their structures. Temperature studies and pH titrations showed that the Ba fragment and the vWF-A and SP domains had conformationally mobile structures. The comparison of the NMR spectra of the SP domains of both factor B and factor D showed that the factor D linewidths were broader than those for factor B, which may result from a range of proteolytically inactive conformations of factor D in the absence of substrate. The NMR spectra from the separate vWF-A and SP domains in combination with that of the Ba fragment generally accounted for that of intact factor B, apart from the perturbation of an upfield-shifted signal from the Ba fragment. A new upfield-shifted signal was observed in the Bb fragment that was not detected in the spectra for the vWF-A or SP domains or intact factor B. Ring current calculations based on homology models or crystal structures predicted that buried hydrophobic methyl-aromatic interactions probably accounted for the upfield-shifted signals, with many arising from the N-terminal subdomain of the SP domain to which the C terminus of the vWF-A domain is directly linked. It was concluded that: (1) the conformation of the free SP domain is better ordered in solution than that of factor D; (2) the conformation of the Ba fragment is affected by its incorporation into factor B; and (3) the proximity of the vWF-A and SP domains within the Bb fragment leads to a conformational change in which conserved charged residues may be important. Allosteric structural rearrangements in the SP domain as the result of its interactions with the vWF-A domain or the Ba fragment provide an explanation of the regulation of the catalytic activity of factor B.
Collapse
Affiliation(s)
- J Hinshelwood
- Department of Biochemistry and Molecular Biology, Royal Free Campus, Royal Free and University College Medical School, University College London, Rowland Hill Street, London, NW3 2PF, UK
| | | |
Collapse
|
49
|
Sahu A, Lambris JD. Complement inhibitors: a resurgent concept in anti-inflammatory therapeutics. IMMUNOPHARMACOLOGY 2000; 49:133-48. [PMID: 10904113 DOI: 10.1016/s0162-3109(00)80299-4] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In addition to its essential role in immune defense, the complement system contributes to tissue damage in many clinical conditions. Thus, there is a pressing need to develop therapeutically effective complement inhibitors to prevent these adverse effects. This concept, though old, received little scientific attention until recently. Data from animal models of diseases that have been produced using complement-deficient, knockout, and transgenic animals, as well as data demonstrating that complement proteins are produced in many important tissue sites (including the brain) have attracted the interest of many basic research scientists and applied scientists from the biotechnology field and larger pharmaceutical firms. This resurgence of interest has generated a wealth of new information in the field of complement inhibition. In this article, we comprehensively review up-to-date information in the field of complement inhibitors.
Collapse
Affiliation(s)
- A Sahu
- Protein Chemistry Laboratory, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | |
Collapse
|
50
|
Wang X, Terzyan S, Tang J, Loy JA, Lin X, Zhang XC. Human plasminogen catalytic domain undergoes an unusual conformational change upon activation. J Mol Biol 2000; 295:903-14. [PMID: 10656799 DOI: 10.1006/jmbi.1999.3397] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of the serine protease plasmin from its zymogen, plasminogen, is the key step in fibrinolysis leading to blood clot dissolution. It also plays critical roles in cell migration, such as in tumor metastasis. Here, we report the crystal structure of an inactive S741A mutant of human plasminogen catalytic domain at 2.0 A resolution. This structure permits a direct comparison with that of the plasmin catalytic unit. Unique conformational differences are present between these two structures that are not seen in other zymogen-enzyme pairs of the trypsin family. The functional significance of these differences and the structural basis of plasminogen activation is discussed in the light of this new structure.
Collapse
Affiliation(s)
- X Wang
- Oklahoma Medical Research Foundation, Crystallography Program, 825 N. E. 13(th) Street, Oklahoma City, OK, 73104, USA
| | | | | | | | | | | |
Collapse
|