Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Christoph Q. Schmidt is active.

Publication


Featured researches published by Christoph Q. Schmidt.


Blood | 2010

Association of factor H autoantibodies with deletions of CFHR1, CFHR3, CFHR4, and with mutations in CFH, CFI, CD46, and C3 in patients with atypical hemolytic uremic syndrome

Iain Moore; Lisa Strain; Isabel Y. Pappworth; David J. Kavanagh; Paul N. Barlow; Andrew P. Herbert; Christoph Q. Schmidt; Scott J. Staniforth; Lucy V. Holmes; Roy Ward; Lynn Morgan; Timothy H.J. Goodship; Kevin J. Marchbank

Factor H autoantibodies have been reported in approximately 10% of patients with atypical hemolytic uremic syndrome (aHUS) and are associated with deficiency of factor H-related proteins 1 and 3. In this study we examined the prevalence of factor H autoantibodies in the Newcastle cohort of aHUS patients, determined whether the presence of such autoantibodies is always associated with deficiency of factor H-related proteins 1 and 3, and examined whether such patients have additional susceptibility factors and/or mutations in the genes encoding complement regulator/activators. We screened 142 patients with aHUS and found factor H autoantibodies in 13 individuals (age 1-11 years). The presence of the autoantibodies was confirmed by Western blotting. By using multiplex ligation-dependent probe amplification we measured complement factor H-related (CFHR)1 and CFHR3 copy number. In 10 of the 13 patients there were 0 copies of CFHR1, and in 3 patients there were 2. In 3 of the patients with 0 copies of CFHR1 there was 1 copy of CFHR3, and these individuals exhibited a novel deletion incorporating CFHR1 and CFHR4. In 5 patients mutations were identified: 1 in CFH, 1 in CFI, 1 in CD46, and 2 in C3. The latter observation emphasizes that multiple concurrent factors may be necessary in individual patients for disease manifestation.


Journal of Immunology | 2008

A New Map of Glycosaminoglycan and C3b Binding Sites on Factor H

Christoph Q. Schmidt; Andrew P. Herbert; David J. Kavanagh; Carina Gandy; Christopher J. Fenton; Bärbel S. Blaum; Malcolm Lyon; Dušan Uhrín; Paul N. Barlow

Human complement factor H, consisting of 20 complement control protein (CCP) modules, is an abundant plasma glycoprotein. It prevents C3b amplification on self surfaces bearing certain polyanionic carbohydrates, while complement activation progresses on most other, mainly foreign, surfaces. Herein, locations of binding sites for polyanions and C3b are reexamined rigorously by overexpressing factor H segments, structural validation, and binding assays. As anticipated, constructs corresponding to CCPs 7–8 and 19–20 bind well in heparin-affinity chromatography. However, CCPs 8–9, previously reported to bind glycosaminoglycans, bind neither to heparin resin nor to heparin fragments in gel-mobility shift assays. Introduction of nonnative residues N-terminal to a construct containing CCPs 8–9, identical to those in proteins used in the previous report, converted this module pair to an artificially heparin-binding one. The module pair CCPs 12–13 does not bind heparin appreciably, notwithstanding previous suggestions to the contrary. We further checked CCPs 10–12, 11–14, 13–15, 10–15, and 8–15 for ability to bind heparin but found very low affinity or none. As expected, constructs corresponding to CCPs 1–4 and 19–20 bind C3b amine coupled to a CM5 chip (Kds of 14 and 3.5 μM, respectively) or a C1 chip (Kds of 10 and 4.5 μM, respectively). Constructs CCPs 7–8 and 6–8 exhibit measurable affinities for C3b according to surface plasmon resonance, although they are weak compared with CCPs 19–20. Contrary to expectations, none of several constructs encompassing modules from CCP 9 to 15 exhibited significant C3b binding in this assay. Thus, we propose a new functional map of factor H.


Nature Structural & Molecular Biology | 2011

Structural basis for engagement by complement factor H of C3b on a self surface.

Hugh P. Morgan; Christoph Q. Schmidt; Mara Guariento; Baerbel S. Blaum; Dominic Gillespie; Andrew P. Herbert; David J. Kavanagh; Haydyn D. T. Mertens; Dmitri I. Svergun; Conny M. Johansson; Dušan Uhrín; Paul N. Barlow; Jonathan P. Hannan

Complement factor H (FH) attenuates C3b molecules tethered by their thioester domains to self surfaces and thereby protects host tissues. Factor H is a cofactor for initial C3b proteolysis that ultimately yields a surface-attached fragment (C3d) corresponding to the thioester domain. We used NMR and X-ray crystallography to study the C3d–FH19–20 complex in atomic detail and identify glycosaminoglycan-binding residues in factor H module 20 of the C3d–FH19–20 complex. Mutagenesis justified the merging of the C3d–FH19–20 structure with an existing C3b–FH1–4 crystal structure. We concatenated the merged structure with the available FH6–8 crystal structure and new SAXS-derived FH1–4, FH8–15 and FH15–19 envelopes. The combined data are consistent with a bent-back factor H molecule that binds through its termini to two sites on one C3b molecule and simultaneously to adjacent polyanionic host-surface markers.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Complement receptor 1 is the host erythrocyte receptor for Plasmodium falciparum PfRh4 invasion ligand

Wai-Hong Tham; Danny W. Wilson; Sash Lopaticki; Christoph Q. Schmidt; Patience B. Tetteh-Quarcoo; Paul N. Barlow; Dave Richard; Jason Corbin; James G. Beeson; Alan F. Cowman

Plasmodium falciparum is responsible for the most severe form of malaria disease in humans, causing more than 1 million deaths each year. As an obligate intracellular parasite, P. falciparum’s ability to invade erythrocytes is essential for its survival within the human host. P. falciparum invades erythrocytes using multiple host receptor–parasite ligand interactions known as invasion pathways. Here we show that CR1 is the host erythrocyte receptor for PfRh4, a major P. falciparum ligand essential for sialic acid–independent invasion. PfRh4 and CR1 interact directly, with a Kd of 2.9 μM. PfRh4 binding is strongly correlated with the CR1 level on the erythrocyte surface. Parasite invasion via sialic acid–independent pathways is reduced in low-CR1 erythrocytes due to limited availability of this receptor on the surface. Furthermore, soluble CR1 can competitively block binding of PfRh4 to the erythrocyte surface and specifically inhibit sialic acid–independent parasite invasion. These results demonstrate that CR1 is an erythrocyte receptor used by the parasite ligand PfRh4 for P. falciparum invasion.


Clinical and Experimental Immunology | 2007

Translational Mini-Review Series on Complement Factor H: Structural and functional correlations for factor H

Christoph Q. Schmidt; Andrew P. Herbert; Henry G. Hocking; Dušan Uhrín; Paul N. Barlow

The 155‐kDa glycoprotein, complement factor H (CFH), is a regulator of complement activation that is abundant in human plasma. Three‐dimensional structures of over half the 20 complement control protein (CCP) modules in CFH have been solved in the context of single‐, double‐ and triple‐module segments. Proven binding sites for C3b occupy the N and C termini of this elongated molecule and may be brought together by a bend in CFH mediated by its central CCP modules. The C‐terminal CCP 20 is key to the ability of the molecule to adhere to polyanionic markers on self‐surfaces where CFH acts to regulate amplification of the alternative pathway of complement. The surface patch on CCP 20 that binds to model glycosaminoglycans has been mapped using nuclear magnetic resonance (NMR), as has a second glycosaminoglycan‐binding patch on CCP 7. These patches include many of the residue positions at which sequence variations have been linked to three complement‐mediated disorders: dense deposit disease, age‐related macular degeneration and atypical haemolytic uraemic syndrome. In one plausible model, CCP 20 anchors CFH to self‐surfaces via a C3b/polyanion composite binding site, CCP 7 acts as a ‘proof‐reader’ to help discriminate self‐ from non‐self patterns of sulphation, and CCPs 1–4 disrupt C3/C5 convertase formation and stability.


Journal of Biological Chemistry | 2007

Structure Shows that a Glycosaminoglycan and Protein Recognition Site in Factor H is Perturbed by Age-Related Macular Degeneration-Linked Single Nucleotide Polymorphism.

Andrew P. Herbert; Jon A. Deakin; Christoph Q. Schmidt; Bärbel S. Blaum; Claire Egan; Viviana P. Ferreira; Michael K. Pangburn; Malcolm Lyon; Dušan Uhrín; Paul N. Barlow

A common single nucleotide polymorphism in the factor H gene predisposes to age-related macular degeneration. Factor H blocks the alternative pathway of complement on self-surfaces bearing specific polyanions, including the glycosaminoglycan chains of proteoglycans. Factor H also binds C-reactive protein, potentially contributing to noninflammatory apoptotic processes. The at risk sequence contains His (rather than Tyr) at position 402 (384 in the mature protein), in the seventh of the 20 complement control protein (CCP) modules (CCP7) of factor H. We expressed both His402 and Tyr402 variants of CCP7, CCP7,8, and CCP6-8. We determined structures of His402 and Tyr402 CCP7 and showed them to be nearly identical. The side chains of His/Tyr402 have similar, solvent-exposed orientations far from interfaces with CCP6 and -8. Tyr402 CCP7 bound significantly more tightly than His402 CCP7 to a heparin affinity column as well as to defined-length sulfated heparin oligosaccharides employed in gel mobility shift assays. This observation is consistent with the position of the 402 side chain on the edge of one of two glycosaminoglycan-binding surface patches on CCP7 that we inferred on the basis of chemical shift perturbation studies with a sulfated heparin tetrasaccharide. According to surface plasmon resonance measurements, Tyr402 CCP6-8 binds significantly more tightly than His402 CCP6-8 to immobilized C-reactive protein. The data support a causal link between H402Y and age-related macular degeneration in which variation at position 402 modulates the response of factor H to age-related changes in the glycosaminoglycan composition and apoptotic activity of the macula.


Journal of Immunology | 2013

Rational Engineering of a Minimized Immune Inhibitor with Unique Triple-Targeting Properties

Christoph Q. Schmidt; Hongjun Bai; Zhuoer Lin; Antonio M. Risitano; Paul N. Barlow; Daniel Ricklin; John D. Lambris

Inadequate control of the complement system is the underlying or aggravating factor in many human diseases. Whereas treatment options that specifically target the alternative pathway (AP) of complement activation are considered highly desirable, no such option is available in the clinic. In this study, we present a successful example of protein engineering, guided by structural insight on the complement regulator factor H (FH), yielding a novel complement-targeted therapeutic (mini-FH) with clinical potential. Despite a 70% reduction in size, mini-FH retained and in some respects exceeded the regulatory activity and cell surface–recognition properties of its parent protein FH, including the recently described recognition of sites of oxidative stress. Importantly, the chosen design extended the functional spectrum of the inhibitor, as mini-FH showed increased binding to the surface-bound opsonins iC3b and C3dg when compared with FH. Thus, mini-FH is equipped with a unique and clinically valuable triple-targeting profile toward diseased host cells, through its binding to sites of ongoing complement activation, markers of oxidative damage, and host surface-specific polyanions. When assessed in a clinically relevant AP-mediated disease model of paroxysmal nocturnal hemoglobinuria, mini-FH largely outperformed FH and indicated advantages over clinically evaluated AP inhibitors. Thus, the rational engineering of a streamlined FH construct not only provided insight into the function of a key complement regulator, but also yielded a novel inhibitor that combines a triple-targeting approach with high AP-specific inhibitory activity (IC50 ∼ 40 nM), which may pave the way toward new options for the treatment of complement-mediated diseases.


Protein Expression and Purification | 2011

Production of biologically active complement factor H in therapeutically useful quantities

Christoph Q. Schmidt; Fern Slingsby; Anna Richards; Paul N. Barlow

Human complement factor H (FH), an abundant 155-kDa plasma glycoprotein with 40 disulphide bonds, regulates the alternative-pathway complement cascade. Mutations and single nucleotide polymorphisms in the FH gene predispose to development of age-related macular degeneration, atypical haemolytic uraemic syndrome and dense deposit disease. Supplementation with FH variants protective against disease is an enticing therapeutic prospect. Current sources of therapeutic FH are restricted to human blood plasma highlighting a need for recombinant material. Previously FH expression in cultured plant, mammalian or insect cells yielded protein amounts inadequate for full characterisation, and orders of magnitude below therapeutic usefulness. Here, the V62,Y402 variant of FH has been produced recombinantly (rFH) in Pichia pastoris cells. Codon-optimisation proved essential whilst exploitation of the yeast mating α-factor peptide ensured secretion. We thereby produced multiple 10s-of-milligram of rFH. Following endoglycosidase H digestion of N-linked glycans, rFH (with eight residual N-acetylglucosamine moieties) was purified on heparin-affinity resin and anion-exchange chromatography. Full-length rFH was verified by mass spectrometry and Western blot using monoclonal antibodies to the C-terminus. Recombinant FH is a single non-aggregated species (by dynamic light scattering) and fully functional in biochemical and biological assays. An additional version of rFH was produced in which eight N-glycosylation sequons were ablated by Asn-Gln substitutions resulting in a glycan-devoid product. Successful production of rFH in this potentially very highly expressing system makes production of therapeutically useful quantities economically viable. Furthermore, ease of genetic manipulation in P. pastoris would allow production of engineered FH versions with enhanced pharmacokinetic and pharmacodynamic properties.


Molecular Immunology | 2012

Factor H autoantibodies in membranoproliferative glomerulonephritis

Timothy H.J. Goodship; Isabel Y. Pappworth; Tibor Toth; Mark Denton; Kris Houlberg; Frances McCormick; David Warland; Iain Moore; Eva-Maria Hunze; Scott J. Staniforth; Christine Hayes; Danielle Paixão Cavalcante; David J. Kavanagh; Lisa Strain; Andrew P. Herbert; Christoph Q. Schmidt; Paul N. Barlow; Claire L. Harris; Kevin J. Marchbank

Factor H autoantibodies are found in ~10% of aHUS patients. Most are associated with complete deficiency of factor H related proteins 1/3 and bind to the C terminal recognition domain. MPGN, like aHUS, is characterised by complement activation. In this study we, therefore, examined the hypothesis that factor H autoantibodies are associated with MPGN. We screened sera from 16 MPGN patients and 100 normal controls using ELISA and detected strongly positive IgG factor H autoantibodies in 2 patients. One patient had type II (DDD) MPGN (male aged 24 yrs) with C3NeF and the other type I (female aged 26 yrs) with no detectable C3NeF. We identified the binding site of the autoantibodies using small SCR domain fragments in the ELISA and showed that the autoantibodies in both patients bound predominately to the N terminal complement regulatory domain of factor H. We measured CFHR 1/3 copy number using MLPA and showed that both patients had 2 copies of CFHR1 and 3. Finally, we examined the functionality of detected factor H autoantibodies using purified patient IgG and observed increased haemolysis when purified IgG from both patients was added to normal human sera prior to incubation with rabbit red blood cells. Thus, in a cohort of MPGN patients we have found a high titre of functionally significant factor H autoantibodies in two patients with MPGN. Antibody depleting therapy may have a role in such patients and we suggest that screening for factor H autoantibodies should be undertaken in all patients with MPGN.


Journal of Immunology | 2014

Identification of Factor H–like Protein 1 as the Predominant Complement Regulator in Bruch’s Membrane: Implications for Age-Related Macular Degeneration

Simon J. Clark; Christoph Q. Schmidt; Anne White; Svetlana Hakobyan; B. Paul Morgan; Paul N. Bishop

The tight regulation of innate immunity on extracellular matrix (ECM) is a vital part of immune homeostasis throughout the human body, and disruption to this regulation in the eye is thought to contribute directly to the progression of age-related macular degeneration (AMD). The plasma complement regulator factor H (FH) is thought to be the main regulator that protects ECM against damaging complement activation. However, in the present study we demonstrate that a truncated form of FH, called FH-like protein 1 (FHL-1), is the main regulatory protein in the layer of ECM under human retina, called Bruch’s membrane. Bruch’s membrane is a major site of AMD disease pathogenesis and where drusen, the hallmark lesions of AMD, form. We show that FHL-1 can passively diffuse through Bruch’s membrane, whereas the full sized, glycosylated, FH cannot. FHL-1 is largely bound to Bruch’s membrane through interactions with heparan sulfate, and we show that the common Y402H polymorphism in the CFH gene, associated with an increased risk of AMD, reduces the binding of FHL-1 to this heparan sulfate. We also show that FHL-1 is retained in drusen whereas FH coats the periphery of the lesions, perhaps inhibiting their clearance. Our results identify a novel mechanism of complement regulation in the human eye, which highlights potential new avenues for therapeutic strategies.

Collaboration


Dive into the Christoph Q. Schmidt's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Ricklin

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

John D. Lambris

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wai-Hong Tham

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan F. Cowman

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge