51 results on '"Herbert AP"'
Search Results
2. An Evaluation of the Complement-Regulating Activities of Human Complement Factor H (FH) Variants Associated With Age-Related Macular Degeneration.
- Author
-
Biggs RM, Makou E, Lauder S, Herbert AP, Barlow PN, and Katti SK
- Subjects
- Animals, Humans, Acceleration, Complement Membrane Attack Complex, Complement System Proteins, Sheep, Complement Factor H genetics, Macular Degeneration genetics
- Abstract
Purpose: Factor H (FH, encoded by CFH) prevents activation of the complement system's alternative pathway (AP) on host tissues. FH impedes C3 convertase (C3bBb) formation, accelerates C3bBb decay, and is a cofactor for factor I (FI)-catalyzed C3b cleavage. Numerous CFH variants are associated with age-related macular degeneration (AMD), but their functional consequences frequently remain undetermined. Here, we conduct functional comparisons between a control version of FH (not AMD linked) and 21 AMD-linked FH variants., Methods: Recombinantly produced, untagged, full-length FH versions were assayed for binding to C3b and decay acceleration of C3bBb using surface-plasmon resonance, FI-cofactor activity using a fluorescent probe of C3b integrity, suppression of C5b-9 assembly on an AP-activating surface, and inhibition of human AP-mediated lysis of sheep erythrocytes., Results: All versions were successfully purified despite below-average yields for Arg2Thr, Arg53Cys, Arg175Pro, Arg175Gln, Ile221Val, Tyr402His, Pro503Ala, Arg567Gly, Gly1194Asp, and Arg1210Cys. Compared to control FH, Arg2Thr, Leu3Val, Ser58Ala, Asp90Gly, Asp130Asn, Gln400Lys, Tyr402His, Gly650Val, Ser890Ile, and Thr965Met showed minimal functional differences. Arg1210C, Arg53His, Arg175Gln, Gly1194Asp, Pro503Ala, Arg53Cys, Arg576Gly, and Arg175Pro (in order of decreasing efficacy) underperformed, while Ile221Val, Arg303Gln, and Arg303Trp were "marginal." We newly identified variants toward the center of the molecule, Pro503Ala and Arg567Gly, as potentially pathogenic., Conclusions: Our approach could be extended to other variants of uncertain significance and to assays for noncanonical FH activities, aiming to facilitate selection of cohorts most likely to benefit from therapeutic FH. This is timely as recombinant therapeutic FH is in development for intravitreal treatment of AMD in patients with reduced FH functionality.
- Published
- 2022
- Full Text
- View/download PDF
3. A Novel Full-Length Recombinant Human Complement Factor H (CFH; GEM103) for the Treatment of Age-Related Macular Degeneration Shows Similar In Vitro Functional Activity to Native CFH.
- Author
-
Biggs RM, Makou E, Lauder S, Herbert AP, Barlow PN, and Katti SK
- Subjects
- Animals, Hemolysis, Humans, Polymorphism, Single Nucleotide, Sheep, Complement Factor H genetics, Complement Factor H metabolism, Macular Degeneration drug therapy, Macular Degeneration genetics, Macular Degeneration metabolism
- Abstract
Purpose: GEM103 is a recombinantly produced full-length version of the human complement factor H (CFH) under clinical investigation for treatment of age-related macular degeneration (AMD) in individuals carrying an AMD risk-associated genetic variant of CFH . This study aimed to investigate the complement pathway-related functions of GEM103 in comparison with those of native human CFH., Methods: Key biological activities of GEM103 and human serum-derived CFH (sdCFH) were compared using four independent functional assays. Assays of C3b binding and C3 convertase decay-accelerating activity (DAA) were performed by surface plasmon resonance (SPR). Cofactor activity (CA) was measured using 8-anilinonaphthalene-1-sulfonic acid as a fluorescent probe of C3b integrity. The abilities of GEM103 and sdCFH to protect sheep erythrocytes from hemolysis by CFH-depleted normal human serum were assessed colorimetrically., Results: In multiple SPR-based assays of C3b binding and DAA, the performance of GEM103 was consistently comparable to that of sdCFH across a range of matching concentrations. The EC
50 ± SD in the fluorescence-based fluid-phase CA assay was 0.21 ± 0.06 µM for GEM103 compared to 0.20 ± 0.09 µM for sdCFH. In hemolysis assays, the EC50 value of 0.33 ± 0.16 µM for GEM103 versus 0.46 ± 0.06 µM for sdCFH were not significantly different ( p = 0.81)., Conclusions: GEM103, a recombinant CFH developed by Gemini Therapeutics, shows activity profiles comparable to sdCFH in all complement-related assays employed in this study, suggesting that GEM103 is equivalent to the native glycoprotein in terms of its in vitro functional activity. These results support further study of GEM103 as a potential therapy for AMD.- Published
- 2022
- Full Text
- View/download PDF
4. Effects of Buprenorphine, Chlorhexidine, and Low-level Laser Therapy on Wound Healing in Mice.
- Author
-
Webb DR, Churchill SR, Hill GD, McGee CA, Shi M, King-Herbert AP, and Blankenship-Paris TL
- Subjects
- Animals, Chlorhexidine, Mice, Wound Healing, Anti-Infective Agents, Local, Buprenorphine, Laser Therapy, Low-Level Light Therapy
- Abstract
Systemic buprenorphine and topical antiseptics such as chlorhexidine are frequently used in research animals to aid in pain control and to reduce infection, respectively. These therapeutics are controversial, especially when used in wound healing studies, due to conflicting data suggesting that they delay wound healing. Low-level laser therapy (LLLT) has been used to aid in wound healing without exerting the systemic effects of therapies such as buprenorphine. We conducted 2 studies to investigate the effects of these common treatment modalities on the rate of wound healing in mice. The first study used models of punch biopsy and dermal abrasion to assess whether buprenorphine HCl or 0.12% chlorhexidine delayed wound healing. The second study investigated the effects of sustained-released buprenorphine, 0.05% chlorhexidine, and LLLT on excisional wound healing. The rate of wound healing was assessed by obtaining photographs on days 0, 2, 4, 7, and 9 for the punch biopsy model in study 1, days 0, 1, 2, 4, 6, 8, 11, and 13 for the dermal abrasion model in study 1, and days 0, 3, 6, and 10 for the mice in study 2. Image J software was used to analyze the photographed wounds to determine the wound area. When comparing the wound area on the above days to the original wound area, no significant differences in healing were observed for any of the treatment groups at any time period for either study. Given the results of these studies, we believe that systemic buprenorphine, topical chlorhexidine, and LLLT can be used without impairing or delaying wound healing in mice.
- Published
- 2021
- Full Text
- View/download PDF
5. Murine Factor H Co-Produced in Yeast With Protein Disulfide Isomerase Ameliorated C3 Dysregulation in Factor H-Deficient Mice.
- Author
-
Kerr H, Herbert AP, Makou E, Abramczyk D, Malik TH, Lomax-Browne H, Yang Y, Pappworth IY, Denton H, Richards A, Marchbank KJ, Pickering MC, and Barlow PN
- Subjects
- Animals, Gene Expression, Immunomodulation, Mice, Mice, Knockout, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins isolation & purification, Yeasts genetics, Yeasts metabolism, Complement C3 immunology, Complement C3 metabolism, Complement Factor H biosynthesis, Complement Factor H deficiency, Protein Disulfide-Isomerases metabolism, Recombinant Proteins metabolism
- Abstract
Recombinant human factor H (hFH) has potential for treating diseases linked to aberrant complement regulation including C3 glomerulopathy (C3G) and dry age-related macular degeneration. Murine FH (mFH), produced in the same host, is useful for pre-clinical investigations in mouse models of disease. An abundance of FH in plasma suggests high doses, and hence microbial production, will be needed. Previously, Pichia pastoris produced useful but modest quantities of hFH. Herein, a similar strategy yielded miniscule quantities of mFH. Since FH has 40 disulfide bonds, we created a P. pastoris strain containing a methanol-inducible codon-modified gene for protein-disulfide isomerase (PDI) and transformed this with codon-modified DNA encoding mFH under the same promoter. What had been barely detectable yields of mFH became multiple 10s of mg/L. Our PDI-overexpressing strain also boosted hFH overproduction, by about tenfold. These enhancements exceeded PDI-related production gains reported for other proteins, all of which contain fewer disulfide-stabilized domains. We optimized fermentation conditions, purified recombinant mFH, enzymatically trimmed down its (non-human) N-glycans, characterised its functions in vitro and administered it to mice. In FH-knockout mice, our de-glycosylated recombinant mFH had a shorter half-life and induced more anti-mFH antibodies than mouse serum-derived, natively glycosylated, mFH. Even sequential daily injections of recombinant mFH failed to restore wild-type levels of FH and C3 in mouse plasma beyond 24 hours after the first injection. Nevertheless, mFH functionality appeared to persist in the glomerular basement membrane because C3-fragment deposition here, a hallmark of C3G, remained significantly reduced throughout and beyond the ten-day dosing regimen., Competing Interests: KM has received consultancy or research income from Gemini Therapeutics Inc, Freeline Therapeutics, MPM Capital, Idorsia Pharmaceuticals Ltd and Catalyst Biosciences. PB is a scientific co-founder of, and has received consultancy and research income from, Gemini Therapeutics Inc. AH is founder and Chief Scientific Officer of Invizius. EM is currently an employee of Invizius. AR has been employed at GlaxoSmithKline since October 2014. Results reported in this work were undertaken during her Wellcome Trust Intermediate Clinical Fellowship. Her spouse, David Kavanagh, is head of the National Renal Complement Therapeutics Centre, UK, and a board member and scientific advisor to Gyroscope Therapeutics Ltd. PNB and AR are co-authors on a recombinant CFH patent application (EP10803266A). The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Kerr, Herbert, Makou, Abramczyk, Malik, Lomax-Browne, Yang, Pappworth, Denton, Richards, Marchbank, Pickering and Barlow.)
- Published
- 2021
- Full Text
- View/download PDF
6. An Engineered Complement Factor H Construct for Treatment of C3 Glomerulopathy.
- Author
-
Yang Y, Denton H, Davies OR, Smith-Jackson K, Kerr H, Herbert AP, Barlow PN, Pickering MC, and Marchbank KJ
- Subjects
- Animals, Complement Factor H chemical synthesis, Complement Factor H genetics, Complement Pathway, Alternative, Cricetinae, Glomerular Basement Membrane metabolism, Glomerulonephritis, Membranoproliferative drug therapy, Half-Life, Mice, Protein Binding, Protein Engineering, Complement C3 metabolism, Complement C3b metabolism, Complement Factor H metabolism, Complement Factor H pharmacokinetics, Glomerulonephritis, Membranoproliferative metabolism
- Abstract
Background C3 glomerulopathy (C3G) is associated with dysregulation of the alternative pathway of complement activation, and treatment options for C3G remain limited. Complement factor H (FH) is a potent regulator of the alternative pathway and might offer a solution, but the mass and complexity of FH makes generation of full-length FH far from trivial. We previously generated a mini-FH construct, with FH short consensus repeats 1-5 linked to repeats 18-20 (FH
1-5^18-20 ), that was effective in experimental C3G. However, the serum t1/2 of FH1-5^18-20 was significantly shorter than that of serum-purified FH. Methods We introduced the oligomerization domain of human FH-related protein 1 (denoted by R1-2) at the carboxy or amino terminus of human FH1-5^18-20 to generate two homodimeric mini-FH constructs (FHR1-2^1-5^18-20 and FH1-5^18-20^R1-2 , respectively) in Chinese hamster ovary cells and tested these constructs using binding, fluid-phase, and erythrocyte lysis assays, followed by experiments in FH-deficient Cfh-/- mice. Results FHR1-2^1-5^18-20 and FH1-5^18-20^R1-2 homodimerized in solution and displayed avid binding profiles on clustered C3b surfaces, particularly FHR1-2^1-5^18-20 Each construct was >10-fold more effective than FH at inhibiting cell surface complement activity in vitro and restricted glomerular basement membrane C3 deposition in vivo significantly better than FH or FH1-5^18-20 FH1-5^18-20^R1-2 had a C3 breakdown fragment binding profile similar to that of FH, a >5-fold increase in serum t1/2 compared with that of FH1-5^18-20 , and significantly better retention in the kidney than FH or FH1-5^18-20 Conclusions FH1-5^18-20^R1-2 may have utility as a treatment option for C3G or other complement-mediated diseases., (Copyright © 2018 by the American Society of Nephrology.)- Published
- 2018
- Full Text
- View/download PDF
7. Factor H C-Terminal Domains Are Critical for Regulation of Platelet/Granulocyte Aggregate Formation.
- Author
-
Blatt AZ, Saggu G, Cortes C, Herbert AP, Kavanagh D, Ricklin D, Lambris JD, and Ferreira VP
- Abstract
Platelet/granulocyte aggregates (PGAs) increase thromboinflammation in the vasculature, and PGA formation is tightly controlled by the complement alternative pathway (AP) negative regulator, Factor H (FH). Mutations in FH are associated with the prothrombotic disease atypical hemolytic uremic syndrome (aHUS), yet it is unknown whether increased PGA formation contributes to the thrombosis seen in patients with aHUS. Here, flow cytometry assays were used to evaluate the effects of aHUS-related mutations on FH regulation of PGA formation and characterize the mechanism. Utilizing recombinant fragments of FH spanning the entire length of the protein, we mapped the regions of FH most critical for limiting AP activity on the surface of isolated human platelets and neutrophils, as well as the regions most critical for regulating PGA formation in human whole blood stimulated with thrombin receptor-activating peptide (TRAP). FH domains 19-20 were the most critical for limiting AP activity on platelets, neutrophils, and at the platelet/granulocyte interface. The role of FH in PGA formation was attributed to its ability to regulate AP-mediated C5a generation. AHUS-related mutations in domains 19-20 caused differential effects on control of PGA formation and AP activity on platelets and neutrophils. Our data indicate FH C-terminal domains are key for regulating PGA formation, thus increased FH protection may have a beneficial impact on diseases characterized by increased PGA formation, such as cardiovascular disease. Additionally, aHUS-related mutations in domains 19-20 have varying effects on control of TRAP-mediated PGA formation, suggesting that some, but not all, aHUS-related mutations may cause increased PGA formation that contributes to excessive thrombosis in patients with aHUS.
- Published
- 2017
- Full Text
- View/download PDF
8. Disease-linked mutations in factor H reveal pivotal role of cofactor activity in self-surface-selective regulation of complement activation.
- Author
-
Kerr H, Wong E, Makou E, Yang Y, Marchbank K, Kavanagh D, Richards A, Herbert AP, and Barlow PN
- Subjects
- Amino Acid Substitution, Animals, Atypical Hemolytic Uremic Syndrome metabolism, Bacterial Proteins chemistry, Bacterial Proteins genetics, Bacterial Proteins metabolism, Binding Sites, Complement C3 Convertase, Alternative Pathway chemistry, Complement C3 Convertase, Alternative Pathway genetics, Complement C3 Convertase, Alternative Pathway metabolism, Complement C3d chemistry, Complement C3d genetics, Complement C3d metabolism, Complement Factor H chemistry, Complement Factor H genetics, Complement Factor H metabolism, Complement Factor I chemistry, Complement Factor I genetics, Complement Factor I metabolism, Erythrocytes chemistry, Hemolysis, Humans, Immobilized Proteins chemistry, Immobilized Proteins genetics, Immobilized Proteins metabolism, Macular Degeneration metabolism, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Protein Interaction Domains and Motifs, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Sheep, Domestic, Solubility, Streptococcus pneumoniae metabolism, Surface Properties, Atypical Hemolytic Uremic Syndrome genetics, Complement Activation, Macular Degeneration genetics, Mutation
- Abstract
Spontaneous activation enables the complement system to respond very rapidly to diverse threats. This activation is efficiently suppressed by complement factor H (CFH) on self-surfaces but not on foreign surfaces. The surface selectivity of CFH, a soluble protein containing 20 complement-control protein modules (CCPs 1-20), may be compromised by disease-linked mutations. However, which of the several functions of CFH drives this self-surface selectivity remains unknown. To address this, we expressed human CFH mutants in Pichia pastoris We found that recombinant I62-CFH (protective against age-related macular degeneration) and V62-CFH functioned equivalently, matching or outperforming plasma-derived CFH, whereas R53H-CFH, linked to atypical hemolytic uremic syndrome (aHUS), was defective in C3bBb decay-accelerating activity (DAA) and factor I cofactor activity (CA). The aHUS-linked CCP 19 mutant D1119G-CFH had virtually no CA on (self-like) sheep erythrocytes ( E
S ) but retained DAA. The aHUS-linked CCP 20 mutant S1191L/V1197A-CFH (LA-CFH) had dramatically reduced CA on ES but was less compromised in DAA. D1119G-CFH and LA-CFH both performed poorly at preventing complement-mediated hemolysis of ES PspCN, a CFH-binding Streptococcus pneumoniae protein domain, binds CFH tightly and increases accessibility of CCPs 19 and 20. PspCN did not improve the DAA of any CFH variant on ES Conversely, PspCN boosted the CA, on ES , of I62-CFH, R53H-CFH, and LA-CFH and also enhanced hemolysis protection by I62-CFH and LA-CFH. We conclude that CCPs 19 and 20 are critical for efficient CA on self-surfaces but less important for DAA. Exposing CCPs 19 and 20 with PspCN and thus enhancing CA on self-surfaces may reverse deficiencies of some CFH variants., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2017
- Full Text
- View/download PDF
9. Impact of Environmental Enrichment Devices on NTP In Vivo Studies.
- Author
-
Churchill SR, Morgan DL, Kissling GE, Travlos GS, and King-Herbert AP
- Subjects
- Animals, Eating physiology, Female, Male, Mice, Rats, Rats, Sprague-Dawley, Toxicity Tests, Animal Welfare statistics & numerical data, Behavior, Animal physiology, Body Weight physiology, Housing, Animal statistics & numerical data
- Abstract
The goal of this study was to determine whether the use of nesting material or polycarbonate shelters as enrichment devices would have an impact on end points commonly measured during the conduct of the National Toxicology Program (NTP) 13-week studies. The study design was consistent with the NTP 13-week toxicity studies. Harlan Sprague-Dawley (HSD) rats and their offspring and B6C3F1/N mice were assigned to control (unenriched) and enriched experimental groups. Body weight, food and water consumption, behavioral observations, fecal content, clinical pathology, gross pathology, organ weights, and histopathology were evaluated. Enriched male mice and male and female rats exhibited decreased feed intake without a subsequent decrease in body weight; this may have been the result of the nesting material reducing the effect of cold stress, thereby allowing for more efficient use of feed. There were statistical differences in some hematological parameters; however, these were not considered physiologically relevant since all values were within the normal range. Gross pathology and histopathological findings were background changes and were not considered enrichment-related. Nesting material and shelters were used frequently and consistently and allowed animals to display species-typical behavior. There was no significant impact on commonly measured end points in HSD rats and B6C3F1/N mice given enrichment devices., (© The Author(s) 2016.)
- Published
- 2016
- Full Text
- View/download PDF
10. Complement Evasion Mediated by Enhancement of Captured Factor H: Implications for Protection of Self-Surfaces from Complement.
- Author
-
Herbert AP, Makou E, Chen ZA, Kerr H, Richards A, Rappsilber J, and Barlow PN
- Subjects
- Bacterial Proteins immunology, Complement C3b immunology, Complement Factor H immunology, Hemoglobinuria, Paroxysmal immunology, Humans, Protein Structure, Tertiary, Streptococcus pneumoniae immunology, Bacterial Proteins chemistry, Complement C3b chemistry, Complement Factor H chemistry, Streptococcus pneumoniae chemistry
- Abstract
In an attempt to evade annihilation by the vertebrate complement system, many microbes capture factor H (FH), the key soluble complement-regulating protein in human plasma. However, FH is normally an active complement suppressor exclusively on self-surfaces and this selective action of FH is pivotal to self versus non-self discrimination by the complement system. We investigated whether the bacterially captured FH becomes functionally enhanced and, if so, how this is achieved at a structural level. We found, using site-directed and truncation mutagenesis, surface plasmon resonance, nuclear magnetic resonance spectroscopy, and cross-linking and mass spectrometry, that the N-terminal domain of Streptococcus pneumoniae protein PspC (PspCN) not only binds FH extraordinarily tightly but also holds it in a previously uncharacterized conformation. Functional enhancement arises from exposure of a C-terminal cryptic second binding site in FH for C3b, the activation-specific fragment of the pivotal complement component, C3. This conformational change of FH doubles its affinity for C3b and increases 5-fold its ability to accelerate decay of the binary enzyme (C3bBb) responsible for converting C3 to C3b in an amplification loop. Despite not sharing critical FH-binding residues, PspCNs from D39 and Tigr4 S. pneumoniae exhibit similar FH-anchoring and enhancing properties. We propose that these bacterial proteins mimic molecular markers of self-surfaces, providing a compelling hypothesis for how FH prevents complement-mediated injury to host tissue while lacking efficacy on virtually all other surfaces. In hemolysis assays with 2-aminoethylisothiouronium bromide-treated erythrocytes that recapitulate paroxysmal nocturnal hemoglobinuria, PspCN enhanced protection of cells by FH, suggesting a new paradigm for therapeutic complement suppression., (Copyright © 2015 The Authors.)
- Published
- 2015
- Full Text
- View/download PDF
11. Creating functional sophistication from simple protein building blocks, exemplified by factor H and the regulators of complement activation.
- Author
-
Makou E, Herbert AP, and Barlow PN
- Subjects
- Animals, Binding Sites, Complement Activating Enzymes genetics, Complement Activating Enzymes metabolism, Complement Factor H chemistry, Complement Factor H genetics, Complement Factor H metabolism, Complement Inactivating Agents chemistry, Complement Inactivating Agents metabolism, Complement Inactivating Agents pharmacology, Complement Inactivator Proteins genetics, Complement Inactivator Proteins metabolism, Humans, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Protein Conformation, Protein Interaction Domains and Motifs, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Recombinant Fusion Proteins pharmacology, Complement Activating Enzymes chemistry, Complement Activation, Complement Inactivator Proteins chemistry, Drug Design, Models, Molecular, Protein Engineering
- Abstract
Complement control protein modules (CCPs) occur in numerous functionally diverse extracellular proteins. Also known as short consensus repeats (SCRs) or sushi domains each CCP contains approximately 60 amino acid residues, including four consensus cysteines participating in two disulfide bonds. Varying in length and sequence, CCPs adopt a β-sandwich type fold and have an overall prolate spheroidal shape with N- and C-termini lying close to opposite poles of the long axis. CCP-containing proteins are important as cytokine receptors and in neurotransmission, cell adhesion, blood clotting, extracellular matrix formation, haemoglobin metabolism and development, but CCPs are particularly well represented in the vertebrate complement system. For example, factor H (FH), a key soluble regulator of the alternative pathway of complement activation, is made up entirely from a chain of 20 CCPs joined by short linkers. Collectively, therefore, the 20 CCPs of FH must mediate all its functional capabilities. This is achieved via collaboration and division of labour among these modules. Structural studies have illuminated the dynamic architectures that allow FH and other CCP-rich proteins to perform their biological functions. These are largely the products of a highly varied set of intramolecular interactions between CCPs. The CCP can act as building block, spacer, highly versatile recognition site or dimerization mediator. Tandem CCPs may form composite binding sites or contribute to flexible, rigid or conformationally 'switchable' segments of the parent proteins., (© 2015 Authors; published by Portland Press Limited.)
- Published
- 2015
- Full Text
- View/download PDF
12. Structural basis for sialic acid-mediated self-recognition by complement factor H.
- Author
-
Blaum BS, Hannan JP, Herbert AP, Kavanagh D, Uhrín D, and Stehle T
- Subjects
- Animals, Binding Sites, Carbohydrate Sequence, Complement C3b metabolism, Complement Factor H metabolism, Complement Pathway, Alternative drug effects, Conserved Sequence, Hemolysis drug effects, Hemolytic-Uremic Syndrome genetics, Humans, Mice, Models, Molecular, Molecular Sequence Data, N-Acetylneuraminic Acid metabolism, Polysaccharides pharmacology, Sheep, Complement Factor H chemistry, N-Acetylneuraminic Acid chemistry
- Abstract
The serum protein complement factor H (FH) ensures downregulation of the complement alternative pathway, a branch of innate immunity, upon interaction with specific glycans on host cell surfaces. Using ligand-based NMR, we screened a comprehensive set of sialylated glycans for binding to FH and solved the crystal structure of a ternary complex formed by the two C-terminal domains of FH, a sialylated trisaccharide and the complement C3b thioester-containing domain. Key residues in the sialic acid binding site are conserved from mice to men, and residues linked to atypical hemolytic uremic syndrome cluster within this binding site, suggesting a possible role for sialic acid as a host marker also in other mammals and a critical role in human renal complement homeostasis. Unexpectedly, the FH sialic acid binding site is structurally homologous to the binding sites of two evolutionarily unrelated proteins. The crystal structure also advances our understanding of bacterial immune evasion strategies.
- Published
- 2015
- Full Text
- View/download PDF
13. Characterization of a factor H mutation that perturbs the alternative pathway of complement in a family with membranoproliferative GN.
- Author
-
Wong EK, Anderson HE, Herbert AP, Challis RC, Brown P, Reis GS, Tellez JO, Strain L, Fluck N, Humphrey A, Macleod A, Richards A, Ahlert D, Santibanez-Koref M, Barlow PN, Marchbank KJ, Harris CL, Goodship TH, and Kavanagh D
- Subjects
- Animals, Complement Factor H chemistry, Complement Factor H immunology, Complement Pathway, Alternative immunology, Crystallography, X-Ray, Erythrocytes cytology, Family Health, Female, Haplotypes, Hereditary Complement Deficiency Diseases, Heterozygote, Humans, Kidney Diseases immunology, Male, Pedigree, Polymorphism, Genetic, Protein Structure, Tertiary, Sheep, Structure-Activity Relationship, Complement Factor H deficiency, Complement Factor H genetics, Complement Pathway, Alternative genetics, Erythrocytes immunology, Glomerulonephritis, Membranoproliferative genetics, Glomerulonephritis, Membranoproliferative immunology, Kidney Diseases genetics
- Abstract
Complement C3 activation is a characteristic finding in membranoproliferative GN (MPGN). This activation can be caused by immune complex deposition or an acquired or inherited defect in complement regulation. Deficiency of complement factor H has long been associated with MPGN. More recently, heterozygous genetic variants have been reported in sporadic cases of MPGN, although their functional significance has not been assessed. We describe a family with MPGN and acquired partial lipodystrophy. Although C3 nephritic factor was shown in family members with acquired partial lipodystrophy, it did not segregate with the renal phenotype. Genetic analysis revealed a novel heterozygous mutation in complement factor H (R83S) in addition to known risk polymorphisms carried by individuals with MPGN. Patients with MPGN had normal levels of factor H, and structural analysis of the mutant revealed only subtle alterations. However, functional analysis revealed profoundly reduced C3b binding, cofactor activity, and decay accelerating activity leading to loss of regulation of the alternative pathway. In summary, this family showed a confluence of common and rare functionally significant genetic risk factors causing disease. Data from our analysis of these factors highlight the role of the alternative pathway of complement in MPGN., (Copyright © 2014 by the American Society of Nephrology.)
- Published
- 2014
- Full Text
- View/download PDF
14. Functional anatomy of complement factor H.
- Author
-
Makou E, Herbert AP, and Barlow PN
- Subjects
- Amino Acid Motifs, Binding Sites, Complement C3b chemistry, Complement C3b physiology, Complement Factor H chemistry, Humans, Models, Molecular, Protein Binding, Protein Interaction Domains and Motifs, Complement Activation, Complement Factor H physiology
- Abstract
Factor H (FH) is a soluble regulator of the proteolytic cascade at the core of the evolutionarily ancient vertebrate complement system. Although FH consists of a single chain of similar protein modules, it has a demanding job description. Its chief role is to prevent complement-mediated injury to healthy host cells and tissues. This entails recognition of molecular patterns on host surfaces combined with control of one of nature's most dangerous examples of a positive-feedback loop. In this way, FH modulates, where and when needed, an amplification process that otherwise exponentially escalates the production of the pro-inflammatory, pro-phagocytic, and pro-cytolytic cleavage products of complement proteins C3 and C5. Mutations and single-nucleotide polymorphisms in the FH gene and autoantibodies against FH predispose individuals to diseases, including age-related macular degeneration, dense-deposit disease, and atypical hemolytic uremic syndrome. Moreover, deletions or variations of genes for FH-related proteins also influence the risk of disease. Numerous pathogens hijack FH and use it for self-defense. As reviewed herein, a molecular understanding of FH function is emerging. While its functional oligomeric status remains uncertain, progress has been achieved in characterizing its three-dimensional architecture and, to a lesser extent, its intermodular flexibility. Models are proposed, based on the reconciliation of older data with a wealth of recent evidence, in which a latent circulating form of FH is activated by its principal target, C3b tethered to a self-surface. Such models suggest hypotheses linking sequence variations to pathophysiology, but improved, more quantitative, functional assays and rigorous data analysis are required to test these ideas.
- Published
- 2013
- Full Text
- View/download PDF
15. Tissue-specific host recognition by complement factor H is mediated by differential activities of its glycosaminoglycan-binding regions.
- Author
-
Clark SJ, Ridge LA, Herbert AP, Hakobyan S, Mulloy B, Lennon R, Würzner R, Morgan BP, Uhrín D, Bishop PN, and Day AJ
- Subjects
- Adult, Aged, Aged, 80 and over, Amino Acid Substitution, Atypical Hemolytic Uremic Syndrome, Autopsy, Binding Sites, Complement Activation genetics, Complement Factor H chemistry, Complement Factor H metabolism, Escherichia coli genetics, Eye pathology, Female, Hemolytic-Uremic Syndrome metabolism, Hemolytic-Uremic Syndrome pathology, Humans, Kidney pathology, Macular Degeneration metabolism, Macular Degeneration pathology, Male, Middle Aged, Mutation, Organ Specificity, Protein Binding, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Complement Factor H genetics, Eye metabolism, Hemolytic-Uremic Syndrome genetics, Heparitin Sulfate metabolism, Kidney metabolism, Macular Degeneration genetics
- Abstract
Complement factor H (CFH) regulates complement activation in host tissues through its recognition of polyanions, which mediate CFH binding to host cell surfaces and extracellular matrix, promoting the deactivation of deposited C3b. These polyanions include heparan sulfate (HS), a glycosaminoglycan with a highly diverse range of structures, for which two regions of CFH (CCP6-8 and CCP19-20) have been implicated in HS binding. Mutations/polymorphisms within these glycosaminoglycan-binding sites have been associated with age-related macular degeneration (AMD) and atypical hemolytic uremic syndrome. In this study, we demonstrate that CFH has tissue-specific binding properties mediated through its two HS-binding regions. Our data show that the CCP6-8 region of CFH binds more strongly to heparin (a highly sulfated form of HS) than CCP19-20, and that their sulfate specificities are different. Furthermore, the HS binding site in CCP6-8, which is affected by the AMD-associated Y402H polymorphism, plays the principal role in host tissue recognition in the human eye, whereas the CCP19-20 region makes the major contribution to the binding of CFH in the human kidney. This helps provide a biochemical explanation for the genetic basis of tissue-specific diseases such as AMD and atypical hemolytic uremic syndrome, and leads to a better understanding of the pathogenic mechanisms for these diseases of complement dysregulation.
- Published
- 2013
- Full Text
- View/download PDF
16. Combination of factor H mutation and properdin deficiency causes severe C3 glomerulonephritis.
- Author
-
Lesher AM, Zhou L, Kimura Y, Sato S, Gullipalli D, Herbert AP, Barlow PN, Eberhardt HU, Skerka C, Zipfel PF, Hamano T, Miwa T, Tung KS, and Song WC
- Subjects
- Animals, Complement C3 metabolism, Complement Factor H deficiency, Complement Factor H genetics, Complement Pathway, Alternative, Disease Models, Animal, Glomerulonephritis, Membranoproliferative metabolism, Glomerulonephritis, Membranoproliferative pathology, Hereditary Complement Deficiency Diseases, Humans, Kidney Glomerulus ultrastructure, Mice, Mice, Inbred C57BL, Mutation, Glomerulonephritis, Membranoproliferative genetics, Kidney Diseases genetics, Properdin deficiency
- Abstract
Factor H (fH) and properdin both modulate complement; however, fH inhibits activation, and properdin promotes activation of the alternative pathway of complement. Mutations in fH associate with several human kidney diseases, but whether inhibiting properdin would be beneficial in these diseases is unknown. Here, we found that either genetic or pharmacological blockade of properdin, which we expected to be therapeutic, converted the mild C3 GN of an fH-mutant mouse to a lethal C3 GN with features of human dense deposit disease. We attributed this phenotypic change to a differential effect of properdin on the dynamics of alternative pathway complement activation in the fluid phase and the cell surface in the fH-mutant mice. Thus, in fH mutation-related C3 glomerulopathy, additional factors that impact the activation of the alternative pathway of complement critically determine the nature and severity of kidney pathology. These results show that therapeutic manipulation of the complement system requires rigorous disease-specific target validation.
- Published
- 2013
- Full Text
- View/download PDF
17. Solution structure of CCP modules 10-12 illuminates functional architecture of the complement regulator, factor H.
- Author
-
Makou E, Mertens HD, Maciejewski M, Soares DC, Matis I, Schmidt CQ, Herbert AP, Svergun DI, and Barlow PN
- Subjects
- Humans, Models, Molecular, Protein Conformation, Complement Factor H chemistry, Complement Factor H metabolism, Magnetic Resonance Spectroscopy
- Abstract
The 155-kDa plasma glycoprotein factor H (FH), which consists of 20 complement control protein (CCP) modules, protects self-tissue but not foreign organisms from damage by the complement cascade. Protection is achieved by selective engagement of FH, via CCPs 1-4, CCPs 6-8 and CCPs 19-20, with polyanion-rich host surfaces that bear covalently attached, activation-specific, fragments of complement component C3. The role of intervening CCPs 9-18 in this process is obscured by lack of structural knowledge. We have concatenated new high-resolution solution structures of overlapping recombinant CCP pairs, 10-11 and 11-12, to form a three-dimensional structure of CCPs 10-12 and validated it by small-angle X-ray scattering of the recombinant triple-module fragment. Superimposing CCP 12 of this 10-12 structure with CCP 12 from the previously solved CCP 12-13 structure yielded an S-shaped structure for CCPs 10-13 in which modules are tilted by 80-110° with respect to immediate neighbors, but the bend between CCPs 10 and 11 is counter to the arc traced by CCPs 11-13. Including this four-CCP structure in interpretation of scattering data for the longer recombinant segments, CCPs 10-15 and 8-15, implied flexible attachment of CCPs 8 and 9 to CCP 10 but compact and intimate arrangements of CCP 14 with CCPs 12, 13 and 15. Taken together with difficulties in recombinant production of module pairs 13-14 and 14-15, the aberrant structure of CCP 13 and the variability of 13-14 linker sequences among orthologues, a structural dependency of CCP 14 on its neighbors is suggested; this has implications for the FH mechanism., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
18. Solution NMR structure of the Ca2+-bound N-terminal domain of CaBP7: a regulator of golgi trafficking.
- Author
-
McCue HV, Patel P, Herbert AP, Lian LY, Burgoyne RD, and Haynes LP
- Subjects
- Amino Acid Sequence, Animals, Binding Sites genetics, Blotting, Western, Calcium-Binding Proteins chemistry, Calcium-Binding Proteins genetics, Cattle, Circular Dichroism, Humans, Hydrophobic and Hydrophilic Interactions, Minor Histocompatibility Antigens, Models, Molecular, Molecular Sequence Data, Phosphotransferases (Alcohol Group Acceptor) genetics, Phosphotransferases (Alcohol Group Acceptor) metabolism, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Protein Transport, Sequence Homology, Amino Acid, Solutions, Surface Properties, Calcium metabolism, Calcium-Binding Proteins metabolism, Golgi Apparatus metabolism, Magnetic Resonance Spectroscopy methods
- Abstract
Calcium-binding protein 7 (CaBP7) is a member of the calmodulin (CaM) superfamily that harbors two high affinity EF-hand motifs and a C-terminal transmembrane domain. CaBP7 has been previously shown to interact with and modulate phosphatidylinositol 4-kinase III-β (PI4KIIIβ) activity in in vitro assays and affects vesicle transport in neurons when overexpressed. Here we show that the N-terminal domain (NTD) of CaBP7 is sufficient to mediate the interaction of CaBP7 with PI4KIIIβ. CaBP7 NTD encompasses the two high affinity Ca(2+) binding sites, and structural characterization through multiangle light scattering, circular dichroism, and NMR reveals unique properties for this domain. CaBP7 NTD binds specifically to Ca(2+) but not Mg(2+) and undergoes significant conformational changes in both secondary and tertiary structure upon Ca(2+) binding. The Ca(2+)-bound form of CaBP7 NTD is monomeric and exhibits an open conformation similar to that of CaM. Ca(2+)-bound CaBP7 NTD has a solvent-exposed hydrophobic surface that is more expansive than observed in CaM or CaBP1. Within this hydrophobic pocket, there is a significant reduction in the number of methionine residues that are conserved in CaM and CaBP1 and shown to be important for target recognition. In CaBP7 NTD, these residues are replaced with isoleucine and leucine residues with branched side chains that are intrinsically more rigid than the flexible methionine side chain. We propose that these differences in surface hydrophobicity, charge, and methionine content may be important in determining highly specific interactions of CaBP7 with target proteins, such as PI4KIIIβ.
- Published
- 2012
- Full Text
- View/download PDF
19. Factor H autoantibodies in membranoproliferative glomerulonephritis.
- Author
-
Goodship TH, Pappworth IY, Toth T, Denton M, Houlberg K, McCormick F, Warland D, Moore I, Hunze EM, Staniforth SJ, Hayes C, Cavalcante DP, Kavanagh D, Strain L, Herbert AP, Schmidt CQ, Barlow PN, Harris CL, and Marchbank KJ
- Subjects
- Adolescent, Adult, Aged, Binding Sites, Antibody, Complement C3, Complement C3 Nephritic Factor analysis, Complement Factor H chemistry, Female, Glomerulonephritis, Membranoproliferative genetics, Humans, Male, Middle Aged, Young Adult, Autoantibodies blood, Autoantibodies immunology, Complement Factor H immunology, Glomerulonephritis, Membranoproliferative immunology
- Abstract
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., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
20. Structural and functional characterization of the product of disease-related factor H gene conversion.
- Author
-
Herbert AP, Kavanagh D, Johansson C, Morgan HP, Blaum BS, Hannan JP, Barlow PN, and Uhrín D
- Subjects
- Complement C3b chemistry, Complement C3d chemistry, Complement Factor H metabolism, Crystallography, X-Ray, Humans, Mutation, Pichia genetics, Pichia metabolism, Protein Conformation, Protein Structure, Tertiary, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Structure-Activity Relationship, Temperature, Complement Factor H chemistry, Complement Factor H genetics, Gene Conversion
- Abstract
Numerous complement factor H (FH) mutations predispose patients to atypical hemolytic uremic syndrome (aHUS) and other disorders arising from inadequately regulated complement activation. No unifying structural or mechanistic consequences have been ascribed to these mutants beyond impaired self-cell protection. The S1191L and V1197A mutations toward the C-terminus of FH, which occur in patients singly or together, arose from gene conversion between CFH encoding FH and CFHR1 encoding FH-related 1. We show that neither single nor double mutations structurally perturbed recombinant proteins consisting of the FH C-terminal modules, 19 and 20 (FH19-20), although all three FH19-20 mutants were poor, compared to wild-type FH19-20, at promoting hemolysis of C3b-coated erythrocytes through competition with full-length FH. Indeed, our new crystal structure of the S1191L mutant of FH19-20 complexed with an activation-specific complement fragment, C3d, was nearly identical to that of the wild-type FH19-20:C3d complex, consistent with mutants binding to C3b with wild-type-like affinity. The S1191L mutation enhanced thermal stability of module 20, whereas the V1197A mutation dramatically decreased it. Thus, although mutant proteins were folded at 37 °C, they differ in conformational rigidity. Neither single substitutions nor double substitutions increased measurably the extent of FH19-20 self-association, nor did these mutations significantly affect the affinity of FH19-20 for three glycosaminoglycans, despite critical roles of module 20 in recognizing polyanionic self-surface markers. Unexpectedly, FH19-20 mutants containing Leu1191 self-associated on a heparin-coated surface to a higher degree than on surfaces coated with dermatan or chondroitin sulfates. Thus, potentially disease-related functional distinctions between mutants, and between FH and FH-related 1, may manifest in the presence of specific glycosaminoglycans.
- Published
- 2012
- Full Text
- View/download PDF
21. Structural analysis of the C-terminal region (modules 18-20) of complement regulator factor H (FH).
- Author
-
Morgan HP, Mertens HD, Guariento M, Schmidt CQ, Soares DC, Svergun DI, Herbert AP, Barlow PN, and Hannan JP
- Subjects
- Complement Factor H genetics, Complement Factor H metabolism, Crystallography, X-Ray, Humans, Protein Structure, Secondary, Scattering, Small Angle, Complement Factor H chemistry
- Abstract
Factor H (FH) is a soluble regulator of the human complement system affording protection to host tissues. It selectively inhibits amplification of C3b, the activation-specific fragment of the abundant complement component C3, in fluid phase and on self-surfaces and accelerates the decay of the alternative pathway C3 convertase, C3bBb. We have determined the crystal structure of the three carboxyl-terminal complement control protein (CCP) modules of FH (FH18-20) that bind to C3b, and which additionally recognize polyanionic markers specific to self-surfaces. These CCPs harbour nearly 30 disease-linked missense mutations. We have also deployed small-angle X-ray scattering (SAXS) to investigate FH18-20 flexibility in solution using FH18-20 and FH19-20 constructs. In the crystal lattice FH18-20 adopts a "J"-shape: A ~122-degree tilt between the structurally highly similar modules 18 and 19 precedes an extended, linear arrangement of modules 19 and 20 as observed in previously determined structures of these two modules alone. However, under solution conditions FH18-20 adopts multiple conformations mediated by flexibility between CCPs 18 and 19. We also pinpoint the locations of disease-associated missense mutations on the module 18 surface and discuss our data in the context of the C3b:FH interaction.
- Published
- 2012
- Full Text
- View/download PDF
22. NMR structure of Hsp12, a protein induced by and required for dietary restriction-induced lifespan extension in yeast.
- Author
-
Herbert AP, Riesen M, Bloxam L, Kosmidou E, Wareing BM, Johnson JR, Phelan MM, Pennington SR, Lian LY, and Morgan A
- Subjects
- Gene Expression Regulation, Fungal, Models, Molecular, Phenotype, Protein Structure, Secondary, Proteomics, Saccharomyces cerevisiae cytology, Saccharomyces cerevisiae physiology, Time Factors, Glucose metabolism, Heat-Shock Proteins chemistry, Heat-Shock Proteins metabolism, Nuclear Magnetic Resonance, Biomolecular, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Dietary restriction (DR) extends lifespan in yeast, worms, flies and mammals, suggesting that it may act via conserved processes. However, the downstream mechanisms by which DR increases lifespan remain unclear. We used a gel based proteomic strategy to identify proteins whose expression was induced by DR in yeast and thus may correlate with longevity. One protein up-regulated by DR was Hsp12, a small heat shock protein induced by various manipulations known to retard ageing. Lifespan extension by growth on 0.5% glucose (DR) was abolished in an hsp12Δ strain, indicating that Hsp12 is essential for the longevity effect of DR. In contrast, deletion of HSP12 had no effect on growth under DR conditions or a variety of environmental stresses, indicating that the effect of Hsp12 on lifespan is not due to increased general stress resistance. Unlike other small heat shock proteins, recombinant Hsp12 displayed negligible in vitro molecular chaperone activity, suggesting that its cellular function does not involve preventing protein aggregation. NMR analysis indicated that Hsp12 is monomeric and intrinsically unfolded in solution, but switches to a 4-helical conformation upon binding to membrane-mimetic SDS micelles. The structure of micelle-bound Hsp12 reported here is consistent with its recently proposed function as a membrane-stabilising 'lipid chaperone'. Taken together, our data suggest that DR-induced Hsp12 expression contributes to lifespan extension, possibly via membrane alterations.
- Published
- 2012
- Full Text
- View/download PDF
23. Crystallographic determination of the disease-associated T1184R variant of complement regulator factor H.
- Author
-
Morgan HP, Jiang J, Herbert AP, Kavanagh D, Uhrin D, Barlow PN, and Hannan JP
- Subjects
- Atypical Hemolytic Uremic Syndrome, Complement Factor H genetics, Crystallography, X-Ray, Models, Molecular, Protein Structure, Tertiary, Complement Factor H chemistry, Hemolytic-Uremic Syndrome genetics, Mutation
- Abstract
The soluble 155 kDa glycoprotein factor H (FH) protects host tissue from damage by the human complement system. It accelerates decay of the alternative-pathway C3 convertase, C3bBb, and is a cofactor for factor I-mediated cleavage of the opsonin C3b. Numerous mutations and single-nucleotide polymorphisms (SNPs) occur in the gene encoding FH and the resulting missense mutations and truncation products result in altered functionality that predisposes to the development of the serious renal condition atypical haemolytic uraemic syndrome (aHUS). Other polymorphisms are linked to membranoproliferative glomerulonephritis and macular degeneration. The two C-terminal modules of FH (FH19-20) harbour numerous aHUS-associated mutations that disrupt the ability of factor H to protect host cells from complement-mediated damage. In this work, the crystal structure of an aHUS-associated T1184R variant of FH19-20 at a resolution of 1.52 Å is described. It is shown that this mutation has negligible structural effects but causes a significant change in the electrostatic surface of these two domains. Mechanisms are discussed by which this mutation may alter FH-ligand interactions, particularly with regard to the extension of a region of this molecule within module 20 that has been associated with the binding of glycosaminoglycans (GAGs) or sialic acid residues.
- Published
- 2011
- Full Text
- View/download PDF
24. Structural basis for engagement by complement factor H of C3b on a self surface.
- Author
-
Morgan HP, Schmidt CQ, Guariento M, Blaum BS, Gillespie D, Herbert AP, Kavanagh D, Mertens HD, Svergun DI, Johansson CM, Uhrín D, Barlow PN, and Hannan JP
- Subjects
- Binding Sites, Complement C3b genetics, Complement C3b metabolism, Complement Factor H genetics, Complement Factor H metabolism, Crystallography, X-Ray, Models, Molecular, Mutagenesis, Nuclear Magnetic Resonance, Biomolecular, Protein Conformation, Complement C3b chemistry, Complement Factor H chemistry
- Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
25. Effects of buprenorphine, meloxicam, and flunixin meglumine as postoperative analgesia in mice.
- Author
-
Tubbs JT, Kissling GE, Travlos GS, Goulding DR, Clark JA, King-Herbert AP, and Blankenship-Paris TL
- Subjects
- Animals, Behavior, Animal, Clonixin therapeutic use, Drinking, Eating, Male, Meloxicam, Mice, Inbred C57BL, Models, Animal, Pain, Postoperative drug therapy, Postoperative Period, Analgesics, Opioid therapeutic use, Anti-Inflammatory Agents, Non-Steroidal therapeutic use, Buprenorphine therapeutic use, Clonixin analogs & derivatives, Hepatectomy veterinary, Mice, Pain, Postoperative veterinary, Thiazines therapeutic use, Thiazoles therapeutic use
- Abstract
C57BL/6NCrl male mice (n = 60; age, 6 to 7 wk) underwent partial hepatectomy or no surgery and were given 1 of 3 analgesics pre- and postoperatively. Food and water consumption, body weight, running wheel activity, locomotor activity, and serum corticosterone concentrations were measured before and after surgery. Mice that were surgically manipulated weighed significantly less on days 1 through 3 after surgery than did mice not manipulated surgically. On the day of surgery, the surgery groups consumed significantly less feed (-1.5±0.35 g) than did nonsurgery groups. There were no differences in water consumption on any day between surgery and nonsurgery groups or among the 3 analgesic groups. For running wheel activity, significant decreases in the surgery groups were seen at day 1 after surgery compared with baseline. Surgery groups that received buprenorphine and meloxicam returned to baseline activity levels on day 2 after surgery. Open-field testing revealed no significant differences in locomotor activity in any groups; however, posttreatment locomotor activity in the buprenorphine nonsurgery group was increased compared with baseline, and posttreatment locomotor activity in the flunixin meglumine surgery group was decreased compared with baseline. Serum corticosterone concentrations were within normal limits regardless of treatment in all groups. Comparison of the overall results indicated that meloxicam and buprenorphine, at the dose given, appear to be suitable postoperative analgesics for partial hepatectomy in mice. Flunixin meglumine at the given dosage (2.5 mg/kg) may not provide adequate analgesia for partial hepatectomy.
- Published
- 2011
26. Analgesic effects of tramadol, carprofen or multimodal analgesia in rats undergoing ventral laparotomy.
- Author
-
Zegre Cannon C, Kissling GE, Goulding DR, King-Herbert AP, and Blankenship-Paris T
- Subjects
- Analgesics, Opioid administration & dosage, Animals, Drug Therapy, Combination, Male, Models, Animal, Rats, Rats, Sprague-Dawley, Analgesia methods, Analgesics administration & dosage, Carbazoles administration & dosage, Laparotomy, Tramadol administration & dosage
- Abstract
In this study, the authors evaluated the analgesic efficacy of tramadol (an opioid-like analgesic), carprofen (a nonsteroidal anti-inflammatory drug) and a combination of both drugs (multimodal therapy) in a rat laparotomy model. The authors randomly assigned rats to undergo either surgery (abdominal laparotomy with visceral manipulation and anesthesia) or anesthesia only. Rats in each group were treated with tramadol (12.5 mg per kg body weight), carprofen (5 mg per kg body weight), a combination of tramadol and carprofen (12.5 mg per kg body weight and 5 mg per kg body weight, respectively) or saline (anesthesia control group only; 5 mg per kg body weight). The authors administered analgesia 10 min before anesthesia, 4 h after surgery or (for the rats that received anesthesia only) anesthesia and 24 h after surgery or anesthesia. They measured locomotor activity, running wheel activity, feed and water consumption, body weight and fecal corticosterone concentration of each animal before and after surgery. Clinical observations were made after surgery or anesthesia to evaluate signs of pain and distress. The authors found that carprofen, tramadol and a combination of carprofen and tramadol were all acceptable analgesia regimens for a rat laparotomy model.
- Published
- 2011
- Full Text
- View/download PDF
27. Evaluation of dosages and routes of administration of tramadol analgesia in rats using hot-plate and tail-flick tests.
- Author
-
Cannon CZ, Kissling GE, Hoenerhoff MJ, King-Herbert AP, and Blankenship-Paris T
- Subjects
- Administration, Oral, Analgesics, Opioid administration & dosage, Animals, Injections, Intraperitoneal veterinary, Injections, Subcutaneous veterinary, Male, Pain Measurement veterinary, Rats, Specific Pathogen-Free Organisms, Tramadol administration & dosage, Analgesics, Opioid therapeutic use, Pain drug therapy, Rats, Sprague-Dawley metabolism, Tramadol therapeutic use
- Abstract
Tramadol is an opioid-like analgesic with relatively mild side effects. Because it is inexpensive and is not classified as a controlled substance by the US federal government, the authors wanted to evaluate its applicability as a practical and effective analgesic in male Sprague Dawley rats. They measured the efficacy of four dosages (4, 12.5, 25 or 50 mg tramadol per kg body weight) and three routes of administration (per os (p.o.) in a flavored gelatin cube, subcutaneous (s.c.) or intraperitoneal (i.p.)) using the hot-plate test and the tail-flick test, which were carried out 1 week apart. Rats that were dosed p.o. were given flavored gelatin cubes without tramadol on the 2 d before testing to help them become acclimated to the gelatin, in an effort to increase the likelihood that they would consume the gelatin on the testing day. Results from the hot-plate and tail-flick tests for rats that were given tramadol p.o. were similar before and after administration, regardless of tramadol dosage, suggesting that this route of administration was not effective. The s.c. route of administration was effective at dosages of 25 mg and 50 mg tramadol per kg body weight, although these dosages also resulted in sedation and skin lesions. The i.p. route of administration was also effective at dosages of 12.5 mg, 25 mg and 50 mg tramadol per kg body weight, though sedation was observed at dosages of 25 mg and 50 mg per kg body weight. Intraperitoneal administration of 12.5 mg tramadol per kg body weight had no notable side effects, and the authors plan to further study this dosage and route of administration in a rodent surgical model of pain.
- Published
- 2010
- Full Text
- View/download PDF
28. Complement factor h autoantibodies and age-related macular degeneration.
- Author
-
Dhillon B, Wright AF, Tufail A, Pappworth I, Hayward C, Moore I, Strain L, Kavanagh D, Barlow PN, Herbert AP, Schmidt CQ, Armbrecht AM, Laude A, Deary IJ, Staniforth SJ, Holmes LV, Goodship TH, and Marchbank KJ
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Blood Donors, Blood Proteins genetics, Case-Control Studies, Complement C3b Inactivator Proteins genetics, Complement Factor H immunology, DNA Primers chemistry, DNA Probes chemistry, Enzyme-Linked Immunosorbent Assay, Female, Gene Dosage, Gene Frequency, Humans, Male, Middle Aged, Nucleic Acid Amplification Techniques, Polymerase Chain Reaction, Young Adult, Autoantibodies blood, Macular Degeneration immunology
- Abstract
Purpose: In this case-control study, the hypothesis that factor H autoantibodies are associated with age-related macular degeneration (AMD) was examined., Methods: One hundred AMD patients (median age, 78 years), 98 age-matched control subjects (median age, 78 years) known not to have AMD, and 100 healthy blood donors (median age, 43 years) were enrolled. An enzyme-linked immunosorbent assay (ELISA) was used to screen for complement factor H autoantibodies and either quantitative polymerase chain reaction (qPCR) or multiplex ligation-dependent probe amplification (MLPA) were performed to measure the copy number of the gene encoding complement factor H-related protein 3 (CFHR3)., Results: There was a significant difference in the median complement factor H autoantibody titer between the three groups (AMD patients, 196 reference units [RU]]; age-match control subjects, 316 RU; and blood donor control subjects, 121 RU; Kruskal-Wallis test, P < 0.001). Pair-wise comparison (Mann-Whitney test) showed that all three groups were significantly different from each other. Two different thresholds were used in the healthy blood donors to identify individuals with complement factor H autoantibodies. Both suggested that the prevalence of factor H autoantibodies was decreased in AMD patients. The CFHR3 copy number was measured as a surrogate for the deletion of the genes encoding complement factor H-related proteins 3 and 1 (CFHR3/1). The allele frequency of the deletion was significantly higher in the age-matched control subjects than in the AMD patients (22.2% vs. 8.2%)., Conclusions: The level of factor H autoantibodies is lower in AMD patients than in age-matched control subjects.
- Published
- 2010
- Full Text
- View/download PDF
29. Lysine and arginine side chains in glycosaminoglycan-protein complexes investigated by NMR, cross-linking, and mass spectrometry: a case study of the factor H-heparin interaction.
- Author
-
Blaum BS, Deakin JA, Johansson CM, Herbert AP, Barlow PN, Lyon M, and Uhrín D
- Subjects
- Amino Acid Sequence, Heparin pharmacology, Humans, Magnetic Resonance Spectroscopy, Models, Molecular, Molecular Sequence Data, Protein Binding, Protein Multimerization drug effects, Protein Structure, Quaternary, Solutions, Trypsin metabolism, Arginine chemistry, Complement Factor H chemistry, Complement Factor H metabolism, Cross-Linking Reagents pharmacology, Heparin metabolism, Lysine chemistry, Mass Spectrometry
- Abstract
We have used the interaction between module 7 of complement factor H (CFH approximately 7) and a fully sulfated heparin tetrasaccharide to exemplify a new approach for studying contributions of basic side chains to the formation of glycosaminoglycan (GAG)-protein complexes. We first employed HISQC and H(2)CN NMR experiments to monitor the side-chain resonances of lysines and arginines in (15)N, (13)C-labeled protein during titrations with a fully sulfated heparin tetrasaccharide under physiological conditions. Under identical conditions and using (15)N-labeled protein, we then cross-linked tetrasaccharide to CFH approximately 7 and confirmed the 1:1 stoichiometry by FT-ICR-MS. We subsequently characterized this covalent protein-GAG conjugate by NMR and further MS techniques. MALDI-TOF MS identified protein fragments obtained via trypsin digestion or chemical fragmentation, yielding information concerning the site of GAG attachment. Combining MS and NMR data allowed us to identify the side chain of K405 as the point of attachment of the cross-linked heparin oligosaccharide to CFH approximately 7. On the basis of the analysis of NMR and MS data of the noncovalent and cross-linked CFH approximately 7-tetrasaccharide complexes, we conclude that the K446 side chain is not essential for binding the tetrasaccharide, despite the large chemical shift perturbations of its backbone amide (15)N and (1)H resonances during titrations. We show that R444 provides the most important charge-charge interaction within a C-terminal heparin-binding subsite of CFH approximately 7 whereas side chains of R404, K405, and K388 are the predominant contributors to an N-terminal binding subsite located in the immediate vicinity of residue 402, which is implicated in age-related macular degeneration (AMD).
- Published
- 2010
- Full Text
- View/download PDF
30. The structure of the KlcA and ArdB proteins reveals a novel fold and antirestriction activity against Type I DNA restriction systems in vivo but not in vitro.
- Author
-
Serfiotis-Mitsa D, Herbert AP, Roberts GA, Soares DC, White JH, Blakely GW, Uhrín D, and Dryden DT
- Subjects
- Amino Acid Sequence, Bacterial Proteins genetics, Bacterial Proteins metabolism, Bordetella pertussis chemistry, DNA Restriction Enzymes metabolism, Deoxyribonucleases, Type I Site-Specific metabolism, Endopeptidase Clp metabolism, Escherichia coli genetics, Escherichia coli Proteins chemistry, Escherichia coli Proteins metabolism, Hydrophobic and Hydrophilic Interactions, Models, Molecular, Molecular Sequence Data, Nuclear Magnetic Resonance, Biomolecular, Protein Folding, Sequence Homology, Amino Acid, Site-Specific DNA-Methyltransferase (Adenine-Specific) metabolism, Bacterial Proteins chemistry, Deoxyribonucleases, Type I Site-Specific antagonists & inhibitors
- Abstract
Plasmids, conjugative transposons and phage frequently encode anti-restriction proteins to enhance their chances of entering a new bacterial host that is highly likely to contain a Type I DNA restriction and modification (RM) system. The RM system usually destroys the invading DNA. Some of the anti-restriction proteins are DNA mimics and bind to the RM enzyme to prevent it binding to DNA. In this article, we characterize ArdB anti-restriction proteins and their close homologues, the KlcA proteins from a range of mobile genetic elements; including an ArdB encoded on a pathogenicity island from uropathogenic Escherichia coli and a KlcA from an IncP-1b plasmid, pBP136 isolated from Bordetella pertussis. We show that all the ArdB and KlcA act as anti-restriction proteins and inhibit the four main families of Type I RM systems in vivo, but fail to block the restriction endonuclease activity of the archetypal Type I RM enzyme, EcoKI, in vitro indicating that the action of ArdB is indirect and very different from that of the DNA mimics. We also present the structure determined by NMR spectroscopy of the pBP136 KlcA protein. The structure shows a novel protein fold and it is clearly not a DNA structural mimic.
- Published
- 2010
- Full Text
- View/download PDF
31. Annexin-II, DNA, and histones serve as factor H ligands on the surface of apoptotic cells.
- Author
-
Leffler J, Herbert AP, Norström E, Schmidt CQ, Barlow PN, Blom AM, and Martin M
- Subjects
- Binding Sites, Complement C4b-Binding Protein metabolism, Flow Cytometry, Glycosaminoglycans metabolism, Humans, Immunoprecipitation, Jurkat Cells, Ligands, Microscopy, Confocal, N-Acetylneuraminic Acid metabolism, Necrosis, Phospholipases A2 metabolism, Phospholipases A2 pharmacology, Protein Binding drug effects, Surface Plasmon Resonance, Annexin A2 metabolism, Apoptosis, Cell Membrane metabolism, Complement Factor H metabolism, DNA, Neoplasm metabolism, Histones metabolism
- Abstract
Apoptotic cells are opsonized by complement components such as C1q and C3b, which increases their susceptibility to phagocytosis. Soluble complement inhibitors such as factor H (fH) also recognize apoptotic cells to minimize the pro-inflammatory effects of downstream complement activation. We used four radiolabeled protein constructs that span different regions of the 20 complement control protein (CCP) modules that make up fH and found that fragments comprising CCPs 6-8, CCPs 8-15, and CCPs 19-20 but not CCPs 1-4, bound to apoptotic Jurkat T cells. There are four possible ligand types on apoptotic cells that could recruit fH: proteins, carbohydrates, lipids, and DNA. We found that CCPs 6-8 of fH bind to annexin-II, a trypsin-insensitive protein that becomes exposed on surfaces of apoptotic cells. The second ligand of fH, which interacts with CCPs 6-8 and 19-20, is DNA. Confocal microscopy showed co-localization of fH with antibodies specific for DNA. fH also binds to histones devoid of DNA, and CCPs 1-4, 6-8, and 8-15 mediate this interaction. Treatment of apoptotic cells with neuraminidase, chondroitinase, heparitinase, and heparinase did not change fH binding. Treatment of apoptotic cells with phospholipase A(2) dramatically increased both binding of fH and cell-surface DNA. We also excluded the possibility that fH interacts with lysophospholipids using surface plasmon resonance and flow cytometry with lipid-coated beads. Identification of annexin-II as one of the fH ligands on apoptotic cells together with the fact that autoantibodies against annexin-II are found in systemic lupus erythematosus provides further insight into understanding the pathogenesis of this disease.
- Published
- 2010
- Full Text
- View/download PDF
32. 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.
- Author
-
Moore I, Strain L, Pappworth I, Kavanagh D, Barlow PN, Herbert AP, Schmidt CQ, Staniforth SJ, Holmes LV, Ward R, Morgan L, Goodship TH, and Marchbank KJ
- Subjects
- Apolipoproteins immunology, Apolipoproteins metabolism, Autoantibodies immunology, Blood Proteins immunology, Blood Proteins metabolism, Child, Child, Preschool, Cohort Studies, Complement C3 immunology, Complement C3 metabolism, Complement C3b Inactivator Proteins immunology, Complement C3b Inactivator Proteins metabolism, Complement Factor H immunology, Complement Factor H metabolism, Complement Factor I immunology, Complement Factor I metabolism, Female, Gene Dosage, Hemolytic-Uremic Syndrome immunology, Humans, Infant, Male, Membrane Cofactor Protein immunology, Membrane Cofactor Protein metabolism, Sequence Deletion, Apolipoproteins genetics, Autoantibodies blood, Blood Proteins genetics, Complement C3 genetics, Complement C3b Inactivator Proteins genetics, Complement Factor H genetics, Complement Factor I genetics, Hemolytic-Uremic Syndrome blood, Hemolytic-Uremic Syndrome genetics, Membrane Cofactor Protein genetics
- Abstract
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.
- Published
- 2010
- Full Text
- View/download PDF
33. The central portion of factor H (modules 10-15) is compact and contains a structurally deviant CCP module.
- Author
-
Schmidt CQ, Herbert AP, Mertens HD, Guariento M, Soares DC, Uhrin D, Rowe AJ, Svergun DI, and Barlow PN
- Subjects
- Amino Acid Sequence, Chromatography, Gel, Humans, Magnetic Resonance Spectroscopy, Models, Molecular, Molecular Sequence Data, Pliability, Protein Structure, Secondary, Scattering, Small Angle, Solutions, Ultracentrifugation, X-Ray Diffraction, Complement Factor H chemistry
- Abstract
The first eight and the last two of 20 complement control protein (CCP) modules within complement factor H (fH) encompass binding sites for C3b and polyanionic carbohydrates. These binding sites cooperate self-surface selectively to prevent C3b amplification, thus minimising complement-mediated damage to host. Intervening fH CCPs, apparently devoid of such recognition sites, are proposed to play a structural role. One suggestion is that the generally small CCPs 10-15, connected by longer-than-average linkers, act as a flexible tether between the two functional ends of fH; another is that the long linkers induce a 180 degrees bend in the middle of fH. To test these hypotheses, we determined the NMR-derived structure of fH12-13 consisting of module 12, shown here to have an archetypal CCP structure, and module 13, which is uniquely short and features a laterally protruding helix-like insertion that contributes to a prominent electropositive patch. The unusually long fH12-13 linker is not flexible. It packs between the two CCPs that are not folded back on each other but form a shallow vee shape; analytical ultracentrifugation and X-ray scattering supported this finding. These two techniques additionally indicate that flanking modules (within fH11-14 and fH10-15) are at least as rigid and tilted relative to neighbours as are CCPs 12 and 13 with respect to one another. Tilts between successive modules are not unidirectional; their principal axes trace a zigzag path. In one of two arrangements for CCPs 10-15 that fit well with scattering data, CCP 14 is folded back onto CCP 13. In conclusion, fH10-15 forms neither a flexible tether nor a smooth bend. Rather, it is compact and has embedded within it a CCP module (CCP 13) that appears to be highly specialised given both its deviant structure and its striking surface charge distribution. A passive, purely structural role for this central portion of fH is unlikely.
- Published
- 2010
- Full Text
- View/download PDF
34. Commentary: update on animal models for NTP studies.
- Author
-
King-Herbert AP, Sills RC, and Bucher JR
- Subjects
- Animals, Mice, Rats, Rats, Inbred F344, Rats, Sprague-Dawley, Carcinogenicity Tests methods, Disease Models, Animal
- Abstract
Based on recommendations of participants at the National Toxicology Program (NTP) workshop "Animal Models for the NTP Cancer Bioassay: Strains and Stocks-Should We Switch?" some modifications to the NTP rodent cancer bioassay were made. The B6C3F1 remains the mouse model used in the cancer bioassay. The use of multiple strains of mice will be explored through a new branch within the NTP, the Host Susceptibility Branch. Several rat models were evaluated; the Harlan Sprague Dawley rat is currently being used in the NTP studies.
- Published
- 2010
- Full Text
- View/download PDF
35. A molecular insight into complement evasion by the staphylococcal complement inhibitor protein family.
- Author
-
Ricklin D, Tzekou A, Garcia BL, Hammel M, McWhorter WJ, Sfyroera G, Wu YQ, Holers VM, Herbert AP, Barlow PN, Geisbrecht BV, and Lambris JD
- Subjects
- Complement C3 Convertase, Alternative Pathway metabolism, Complement C3 Convertase, Alternative Pathway physiology, Complement C3b chemistry, Complement Factor B metabolism, Complement Factor H metabolism, Complement Inactivator Proteins metabolism, Humans, Protein Binding immunology, Staphylococcus aureus pathogenicity, Virulence, Complement C3b metabolism, Complement Inactivator Proteins physiology, Multigene Family immunology, Staphylococcus aureus immunology
- Abstract
Staphylococcus aureus possesses an impressive arsenal of complement evasion proteins that help the bacterium escape attack of the immune system. The staphylococcal complement inhibitor (SCIN) protein exhibits a particularly high potency and was previously shown to block complement by acting at the level of the C3 convertases. However, many details about the exact binding and inhibitory mechanism remained unclear. In this study, we demonstrate that SCIN directly binds with nanomolar affinity to a functionally important area of C3b that lies near the C terminus of its beta-chain. Direct competition of SCIN with factor B for C3b slightly decreased the formation of surface-bound convertase. However, the main inhibitory effect can be attributed to an entrapment of the assembled convertase in an inactive state. Whereas native C3 is still able to bind to the blocked convertase, no generation and deposition of C3b could be detected in the presence of SCIN. Furthermore, SCIN strongly competes with the binding of factor H to C3b and influences its regulatory activities: the SCIN-stabilized convertase was essentially insensitive to decay acceleration by factor H and the factor I- and H-mediated conversion of surface-bound C3b to iC3b was significantly reduced. By targeting a key area on C3b, SCIN is able to block several essential functions within the alternative pathway, which explains the high potency of the inhibitor. Our findings provide an important insight into complement evasion strategies by S. aureus and may act as a base for further functional studies.
- Published
- 2009
- Full Text
- View/download PDF
36. 1H, 15N and 13C resonance assignment of the pair of Factor-I like modules of the complement protein C7.
- Author
-
Phelan MM, Thai CT, Herbert AP, Bella J, Uhrín D, Ogata RT, Barlow PN, and Bramham J
- Subjects
- Amino Acid Sequence, Carbon Isotopes chemistry, Molecular Sequence Data, Nitrogen Isotopes chemistry, Protein Conformation, Protein Structure, Tertiary, Protons, Complement C7 chemistry, Complement C7 ultrastructure, Fibrinogen chemistry, Fibrinogen ultrastructure, Magnetic Resonance Spectroscopy methods
- Abstract
The carboxy terminus of human complement component C7 comprises two Factor I-like Modules (FIMs) which are essential for formation of the Membrane Attack Complex, the terminal pathway of the innate immune system. C7-FIMs is a 16.9 kDa, recombinant, disulphide-rich, protein encompassing this C-terminal domain. Using conventional triple resonance experiments 93% of the (1)H, (15)N and (13)C assignment has been achieved, accounting for all assignment apart from a flexible N-terminus cloning artefact and an undefined loop. The chemical shifts have been deposited in the BioMagResBank; Accession No. 15996.
- Published
- 2009
- Full Text
- View/download PDF
37. The binding of factor H to a complex of physiological polyanions and C3b on cells is impaired in atypical hemolytic uremic syndrome.
- Author
-
Ferreira VP, Herbert AP, Cortés C, McKee KA, Blaum BS, Esswein ST, Uhrín D, Barlow PN, Pangburn MK, and Kavanagh D
- Subjects
- Animals, Cells, Cultured, Complement Factor H genetics, Erythrocytes pathology, Genetic Predisposition to Disease, Hemolytic-Uremic Syndrome immunology, Hemolytic-Uremic Syndrome pathology, Humans, Polyelectrolytes, Polymers metabolism, Protein Binding genetics, Sheep, Complement C3b metabolism, Complement Factor H metabolism, Erythrocytes immunology, Hemolytic-Uremic Syndrome genetics, Heparin metabolism, Mutation
- Abstract
Factor H (fH) is essential for complement homeostasis in fluid-phase and on surfaces. Its two C-terminal domains (CCP 19-20) anchor fH to self-surfaces where it prevents C3b amplification in a process requiring its N-terminal four domains. In atypical hemolytic uremic syndrome (aHUS), mutations clustering toward the C terminus of fH may disrupt interactions with surface-associated C3b or polyanions and thereby diminish the ability of fH to regulate complement. To test this, we compared a recombinant protein encompassing CCP 19-20 with 16 mutants. The mutations had only very limited and localized effects on protein structure. Although we found four aHUS-linked fH mutations that decreased binding to C3b and/or to heparin (a model compound for cell surface polyanionic carbohydrates), we identified five aHUS-associated mutants with increased affinity for either or both ligands. Strikingly, these variable affinities for the individual ligands did not correlate with the extent to which all the aHUS-associated mutants were found to be impaired in a more physiological assay that measured their ability to inhibit cell surface complement functions of full-length fH. Taken together, our data suggest that disruption of a complex fH-self-surface recognition process, involving a balance of affinities for protein and physiological carbohydrate ligands, predisposes to aHUS.
- Published
- 2009
- Full Text
- View/download PDF
38. Molecular basis of the interaction between complement receptor type 2 (CR2/CD21) and Epstein-Barr virus glycoprotein gp350.
- Author
-
Young KA, Herbert AP, Barlow PN, Holers VM, and Hannan JP
- Subjects
- Amino Acid Substitution, Binding Sites, Cell Line, Tumor, Humans, Models, Molecular, Mutagenesis, Site-Directed, Mutant Proteins genetics, Mutant Proteins metabolism, Mutation, Missense, Point Mutation, Protein Binding, Protein Interaction Domains and Motifs, Protein Interaction Mapping, Protein Structure, Tertiary, Receptors, Complement 3d genetics, Viral Matrix Proteins genetics, Herpesvirus 4, Human physiology, Receptors, Complement 3d metabolism, Viral Matrix Proteins metabolism, Virus Attachment
- Abstract
The binding of the Epstein-Barr virus glycoprotein gp350 by complement receptor type 2 (CR2) is critical for viral attachment to B lymphocytes. We set out to test hypotheses regarding the molecular nature of this interaction by developing an enzyme-linked immunosorbent assay (ELISA) for the efficient analysis of the gp350-CR2 interaction by utilizing wild-type and mutant forms of recombinant gp350 and also of the CR2 N-terminal domains SCR1 and SCR2 (designated CR2 SCR1-2). To delineate the CR2-binding site on gp350, we generated 17 gp350 single-site substitutions targeting an area of gp350 that has been broadly implicated in the binding of both CR2 and the major inhibitory anti-gp350 monoclonal antibody (MAb) 72A1. These site-directed mutations identified a novel negatively charged CR2-binding surface described by residues Glu-21, Asp-22, Glu-155, Asp-208, Glu-210, and Asp-296. We also identified gp350 amino acid residues involved in non-charge-dependent interactions with CR2, including Tyr-151, Ile-160, and Trp-162. These data were supported by experiments in which phycoerythrin-conjugated wild-type and mutant forms of gp350 were incubated with CR2-expressing K562 cells and binding was assessed by flow cytometry. The ELISA was further utilized to identify several positively charged residues (Arg-13, Arg-28, Arg-36, Lys-41, Lys-57, Lys-67, Arg-83, and Arg-89) within SCR1-2 of CR2 that are involved in the binding interaction with gp350. These experiments allowed a comparison of those CR2 residues that are important for binding gp350 to those that define the epitope for an effective inhibitory anti-CR2 MAb, 171 (Asn-11, Arg-13, Ser-32, Thr-34, Arg-36, and Tyr-64). The mutagenesis data were used to calculate a model of the CR2-gp350 complex using the soft-docking program HADDOCK.
- Published
- 2008
- Full Text
- View/download PDF
39. Structural basis and functional effects of the interaction between complement inhibitor C4b-binding protein and DNA.
- Author
-
Okroj M, Jenkins HT, Herbert AP, Barlow PN, and Blom AM
- Subjects
- Amino Acids metabolism, Binding, Competitive, Complement C3-C5 Convertases metabolism, Complement C3b metabolism, Complement C4b metabolism, Complement C4b-Binding Protein, Electrophoretic Mobility Shift Assay, Heparin metabolism, Humans, Models, Molecular, Protein Binding, Protein Structure, Secondary, Structure-Activity Relationship, DNA chemistry, DNA metabolism, Histocompatibility Antigens chemistry, Histocompatibility Antigens metabolism
- Abstract
Human C4b-binding protein (C4BP) is a soluble, multiple-subunit inhibitor of complement that circulates in blood. Recently C4BP was shown to bind DNA, reduce DNA release from necrotic cells and limit DNA-mediated complement activation in solution. Herein we employed nuclear magnetic resonance spectroscopy to measure chemical shift perturbations and used them to restrain the computational docking of a B-form 10-base-pair DNA molecule onto the solution structure of C4BP alpha-chain complement control protein (CCP) domains 1-2 (C4BP12). Six amino acid residues located on one face of the interdomain junction - Val(38), Ser(40), Thr(43), Tyr(62), Lys(63) and Arg(64) - exhibited the largest chemical shift changes. In the model, the DNA lies in a cleft formed by the interdomain interface. The double-helix is perpendicular to the long axis of C4BP12 consistent with the multiple arms of C4BP binding to adjacent sites on a longer DNA molecule. The DNA lies in a region previously shown to bind C4b and heparin and these molecules (but not C3b) inhibited the DNA-C4BP interaction. Nonetheless, crucial C4BP functions such as cofactor activity for factor I cleavage of C4b and C3b, and decay acceleration of the classical C3 convertase appeared not to be affected by the presence of DNA. Taken together these results reinforce the case for the occupation of some of the seven arms of C4BP in a multivalent interaction with DNA or surface bound glycosaminoglycans while other arms engage C4b or C3b.
- Published
- 2008
- Full Text
- View/download PDF
40. A new map of glycosaminoglycan and C3b binding sites on factor H.
- Author
-
Schmidt CQ, Herbert AP, Kavanagh D, Gandy C, Fenton CJ, Blaum BS, Lyon M, Uhrín D, and Barlow PN
- Subjects
- Binding Sites immunology, Chromatography, Affinity, Complement C3b chemistry, Complement Factor H chemistry, Complement Factor H genetics, Complement Pathway, Alternative immunology, Complement System Proteins chemistry, Complement System Proteins metabolism, Glycosaminoglycans biosynthesis, Glycosaminoglycans genetics, Heparin metabolism, Humans, Magnetic Resonance Spectroscopy, Peptide Fragments chemistry, Peptide Fragments metabolism, Peptide Mapping, Polyelectrolytes, Polymers metabolism, Protein Folding, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins genetics, Complement C3b metabolism, Complement Factor H metabolism, Glycosaminoglycans metabolism
- Abstract
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 (K(d)s of 14 and 3.5 microM, respectively) or a C1 chip (K(d)s of 10 and 4.5 microM, 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.
- Published
- 2008
- Full Text
- View/download PDF
41. Structure of the N-terminal region of complement factor H and conformational implications of disease-linked sequence variations.
- Author
-
Hocking HG, Herbert AP, Kavanagh D, Soares DC, Ferreira VP, Pangburn MK, Uhrín D, and Barlow PN
- Subjects
- Complement C3b genetics, Complement C3b metabolism, Complement Factor B genetics, Complement Factor B metabolism, Complement Factor H chemistry, Complement Factor H genetics, Complement Factor H metabolism, Humans, Nuclear Magnetic Resonance, Biomolecular, Protein Structure, Tertiary genetics, Amino Acid Substitution, Genetic Diseases, Inborn genetics, Genetic Diseases, Inborn metabolism, Kidney Diseases genetics, Kidney Diseases metabolism, Polymorphism, Single Nucleotide
- Abstract
Factor H is a regulatory glycoprotein of the complement system. We expressed the three N-terminal complement control protein modules of human factor H (FH1-3) and confirmed FH1-3 to be the minimal unit with cofactor activity for C3b proteolysis by factor I. We reconstructed FH1-3 from NMR-derived structures of FH1-2 and FH2-3 revealing an approximately 105-A-long rod-like arrangement of the modules. In structural comparisons with other C3b-engaging proteins, factor H module 3 most closely resembles factor B module 3, consistent with factor H competing with factor B for binding C3b. Factor H modules 1, 2, and 3 each has a similar backbone structure to first, second, and third modules, respectively, of functional sites in decay accelerating factor and complement receptor type 1; the equivalent intermodular tilt and twist angles are also broadly similar. Resemblance between molecular surfaces is closest for first modules but absent in the case of second modules. Substitution of buried Val-62 with Ile (a factor H single nucleotide polymorphism potentially protective for age-related macular degeneration and dense deposit disease) causes rearrangements within the module 1 core and increases thermal stability but does not disturb the interface with module 2. Replacement of partially exposed (in module 1) Arg-53 by His (an atypical hemolytic uremic syndrome-linked mutation) did not impair structural integrity at 37 degrees C, but this FH1-2 mutant was less stable at higher temperatures; furthermore, chemical shift differences indicated potential for small structural changes at the module 1-2 interface.
- Published
- 2008
- Full Text
- View/download PDF
42. Translational mini-review series on complement factor H: structural and functional correlations for factor H.
- Author
-
Schmidt CQ, Herbert AP, Hocking HG, Uhrín D, and Barlow PN
- Subjects
- Amino Acid Sequence, Binding Sites, Complement C3 immunology, Complement Factor H chemistry, Complement Factor H immunology, Humans, Models, Molecular, Molecular Sequence Data, Mutation, Protein Conformation, Sequence Alignment, Structure-Activity Relationship, Complement Factor H genetics
- Abstract
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.
- Published
- 2008
- Full Text
- View/download PDF
43. Structural basis for complement factor H linked age-related macular degeneration.
- Author
-
Prosser BE, Johnson S, Roversi P, Herbert AP, Blaum BS, Tyrrell J, Jowitt TA, Clark SJ, Tarelli E, Uhrín D, Barlow PN, Sim RB, Day AJ, and Lea SM
- Subjects
- Binding Sites, Complement Factor H genetics, Crystallography, X-Ray, Gene Products, gag chemistry, Gene Products, gag genetics, Gene Products, gag metabolism, Ligands, Models, Molecular, Mutation genetics, Polysaccharides chemistry, Polysaccharides metabolism, Protein Structure, Quaternary, Protein Structure, Tertiary, Sucrose analogs & derivatives, Sucrose chemistry, Sucrose metabolism, Surface Properties, Aging physiology, Complement Factor H chemistry, Complement Factor H metabolism
- Abstract
Nearly 50 million people worldwide suffer from age-related macular degeneration (AMD), which causes severe loss of central vision. A single-nucleotide polymorphism in the gene for the complement regulator factor H (FH), which causes a Tyr-to-His substitution at position 402, is linked to approximately 50% of attributable risks for AMD. We present the crystal structure of the region of FH containing the polymorphic amino acid His402 in complex with an analogue of the glycosaminoglycans (GAGs) that localize the complement regulator on the cell surface. The structure demonstrates direct coordination of ligand by the disease-associated polymorphic residue, providing a molecular explanation of the genetic observation. This glycan-binding site occupies the center of an extended interaction groove on the regulator's surface, implying multivalent binding of sulfated GAGs. This finding is confirmed by structure-based site-directed mutagenesis, nuclear magnetic resonance-monitored binding experiments performed for both H402 and Y402 variants with this and another model GAG, and analysis of an extended GAG-FH complex.
- Published
- 2007
- Full Text
- View/download PDF
44. Structure shows that a glycosaminoglycan and protein recognition site in factor H is perturbed by age-related macular degeneration-linked single nucleotide polymorphism.
- Author
-
Herbert AP, Deakin JA, Schmidt CQ, Blaum BS, Egan C, Ferreira VP, Pangburn MK, Lyon M, Uhrín D, and Barlow PN
- Subjects
- Aging genetics, Aging pathology, Anticoagulants metabolism, Anticoagulants pharmacology, Apoptosis physiology, Binding Sites physiology, Complement Factor H chemistry, Heparin metabolism, Heparin pharmacology, Histidine genetics, Humans, Macular Degeneration metabolism, Protein Structure, Quaternary, Protein Structure, Tertiary, Surface Plasmon Resonance, Tyrosine genetics, Complement Factor H genetics, Complement Factor H metabolism, Glycosaminoglycans metabolism, Macular Degeneration genetics, Macular Degeneration pathology, Polymorphism, Single Nucleotide
- Abstract
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 His(402) and Tyr(402) variants of CCP7, CCP7,8, and CCP6-8. We determined structures of His(402) and Tyr(402) CCP7 and showed them to be nearly identical. The side chains of His/Tyr(402) have similar, solvent-exposed orientations far from interfaces with CCP6 and -8. Tyr(402) CCP7 bound significantly more tightly than His(402) 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, Tyr(402) CCP6-8 binds significantly more tightly than His(402) 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.
- Published
- 2007
- Full Text
- View/download PDF
45. Critical role of the C-terminal domains of factor H in regulating complement activation at cell surfaces.
- Author
-
Ferreira VP, Herbert AP, Hocking HG, Barlow PN, and Pangburn MK
- Subjects
- Animals, Binding Sites, Binding, Competitive, Complement Factor H genetics, Erythrocytes immunology, Hemolysis immunology, Humans, Protein Structure, Tertiary genetics, Protein Structure, Tertiary physiology, Recombinant Proteins genetics, Recombinant Proteins metabolism, Complement Activation, Complement C3b metabolism, Complement Factor H metabolism, Erythrocyte Membrane immunology
- Abstract
The plasma protein factor H primarily controls the activation of the alternative pathway of complement. The C-terminal of factor H is known to be involved in protection of host cells from complement attack. In the present study, we show that domains 19-20 alone are capable of discriminating between host-like and complement-activating cells. Furthermore, although factor H possesses three binding sites for C3b, binding to cell-bound C3b can be almost completely inhibited by the single site located in domains 19-20. All of the regulatory activities of factor H are expressed by the N-terminal four domains, but these activities toward cell-bound C3b are inhibited by isolated recombinant domains 19-20 (rH 19-20). Direct competition with the N-terminal site is unlikely to explain this because regulation of fluid phase C3b is unaffected by domains 19-20. Finally, we show that addition of isolated rH 19-20 to normal human serum leads to aggressive complement-mediated lysis of normally nonactivating sheep erythrocytes and moderate lysis of human erythrocytes, which possess membrane-bound regulators of complement. Taken together, the results highlight the importance of the cell surface protective functions exhibited by factor H compared with other complement regulatory proteins. The results may also explain why atypical hemolytic uremic syndrome patients with mutations affecting domains 19-20 can maintain complement homeostasis in plasma while their complement system attacks erythrocytes, platelets, endothelial cells, and kidney tissue.
- Published
- 2006
- Full Text
- View/download PDF
46. Disease-associated sequence variations congregate in a polyanion recognition patch on human factor H revealed in three-dimensional structure.
- Author
-
Herbert AP, Uhrín D, Lyon M, Pangburn MK, and Barlow PN
- Subjects
- Amino Acid Sequence, Complement C3d chemistry, Heparin chemistry, Humans, Magnetic Resonance Spectroscopy methods, Molecular Sequence Data, Pichia metabolism, Polymorphism, Genetic, Protein Conformation, Saccharomyces cerevisiae metabolism, Sequence Homology, Amino Acid, Anions chemistry, Complement Factor H chemistry
- Abstract
Mutations and polymorphisms in the regulator of complement activation, factor H, have been linked to atypical hemolytic uremic syndrome (aHUS), membranoproliferative glomerulonephritis, and age-related macular degeneration. Many aHUS patients carry mutations in the two C-terminal modules of factor H, which normally confer upon this abundant 155-kDa plasma glycoprotein its ability to selectively bind self-surfaces and prevent them from inappropriately triggering the complement cascade via the alternative pathway. In the current study, the three-dimensional solution structure of the C-terminal module pair of factor H has been determined. A binding site for a fully sulfated heparin-derived tetrasaccharide has been delineated using chemical shift mapping and the C3d/C3b-binding site inferred from sequence comparisons and computational docking. The resultant information allows assessment of the likely consequences of aHUS-associated amino acid substitutions in this critical region of factor H. It is striking that, excepting those likely to perturb the three-dimensional structure, aHUS-associated missense mutations congregate in the polyanion-binding site delineated in this study, thus potentially disrupting a vital mechanism for control of complement on self-surfaces in the microvasculature of the kidney. It is intriguing that a single nucleotide polymorphism predisposing to age-related macular degeneration occupies another region of factor H that harbors a polyanion-binding site.
- Published
- 2006
- Full Text
- View/download PDF
47. Disease-associated sequence variations in factor H: a structural biology approach.
- Author
-
Herbert AP, Soares DC, Pangburn MK, and Barlow PN
- Subjects
- Aging physiology, Animals, Complement Activation, Hemolytic-Uremic Syndrome immunology, Hemolytic-Uremic Syndrome pathology, Humans, Macular Degeneration immunology, Macular Degeneration pathology, Models, Molecular, Molecular Sequence Data, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms metabolism, Amino Acid Sequence, Complement Factor H chemistry, Complement Factor H genetics, Complement Factor H metabolism, Complement Inactivating Agents chemistry, Complement Inactivating Agents metabolism, Polymorphism, Genetic, Protein Conformation
- Published
- 2006
- Full Text
- View/download PDF
48. Opportunities for new therapies based on the natural regulators of complement activation.
- Author
-
Brook E, Herbert AP, Jenkins HT, Soares DC, and Barlow PN
- Subjects
- Complement Pathway, Alternative drug effects, Complement Pathway, Alternative physiology, Complement Pathway, Classical drug effects, Complement Pathway, Classical physiology, Complement System Proteins genetics, Elapid Venoms pharmacology, Homeostasis, Humans, Kinetics, Models, Biological, Models, Molecular, Protein Conformation, Complement Activation physiology
- Abstract
While the complement system is an essential component of immunity, shutting down all or part of it could be beneficial in a wide range of clinical situations. Designer, small-molecule, protease inhibitors and antagonists of protein-protein interactions are under development, while an approach based on a humanized monoclonal antibody to the C5 component works effectively against the later stages of complement activation and is close to completing clinical trials. The cobra venom factor depletes plasma of essential complement components, and a humanized (nonimmunogenic) version is being sought. Perhaps the most promising approach to comprehensive complement downregulation, however, is the exploitation of innate regulators of complement activation, with two products in clinical trials. The potential for more efficacious complement blockers of this kind is growing because of better targeting, but a deeper knowledge at the atomic level of mechanisms of action of these regulators is needed to underpin a rational approach to design of still more potent complement inhibitors.
- Published
- 2005
- Full Text
- View/download PDF
49. Effects of immobilization restraint on Syrian golden hamsters.
- Author
-
King-Herbert AP, Hesterburg TW, Thevenaz PP, Hamm TE Jr, Moss OR, Janszen DB, and Everitt JI
- Subjects
- Administration, Inhalation, Animals, Body Temperature physiology, Body Weight physiology, Corticosterone blood, Cricetinae, Drinking physiology, Eating physiology, Hydrocortisone blood, Male, Mesocricetus blood, Immobilization physiology, Mesocricetus physiology, Toxicity Tests methods
- Abstract
Rodent nose-only inhalation toxicology systems comprise whole-body immobilization in plastic restraint tubes. This method of restraint is known to have a variety of effects on animals. In the studies reported here, two independent toxicology laboratories examined the effects of inhalation tube restraint in Syrian golden hamsters, a species that has recently gained importance in inhalation studies of fibrous particulates. Body weight, food and water consumption, core body temperature, and plasma cortisol and corticosterone concentrations were assessed in animals immobilized in nose-only inhalation tubes, and the results were compared with those from unrestrained cage-control animals. Animals were immobilized for either 6 h/ day, 5 days/week for 13 weeks (subchronic), or 4 h/day for 14 consecutive days (subacute), mimicking exposure conditions commonly used in nose-only inhalation studies. Tube restraint was found to induce a marked decrease in body weight, which increased in response to cessation of restraint. The body weight decrement was associated with significant differences in food and water consumption between the restrained and control groups in the subacute study and only food consumption in the subchronic study. During the restraint period, core body temperature in the immobilized animals increased slightly but not above the normal range for this species. Plasma cortisol and corticosterone concentrations were not significantly increased with use of restraint, compared with values in controls. Immobilization-associated body weight depression in Syrian golden hamsters is important for the evaluation of nose-only inhalation study results because many normal physiologic parameters, as well as toxicant-induced effects, are associated with body weight status.
- Published
- 1997
50. The cytopathology of benign breast disease: criteria for diagnosis and an assessment of accuracy.
- Author
-
Herbert AP
- Subjects
- Biopsy, Needle statistics & numerical data, Diagnosis, Differential, Female, Humans, Reproducibility of Results, Biopsy, Needle methods, Breast Diseases pathology, Breast Neoplasms pathology
- Published
- 1989
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.