133 results on '"Millhauser GL"'
Search Results
2. Rare Variant Analysis of Human and Rodent Obesity Genes in Individuals with Severe Childhood Obesity
- Author
-
Hendricks, AE, Bochukova, EG, Marenne, G, Keogh, JM, Atanassova, N, Bounds, R, Wheeler, E, Mistry, V, Henning, E, Körner, A, Muddyman, D, McCarthy, S, Hinney, A, Hebebrand, J, Scott, RA, Langenberg, C, Wareham, NJ, Surendran, P, Howson, JM, Butterworth, AS, Danesh, J, Nordestgaard, BG, Nielsen, SF, Afzal, S, Papadia, S, Ashford, S, Garg, S, Millhauser, GL, Palomino, RI, Kwasniewska, A, Tachmazidou, I, O'Rahilly, S, Zeggini, E, Barroso, I, Farooqi, IS, Benzeval, M, Burton, J, Buck, N, Jäckle, A, Kumari, M, Laurie, H, Lynn, P, Pudney, S, Rabe, B, Wolke, D, Overvad, K, Tjønneland, A, Clavel-Chapelon, F, Kaaks, R, Boeing, H, Trichopoulou, A, Ferrari, P, Palli, D, Krogha, V, Panico, S, Tuminoa, R, Matullo, G, Boer, J, Van Der Schouw, Y, Weiderpass, E, Quiros, JR, Sánchez, MJ, Navarro, C, Moreno-Iribas, C, Arriola, L, Melander, O, Wennberg, P, Key, TJ, Riboli, E, Turki, SA, Anderson, CA, Anney, R, Antony, D, Soler Artigas, M, Ayub, M, Bala, S, Barrett, JC, Beales, P, Bentham, J, Bhattacharyaa, S, Birney, E, Blackwooda, D, Bobrow, M, Bolton, PF, Boustred, C, Breen, G, Calissanoa, M, Carss, K, Charlton, R, Chatterjee, K, Chen, L, Ciampia, A, Cirak, S, Clapham, P, Clement, G, Coates, G, Coccaa, M, Collier, DA, Cosgrove, C, Coxa, T, and Crooks, Lucy
- Abstract
© 2017 The Author(s). Obesity is a genetically heterogeneous disorder. Using targeted and whole-exome sequencing, we studied 32 human and 87 rodent obesity genes in 2,548 severely obese children and 1,117 controls. We identified 52 variants contributing to obesity in 2% of cases including multiple novel variants in GNAS, which were sometimes found with accelerated growth rather than short stature as described previously. Nominally significant associations were found for rare functional variants in BBS1, BBS9, GNAS, MKKS, CLOCK and ANGPTL6. The p.S284X variant in ANGPTL6 drives the association signal (rs201622589, MAF∼0.1%, odds ratio = 10.13, p-value = 0.042) and results in complete loss of secretion in cells. Further analysis including additional case-control studies and population controls (N = 260,642) did not support association of this variant with obesity (odds ratio = 2.34, p-value = 2.59 × 10-3), highlighting the challenges of testing rare variant associations and the need for very large sample sizes. Further validation in cohorts with severe obesity and engineering the variants in model organisms will be needed to explore whether human variants in ANGPTL6 and other genes that lead to obesity when deleted in mice, do contribute to obesity. Such studies may yield druggable targets for weight loss therapies.
- Published
- 2017
3. Exploring the peptide 310-helix ? ?-helix equilibrium with double label electron spin resonance
- Author
-
Millhauser Gl and Wayne R. Fiori
- Subjects
Nitroxide mediated radical polymerization ,Chemistry ,Organic Chemistry ,Biophysics ,General Medicine ,Site-directed spin labeling ,Biochemistry ,law.invention ,Biomaterials ,Nuclear magnetic resonance ,law ,310 helix ,Helix ,Electron paramagnetic resonance ,Spin label ,Peptide sequence ,Alpha helix - Abstract
Over the last several years we have used spin labeling as a means for exploring the structure of helical peptides. Two nitroxide labels are engineered into a peptide sequence and distances are ranked with electron spin resonance (ESR). We have found that there is a significant amount of 3(10)-helix in 16-residue model peptides containing only L-amino acids. This review covers several facets of the methodology including spin labeling strategy, interpretation of ESR spectra and the influence of molecular dynamics on the spectral line shapes. Also covered are recent findings of a length-dependent 3(10)-helix-->alpha-helix transition and the role of Arg+ in the stabilization of specific helix structures.
- Published
- 1995
- Full Text
- View/download PDF
4. Functional analysis of the Ala67Thr polymorphism in agouti related protein associated with anorexia nervosa and leanness
- Author
-
de Rijke, C.E., Jackson, PJ, Garner, K.M., van Rozen, RJ, Douglas, NR, Kas, M.J.H., Millhauser, GL, and Adan, R.A.H.
- Published
- 2005
5. Functional analysis of the Ala67Thr polymorphism in agouti related protein associated with anorexia nervosa and leanness.
- Author
-
Translational Neuroscience, de Rijke, C.E., Jackson, PJ, Garner, K.M., van Rozen, RJ, Douglas, NR, Kas, M.J.H., Millhauser, GL, Adan, R.A.H., Translational Neuroscience, de Rijke, C.E., Jackson, PJ, Garner, K.M., van Rozen, RJ, Douglas, NR, Kas, M.J.H., Millhauser, GL, and Adan, R.A.H.
- Published
- 2005
6. Rare Variant Analysis of Human and Rodent Obesity Genes in Individuals with Severe Childhood Obesity
- Author
-
Hendricks, AE, Bochukova, EG, Marenne, G, Keogh, JM, Atanassova, N, Bounds, R, Wheeler, E, Mistry, V, Henning, E, Körner, A, Muddyman, D, McCarthy, S, Hinney, A, Hebebrand, J, Scott, RA, Langenberg, C, Wareham, NJ, Surendran, P, Howson, JM, Butterworth, AS, Danesh, J, Nordestgaard, BG, Nielsen, SF, Afzal, S, Papadia, S, Ashford, S, Garg, S, Millhauser, GL, Palomino, RI, Kwasniewska, A, Tachmazidou, I, O'Rahilly, S, Zeggini, E, Barroso, I, Farooqi, IS, Understanding Society Scientific Group, EPIC-CVD Consortium, and UK10K Consortium
- Subjects
Male ,Models, Molecular ,Pediatric Obesity ,Protein Conformation ,Genetic Variation ,Rodentia ,3. Good health ,Obesity, Morbid ,Pedigree ,Mice ,Case-Control Studies ,Mutation ,Chromogranins ,GTP-Binding Protein alpha Subunits, Gs ,Odds Ratio ,Animals ,Humans ,Female ,Genetic Predisposition to Disease ,Genetic Association Studies - Abstract
Obesity is a genetically heterogeneous disorder. Using targeted and whole-exome sequencing, we studied 32 human and 87 rodent obesity genes in 2,548 severely obese children and 1,117 controls. We identified 52 variants contributing to obesity in 2% of cases including multiple novel variants in GNAS, which were sometimes found with accelerated growth rather than short stature as described previously. Nominally significant associations were found for rare functional variants in BBS1, BBS9, GNAS, MKKS, CLOCK and ANGPTL6. The p.S284X variant in ANGPTL6 drives the association signal (rs201622589, MAF~0.1%, odds ratio = 10.13, p-value = 0.042) and results in complete loss of secretion in cells. Further analysis including additional case-control studies and population controls (N = 260,642) did not support association of this variant with obesity (odds ratio = 2.34, p-value = 2.59 × 10(-3)), highlighting the challenges of testing rare variant associations and the need for very large sample sizes. Further validation in cohorts with severe obesity and engineering the variants in model organisms will be needed to explore whether human variants in ANGPTL6 and other genes that lead to obesity when deleted in mice, do contribute to obesity. Such studies may yield druggable targets for weight loss therapies.
7. Photocatalytic Generation of Singlet Oxygen by Graphitic Carbon Nitride for Antibacterial Applications.
- Author
-
DuBois DB, Rivera I, Liu Q, Yu B, Singewald K, Millhauser GL, Saltikov C, and Chen S
- Abstract
Carbon-based functional nanocomposites have emerged as potent antimicrobial agents and can be exploited as a viable option to overcome antibiotic resistance of bacterial strains. In the present study, graphitic carbon nitride nanosheets are prepared by controlled calcination of urea. Spectroscopic measurements show that the nanosheets consist of abundant carbonyl groups and exhibit apparent photocatalytic activity under UV photoirradiation towards the selective production of singlet oxygen. Therefore, the nanosheets can effectively damage the bacterial cell membranes and inhibit the growth of bacterial cells, such as Gram-negative Escherichia coli , as confirmed in photodynamic, fluorescence microscopy, and scanning electron microscopy measurements. The results from this research highlight the unique potential of carbon nitride derivatives as potent antimicrobial agents.
- Published
- 2024
- Full Text
- View/download PDF
8. N-glycosylation is a potent regulator of prion protein neurotoxicity.
- Author
-
Schilling KM, Jorwal P, Ubilla-Rodriguez NC, Assafa TE, Gatdula JRP, Vultaggio JS, Harris DA, and Millhauser GL
- Abstract
The C-terminal domain of the cellular prion protein (PrP
C ) contains two N-linked glycosylation sites, the occupancy of which impacts disease pathology. In this study, we demonstrate that glycans at these sites are required to maintain an intramolecular interaction with the N-terminal domain, mediated through a previously identified copper-histidine tether, which suppresses the neurotoxic activity of PrPC . NMR and electron paramagnetic resonance spectroscopy demonstrate that the glycans refine the structure of the protein's interdomain interaction. Using whole-cell patch-clamp electrophysiology, we further show that cultured cells expressing PrP molecules with mutated glycosylation sites display large, spontaneous inward currents, a correlate of PrP-induced neurotoxicity. Our findings establish a structural basis for the role of N-linked glycans in maintaining a nontoxic, physiological fold of PrPC ., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
9. Alzheimer's Drug PBT2 Interacts with the Amyloid β 1-42 Peptide Differently than Other 8-Hydroxyquinoline Chelating Drugs.
- Author
-
Summers KL, Roseman G, Schilling KM, Dolgova NV, Pushie MJ, Sokaras D, Kroll T, Harris HH, Millhauser GL, Pickering IJ, and George GN
- Subjects
- Chelating Agents pharmacology, Chelating Agents therapeutic use, Copper chemistry, Humans, Ions, Metals, Oxyquinoline chemistry, Oxyquinoline pharmacology, Peptide Fragments, Solvents, Zinc, Alzheimer Disease drug therapy, Amyloid beta-Peptides chemistry, Clioquinol analogs & derivatives, Clioquinol chemistry
- Abstract
Although Alzheimer's disease (AD) was first described over a century ago, it remains the leading cause of age-related dementia. Innumerable changes have been linked to the pathology of AD; however, there remains much discord regarding which might be the initial cause of the disease. The "amyloid cascade hypothesis" proposes that the amyloid β (Aβ) peptide is central to disease pathology, which is supported by elevated Aβ levels in the brain before the development of symptoms and correlations of amyloid burden with cognitive impairment. The "metals hypothesis" proposes a role for metal ions such as iron, copper, and zinc in the pathology of AD, which is supported by the accumulation of these metals within amyloid plaques in the brain. Metals have been shown to induce aggregation of Aβ, and metal ion chelators have been shown to reverse this reaction in vitro . 8-Hydroxyquinoline-based chelators showed early promise as anti-Alzheimer's drugs. Both 5-chloro-7-iodo-8-hydroxyquinoline (CQ) and 5,7-dichloro-2-[(dimethylamino)methyl]-8-hydroxyquinoline (PBT2) underwent unsuccessful clinical trials for the treatment of AD. To gain insight into the mechanism of action of 8HQs, we have investigated the potential interaction of CQ, PBT2, and 5,7-dibromo-8-hydroxyquinoline (B2Q) with Cu(II)-bound Aβ(1-42) using X-ray absorption spectroscopy (XAS), high energy resolution fluorescence detected (HERFD) XAS, and electron paramagnetic resonance (EPR). By XAS, we found CQ and B2Q sequestered ∼83% of the Cu(II) from Aβ(1-42), whereas PBT2 sequestered only ∼59% of the Cu(II) from Aβ(1-42), suggesting that CQ and B2Q have a higher relative Cu(II) affinity than PBT2. From our EPR, it became clear that PBT2 sequestered Cu(II) from a heterogeneous mixture of Cu(II)Aβ(1-42) species in solution, leaving a single Cu(II)Aβ(1-42) species. It follows that the Cu(II) site in this Cu(II)Aβ(1-42) species is inaccessible to PBT2 and may be less solvent-exposed than in other Cu(II)Aβ(1-42) species. We found no evidence to suggest that these 8HQs form ternary complexes with Cu(II)Aβ(1-42).
- Published
- 2022
- Full Text
- View/download PDF
10. EPR of copper centers in the prion protein.
- Author
-
Quintanar L and Millhauser GL
- Subjects
- Binding Sites, Copper chemistry, Electron Spin Resonance Spectroscopy methods, Zinc metabolism, Prion Proteins chemistry, Prion Proteins metabolism, Prions chemistry, Prions metabolism
- Abstract
Most proteins implicated in neurodegenerative diseases bind metal ions, notably copper and zinc. Metal ion binding may be part of the protein's function or, alternatively, may promote a deleterious gain of function. With regard to Cu
2+ ions, electron paramagnetic resonance techniques have proven to be instrumental in determining the biophysical characteristics of the copper binding sites, as well as structural features of the coordinating protein and how they are impacted by metal binding. Here, the most useful methods are described as they apply to the prion protein, which serves as a model for the broader spectrum of neurodegenerative proteins., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
11. Photodynamic Activity of Graphene Oxide/Polyaniline/Manganese Oxide Ternary Composites toward Both Gram-Positive and Gram-Negative Bacteria.
- Author
-
Chata G, Nichols F, Mercado R, Assafa T, Millhauser GL, Saltikov C, and Chen S
- Subjects
- Aniline Compounds, Anti-Bacterial Agents pharmacology, Escherichia coli, Gram-Negative Bacteria, Gram-Positive Bacteria, Manganese Compounds, Oxides pharmacology, Anti-Infective Agents, Graphite pharmacology
- Abstract
Graphene derivatives have been attracting extensive interest as effective antimicrobial agents. In the present study, ternary nanocomposites are prepared based on graphene oxide quantum dots (GOQD), polyaniline (PANI), and manganese oxides. Because of the hydrophilic GOQD and PANI, the resulting GPM nanocomposites are readily dispersible in water and upon photoirradiation at 365 nm exhibit antimicrobial activity toward both Gram-negative Escherichia coli ( E. coli ) and Gram-positive Staphylococcus epidermidis ( S. epidermidis ). Notably, the nanocomposite with a high Mn
2+ and Mn4+ content is found to be far more active than that with a predominant Mn3+ component, although both samples feature a similar elemental composition and average Mn valence state. The bactericidal activity is largely ascribed to the photocatalytic production of hydroxy radicals and photogenerated holes; both are known to exert oxidative stress on bacterial cells. Further antimicrobial contributions may arise from the strong affinity of the nanocomposites to the cell surfaces. These results suggest that the metal valence state may be a critical parameter in the design and engineering of high-performance antimicrobial agents based on metal oxide nanocomposites.- Published
- 2021
- Full Text
- View/download PDF
12. Copper(II) Binding to PBT2 Differs from That of Other 8-Hydroxyquinoline Chelators: Implications for the Treatment of Neurodegenerative Protein Misfolding Diseases.
- Author
-
Summers KL, Roseman GP, Sopasis GJ, Millhauser GL, Harris HH, Pickering IJ, and George GN
- Subjects
- Animals, Chelating Agents chemical synthesis, Chelating Agents chemistry, Clioquinol chemistry, Clioquinol therapeutic use, Coordination Complexes chemical synthesis, Coordination Complexes chemistry, Copper chemistry, Density Functional Theory, Humans, Ligands, Molecular Structure, Neuroprotective Agents chemical synthesis, Neuroprotective Agents chemistry, X-Ray Absorption Spectroscopy, Alzheimer Disease drug therapy, Chelating Agents therapeutic use, Clioquinol analogs & derivatives, Coordination Complexes therapeutic use, Copper therapeutic use, Neuroprotective Agents pharmacology, Proteostasis Deficiencies drug therapy
- Abstract
PBT2 (5,7-dichloro-2-[(dimethylamino)methyl]-8-hydroxyquinoline) is a small Cu(II)-binding drug that has been investigated in the treatment of neurodegenerative diseases, namely, Alzheimer's disease (AD). PBT2 is thought to be highly effective at crossing the blood-brain barrier and has been proposed to exert anti-Alzheimer's effects through the modulation of metal ion concentrations in the brain, specifically the sequestration of Cu(II) from amyloid plaques. However, despite promising initial results in animal models and in clinical trials where PBT2 was shown to improve cognitive function, larger-scale clinical trials did not find PBT2 to have a significant effect on the amyloid plaque burden compared with controls. We propose that the results of these clinical trials likely point to a more complex mechanism of action for PBT2 other than simple Cu(II) sequestration. To this end, herein we have investigated the solution chemistry of Cu(II) coordination by PBT2 primarily using X-ray absorption spectroscopy (XAS), high-energy-resolution fluorescence-detected XAS, and electron paramagnetic resonance. We propose that a novel bis -PBT2 Cu(II) complex with asymmetric coordination may coexist in solution with a symmetric four-coordinate Cu(II)- bis -PBT2 complex distorted from coplanarity. Additionally, PBT2 is a more flexible ligand than other 8HQs because it can act as both a bidentate and a tridentate ligand as well as coordinate Cu(II) in both 1:1 and 2:1 PBT2/Cu(II) complexes.
- Published
- 2020
- Full Text
- View/download PDF
13. Membrane orientation and oligomerization of the melanocortin receptor accessory protein 2.
- Author
-
Chen V, Bruno AE, Britt LL, Hernandez CC, Gimenez LE, Peisley A, Cone RD, and Millhauser GL
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Cell Membrane genetics, HEK293 Cells, Humans, Protein Domains, Adaptor Proteins, Signal Transducing metabolism, Cell Membrane metabolism, Protein Multimerization
- Abstract
The melanocortin receptor accessory protein 2 (MRAP2) plays a pivotal role in the regulation of several G protein-coupled receptors that are essential for energy balance and food intake. MRAP2 loss-of-function results in obesity in mammals. MRAP2 and its homolog MRAP1 have an unusual membrane topology and are the only known eukaryotic proteins that thread into the membrane in both orientations. In this study, we demonstrate that the conserved polybasic motif that dictates the membrane topology and dimerization of MRAP1 does not control the membrane orientation and dimerization of MRAP2. We also show that MRAP2 dimerizes through its transmembrane domain and can form higher-order oligomers that arrange MRAP2 monomers in a parallel orientation. Investigating the molecular details of MRAP2 structure is essential for understanding the mechanism by which it regulates G protein-coupled receptors and will aid in elucidating the pathways involved in metabolic dysfunction., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Chen et al.)
- Published
- 2020
- Full Text
- View/download PDF
14. Evidence for aggregation-independent, PrP C -mediated Aβ cellular internalization.
- Author
-
Foley AR, Roseman GP, Chan K, Smart A, Finn TS, Yang K, Lokey RS, Millhauser GL, and Raskatov JA
- Subjects
- HEK293 Cells, Humans, Amyloid beta-Peptides metabolism, Peptide Fragments metabolism, PrPC Proteins metabolism
- Abstract
Evidence linking amyloid beta (Aβ) cellular uptake and toxicity has burgeoned, and mechanisms underlying this association are subjects of active research. Two major, interconnected questions are whether Aβ uptake is aggregation-dependent and whether it is sequence-specific. We recently reported that the neuronal uptake of Aβ depends significantly on peptide chirality, suggesting that the process is predominantly receptor-mediated. Over the past decade, the cellular prion protein (PrP
C ) has emerged as an important mediator of Aβ-induced toxicity and of neuronal Aβ internalization. Here, we report that the soluble, nonfibrillizing Aβ (1-30) peptide recapitulates full-length Aβ stereoselective cellular uptake, allowing us to decouple aggregation from cellular, receptor-mediated internalization. Moreover, we found that Aβ (1-30) uptake is also dependent on PrPC expression. NMR-based molecular-level characterization identified the docking site on PrPC that underlies the stereoselective binding of Aβ (1-30). Our findings therefore identify a specific sequence within Aβ that is responsible for the recognition of the peptide by PrPC , as well as PrPC -dependent cellular uptake. Further uptake stereodifferentiation in PrPC -free cells points toward additional receptor-mediated interactions as likely contributors for Aβ cellular internalization. Taken together, our results highlight the potential of targeting cellular surface receptors to inhibit Aβ cellular uptake as an alternative route for future therapeutic development for Alzheimer's disease., Competing Interests: The authors declare no competing interest.- Published
- 2020
- Full Text
- View/download PDF
15. Both N-Terminal and C-Terminal Histidine Residues of the Prion Protein Are Essential for Copper Coordination and Neuroprotective Self-Regulation.
- Author
-
Schilling KM, Tao L, Wu B, Kiblen JTM, Ubilla-Rodriguez NC, Pushie MJ, Britt RD, Roseman GP, Harris DA, and Millhauser GL
- Subjects
- Animals, DNA Repeat Expansion, Histidine metabolism, Mice, Models, Molecular, Molecular Dynamics Simulation, Prion Proteins genetics, Protein Conformation, Protein Domains, Protein Folding, Copper metabolism, Mutation, Prion Proteins chemistry, Prion Proteins metabolism
- Abstract
The cellular prion protein (PrP
C ) comprises two domains: a globular C-terminal domain and an unstructured N-terminal domain. Recently, copper has been observed to drive tertiary contact in PrPC , inducing a neuroprotective cis interaction that structurally links the protein's two domains. The location of this interaction on the C terminus overlaps with the sites of human pathogenic mutations and toxic antibody docking. Combined with recent evidence that the N terminus is a toxic effector regulated by the C terminus, there is an emerging consensus that this cis interaction serves a protective role, and that the disruption of this interaction by misfolded PrP oligomers may be a cause of toxicity in prion disease. We demonstrate here that two highly conserved histidines in the C-terminal domain of PrPC are essential for the protein's cis interaction, which helps to protect against neurotoxicity carried out by its N terminus. We show that simultaneous mutation of these histidines drastically weakens the cis interaction and enhances spontaneous cationic currents in cultured cells, the first C-terminal mutant to do so. Whereas previous studies suggested that Cu2+ coordination was localized solely to the protein's N-terminal domain, we find that both domains contribute equatorially coordinated histidine residue side-chains, resulting in a novel bridging interaction. We also find that extra N-terminal histidines in pathological familial mutations involving octarepeat expansions inhibit this interaction by sequestering copper from the C terminus. Our findings further establish a structural basis for PrPC 's C-terminal regulation of its otherwise toxic N terminus., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
16. Intrinsic toxicity of the cellular prion protein is regulated by its conserved central region.
- Author
-
Roseman GP, Wu B, Wadolkowski MA, Harris DA, and Millhauser GL
- Subjects
- Cell Line, Cell Survival physiology, HEK293 Cells, Humans, Magnetic Resonance Spectroscopy methods, Conserved Sequence physiology, PrPC Proteins metabolism, Prion Proteins metabolism
- Abstract
The conserved central region (CR) of PrP
C has been hypothesized to serve as a passive linker connecting the protein's toxic N-terminal and globular C-terminal domains. Yet, deletion of the CR causes neonatal fatality in mice, implying the CR possesses a protective function. The CR encompasses the regulatory α-cleavage locus, and additionally facilitates a regulatory metal ion-promoted interaction between the PrPC N- and C-terminal domains. To elucidate the role of the CR and determine why CR deletion generates toxicity, we designed PrPC constructs wherein either the cis-interaction or α-cleavage are selectively prevented. These constructs were interrogated using nuclear magnetic resonance, electrophysiology, and cell viability assays. Our results demonstrate the CR is not a passive linker and the native sequence is crucial for its protective role over the toxic N-terminus, irrespective of α-cleavage or the cis-interaction. Additionally, we find that the CR facilitates homodimerization of PrPC , attenuating the toxicity of the N-terminus., (© 2020 Federation of American Societies for Experimental Biology.)- Published
- 2020
- Full Text
- View/download PDF
17. Determination of the melanocortin-4 receptor structure identifies Ca 2+ as a cofactor for ligand binding.
- Author
-
Yu J, Gimenez LE, Hernandez CC, Wu Y, Wein AH, Han GW, McClary K, Mittal SR, Burdsall K, Stauch B, Wu L, Stevens SN, Peisley A, Williams SY, Chen V, Millhauser GL, Zhao S, Cone RD, and Stevens RC
- Subjects
- Crystallography, X-Ray, Cyclic AMP chemistry, Humans, Ligands, Melanocyte-Stimulating Hormones chemistry, Melanocyte-Stimulating Hormones pharmacology, Mutation, Potassium Channels, Inwardly Rectifying chemistry, Protein Binding, Protein Multimerization, Protein Structure, Secondary, Receptor, Melanocortin, Type 4 antagonists & inhibitors, Receptor, Melanocortin, Type 4 genetics, Receptors, G-Protein-Coupled antagonists & inhibitors, Receptors, G-Protein-Coupled genetics, Signal Transduction, Calcium chemistry, Receptor, Melanocortin, Type 4 chemistry, Receptors, G-Protein-Coupled chemistry
- Abstract
The melanocortin-4 receptor (MC4R) is involved in energy homeostasis and is an important drug target for syndromic obesity. We report the structure of the antagonist SHU9119-bound human MC4R at 2.8-angstrom resolution. Ca
2+ is identified as a cofactor that is complexed with residues from both the receptor and peptide ligand. Extracellular Ca2+ increases the affinity and potency of the endogenous agonist α-melanocyte-stimulating hormone at the MC4R by 37- and 600-fold, respectively. The ability of the MC4R crystallized construct to couple to ion channel Kir7.1, while lacking cyclic adenosine monophosphate stimulation, highlights a heterotrimeric GTP-binding protein (G protein)-independent mechanism for this signaling modality. MC4R is revealed as a structurally divergent G protein-coupled receptor (GPCR), with more similarity to lipidic GPCRs than to the homologous peptidic GPCRs., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2020
- Full Text
- View/download PDF
18. Production of Artificially Doubly Glycosylated, 15 N Labeled Prion Protein for NMR Studies Using a pH-Scanning Volatile Buffer System.
- Author
-
Schilling KM, Ubilla-Rodriguez NC, Wells CW, and Millhauser GL
- Subjects
- Hydrogen-Ion Concentration, Magnetic Resonance Imaging, Magnetic Resonance Spectroscopy, Polysaccharides, Prion Proteins
- Abstract
Bacterially expressed proteins used in NMR studies lack glycans, and proteins from other organisms are neither
15 N labeled nor glycosylated homogeneously. Here, we add two artificial glycans to uniformly15 N labeled prion protein using a buffer system that evolves over a pH range to accommodate the conflicting pH requirements of the substrate and enzymes without the need to fine-tune buffer conditions. NMR and CD spectroscopy of the protein indicates that the glycans do not influence its fold.- Published
- 2020
- Full Text
- View/download PDF
19. First Synthesis of Mn-Doped Cesium Lead Bromide Perovskite Magic Sized Clusters at Room Temperature.
- Author
-
Xu K, Vickers ET, Luo B, Allen AC, Chen E, Roseman G, Wang Q, Kliger DS, Millhauser GL, Yang W, Li X, and Zhang JZ
- Abstract
Mn-doped CsPbBr
3 perovskite magic sized clusters (PMSCs) are synthesized for the first time using benzoic acid and benzylamine as passivating ligands and MnCl2 ·4H2 O and MnBr2 as the Mn2+ dopant sources at room temperature. The same approach is used to prepare Mn-doped CsPbBr3 perovskite quantum dots (PQDs). The concentration of MnX2 (X = Cl or Br) affects the excitonic absorption of the PMSCs and PQDs. A higher concentration of MnX2 favors PMSCs over PQDs as well as higher photoluminescence (PL) quantum yields (QYs) and PL stability. The large ratio between the characteristic Mn emission (∼590 nm) and the host band-edge emission shows efficient energy transfer from the host exciton to the Mn2+ dopant. PL excitation, electron paramagnetic resonance, and time-resolved PL results all support Mn2+ doping in CsPbBr3 , which likely replaces Pb2+ ions. This study establishes a new method for synthesizing Mn-doped PMSCs with good PL stability, high PLQY and highly effective passivation.- Published
- 2020
- Full Text
- View/download PDF
20. Antimicrobial activity of graphene oxide quantum dots: impacts of chemical reduction.
- Author
-
Rojas-Andrade MD, Nguyen TA, Mistler WP, Armas J, Lu JE, Roseman G, Hollingsworth WR, Nichols F, Millhauser GL, Ayzner A, Saltikov C, and Chen S
- Abstract
Design and engineering of graphene-based functional nanomaterials for effective antimicrobial applications has been attracting extensive interest. In the present study, graphene oxide quantum dots (GOQDs) were prepared by chemical exfoliation of carbon fibers and exhibited apparent antimicrobial activity. Transmission electron microscopic measurements showed that the lateral length ranged from a few tens to a few hundred nanometers. Upon reduction by sodium borohydride, whereas the UV-vis absorption profile remained largely unchanged, steady-state photoluminescence measurements exhibited a marked blue-shift and increase in intensity of the emission, due to (partial) removal of phenanthroline-like structural defects within the carbon skeletons. Consistent results were obtained in Raman and time-resolved photoluminescence measurements. Interestingly, the samples exhibited apparent, but clearly different, antimicrobial activity against Staphylococcus epidermidis cells. In the dark and under photoirradiation (400 nm), the as-produced GOQDs exhibited markedly higher cytotoxicity than the chemically reduced counterparts, likely because of (i) effective removal by NaBH
4 reduction of redox-active phenanthroline-like moieties that interacted with the electron-transport chain of the bacterial cells, and (ii) diminished production of hydroxyl radicals that were potent bactericidal agents after chemical reduction as a result of increased conjugation within the carbon skeletons., Competing Interests: There are no conflicts to declare., (This journal is © The Royal Society of Chemistry.)- Published
- 2020
- Full Text
- View/download PDF
21. Charge Characteristics of Agouti-Related Protein Implicate Potent Involvement of Heparan Sulfate Proteoglycans in Metabolic Function.
- Author
-
Chen J, Chen V, Kawamura T, Hoang I, Yang Y, Wong AT, McBride R, Repunte-Canonigo V, Millhauser GL, and Sanna PP
- Abstract
The endogenous melanocortin peptide agouti-related protein (AgRP) plays a well-known role in foraging, but its contribution to metabolic regulation is less understood. Mature AgRP
(83-132) has distinct residues for melanocortin receptor binding and heparan sulfate interactions. Here, we show that AgRP increases ad libitum feeding and operant responding for food in mice, decreases oxygen consumption, and lowers body temperature and activity, indicating lower energy expenditure. AgRP increased the respiratory exchange ratio, indicating a reduction of fat oxidation and a shift toward carbohydrates as the primary fuel source. The duration and intensity of AgRP's effects depended on the density of its positively charged amino acids, suggesting that its orexigenic and metabolic effects depend on its affinity for heparan sulfate. These findings may have major clinical implications by unveiling the critical involvement of interactions between AgRP and heparan sulfate to the central regulation of energy expenditure, fat utilization, and possibly their contribution to metabolic disease., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
22. Altered Domain Structure of the Prion Protein Caused by Cu 2+ Binding and Functionally Relevant Mutations: Analysis by Cross-Linking, MS/MS, and NMR.
- Author
-
McDonald AJ, Leon DR, Markham KA, Wu B, Heckendorf CF, Schilling K, Showalter HD, Andrews PC, McComb ME, Pushie MJ, Costello CE, Millhauser GL, and Harris DA
- Subjects
- Animals, Cell Line, Copper metabolism, Cross-Linking Reagents chemistry, Cross-Linking Reagents metabolism, Humans, Magnetic Resonance Spectroscopy methods, Mice, Prion Proteins metabolism, Protein Binding, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms metabolism, Tandem Mass Spectrometry methods, Copper chemistry, Molecular Dynamics Simulation, Mutation, Prion Proteins chemistry, Prion Proteins genetics, Protein Domains
- Abstract
The cellular isoform of the prion protein (PrP
C ) serves as precursor to the infectious isoform (PrPSc ), and as a cell-surface receptor, which binds misfolded protein oligomers as well as physiological ligands such as Cu2+ ions. PrPC consists of two domains: a flexible N-terminal domain and a structured C-terminal domain. Both the physiological and pathological functions of PrP depend on intramolecular interactions between these two domains, but the specific amino acid residues involved have proven challenging to define. Here, we employ a combination of chemical cross-linking, mass spectrometry, NMR, molecular dynamics simulations, and functional assays to identify residue-level contacts between the N- and C-terminal domains of PrPC . We also determine how these interdomain contacts are altered by binding of Cu2+ ions and by functionally relevant mutations. Our results provide a structural basis for interpreting both the normal and toxic activities of PrP., (Copyright © 2019 Elsevier Ltd. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
23. X-ray Absorption Spectroscopy Investigations of Copper(II) Coordination in the Human Amyloid β Peptide.
- Author
-
Summers KL, Schilling KM, Roseman G, Markham KA, Dolgova NV, Kroll T, Sokaras D, Millhauser GL, Pickering IJ, and George GN
- Subjects
- Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Copper metabolism, Histidine metabolism, Humans, Hydrogen-Ion Concentration, Oxidation-Reduction, Peptide Fragments metabolism, X-Ray Absorption Spectroscopy, Amyloid beta-Peptides chemistry, Copper chemistry, Histidine chemistry, Peptide Fragments chemistry
- Abstract
Alzheimer's disease (AD) is the main cause of age-related dementia and currently affects approximately 5.7 million Americans. Major brain changes associated with AD pathology include accumulation of amyloid beta (Aβ) protein fragments and formation of extracellular amyloid plaques. Redox-active metals mediate oligomerization of Aβ, and the resultant metal-bound oligomers have been implicated in the putative formation of harmful, reactive species that could contribute to observed oxidative damage. In isolated plaque cores, Cu(II) is bound to Aβ via histidine residues. Despite numerous structural studies of Cu(II) binding to synthetic Aβ in vitro, there is still uncertainty surrounding Cu(II) coordination in Aβ. In this study, we used X-ray absorption spectroscopy (XAS) and high energy resolution fluorescence detected (HERFD) XAS to investigate Cu(II) coordination in Aβ(1-42) under various solution conditions. We found that the average coordination environment in Cu(II)Aβ(1-42) is sensitive to X-ray photoreduction, changes in buffer composition, peptide concentration, and solution pH. Fitting of the extended X-ray absorption fine structure (EXAFS) suggests Cu(II) is bound in a mixture of coordination environments in monomeric Aβ(1-42) under all conditions studied. However, it was evident that on average only a single histidine residue coordinates Cu(II) in monomeric Aβ(1-42) at pH 6.1, in addition to 3 other oxygen or nitrogen ligands. Cu(II) coordination in Aβ(1-42) at pH 7.4 is similarly 4-coordinate with oxygen and nitrogen ligands, although an average of 2 histidine residues appear to coordinate at this pH. At pH 9.0, the average Cu(II) coordination environment in Aβ(1-42) appears to be 5-coordinate with oxygen and nitrogen ligands, including two histidine residues.
- Published
- 2019
- Full Text
- View/download PDF
24. Molecular Features of the Zn 2+ Binding Site in the Prion Protein Probed by 113 Cd NMR.
- Author
-
Markham KA, Roseman GP, Linsley RB, Lee HW, and Millhauser GL
- Subjects
- Amino Acid Sequence, Binding Sites, Cadmium chemistry, Histidine chemistry, Imidazoles chemistry, Models, Molecular, Mutation, Prion Proteins genetics, Protein Binding, Nuclear Magnetic Resonance, Biomolecular, Prion Proteins chemistry, Prion Proteins metabolism, Zinc metabolism
- Abstract
The cellular prion protein (PrP
C ) is a zinc-binding protein that contributes to the regulation of Zn2+ and other divalent species of the central nervous system. Zn2+ coordinates to the flexible, N-terminal repeat region of PrPC and drives a tertiary contact between this repeat region and a well-defined cleft of the C-terminal domain. The tertiary structure promoted by Zn2+ is thought to regulate inherent PrPC toxicity. Despite the emerging consensus regarding the interaction between Zn2+ and PrPC , there is little direct spectroscopic confirmation of the metal ion's coordination details. Here, we address this conceptual gap by using Cd2+ as a surrogate for Zn2+ . NMR finds that Cd2+ binds exclusively to the His imidazole side chains of the repeat segment, with a dissociation constant of ∼1.2 mM, and promotes an N-terminal-C-terminal cis interaction very similar to that observed with Zn2+ . Analysis of113 Cd NMR spectra of PrPC , along with relevant control proteins and peptides, suggests that coordination of Cd2+ in the full-length protein is consistent with a three- or four-His geometry. Examination of the mutation E199K in mouse PrPC (E200K in humans), responsible for inherited Creutzfeldt-Jakob disease, finds that the mutation lowers metal ion affinity and weakens the cis interaction. These findings not only provide deeper insight into PrPC metal ion coordination but they also suggest new perspectives on the role of familial mutations in prion disease., (Copyright © 2019 Biophysical Society. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
25. Late onset obesity in mice with targeted deletion of potassium inward rectifier Kir7.1 from cells expressing the melanocortin-4 receptor.
- Author
-
Anderson EJP, Ghamari-Langroudi M, Cakir I, Litt MJ, Chen V, Reggiardo RE, Millhauser GL, and Cone RD
- Subjects
- Animals, Feeding Behavior physiology, Female, Male, Membrane Potentials, Mice, Inbred C57BL, Mice, Knockout, Potassium Channels, Inwardly Rectifying genetics, Hypothalamus physiopathology, Neurons physiology, Obesity physiopathology, Potassium Channels, Inwardly Rectifying physiology, Receptor, Melanocortin, Type 4 physiology
- Abstract
Energy stores in fat tissue are determined in part by the activity of hypothalamic neurones expressing the melanocortin-4 receptor (MC4R). Even a partial reduction in MC4R expression levels in mice, rats or humans produces hyperphagia and morbid obesity. Thus, it is of great interest to understand the molecular basis of neuromodulation by the MC4R. The MC4R is a G protein-coupled receptor that signals efficiently through Gα
S , and this signalling pathway is essential for normal MC4R function in vivo. However, previous data from hypothalamic slice preparations indicated that activation of the MC4R depolarised neurones via G protein-independent regulation of the ion channel Kir7.1. In the present study, we show that deletion of Kcnj13 (ie, the gene encoding Kir7.1) specifically from MC4R neurones produced resistance to melanocortin peptide-induced depolarisation of MC4R paraventricular nucleus neurones in brain slices, resistance to the sustained anorexic effect of exogenously administered melanocortin peptides, late onset obesity, increased linear growth and glucose intolerance. Some MC4R-mediated phenotypes appeared intact, including Agouti-related peptide-induced stimulation of food intake and MC4R-mediated induction of peptide YY release from intestinal L cells. Thus, a subset of the consequences of MC4R signalling in vivo appears to be dependent on expression of the Kir7.1 channel in MC4R cells., (© 2018 British Society for Neuroendocrinology.)- Published
- 2019
- Full Text
- View/download PDF
26. Photo-enhanced antibacterial activity of ZnO/graphene quantum dot nanocomposites.
- Author
-
Liu J, Rojas-Andrade MD, Chata G, Peng Y, Roseman G, Lu JE, Millhauser GL, Saltikov C, and Chen S
- Abstract
Synthesis of new, highly active antibacterial agents has become increasingly important in light of emerging antibiotic resistance. In the present study, ZnO/graphene quantum dot (GQD) nanocomposites were produced by a facile hydrothermal method and characterized by an array of microscopic and spectroscopic measurements, including transmission electron microscopy, X-ray photoelectron spectroscopy, UV-vis and photoluminescence spectroscopy. Antibacterial activity of the ZnO/GQD nanocomposites was evaluated with Escherichia coli within the context of minimum inhibitory concentration and the reduction of the number of bacterial colonies in a standard plate count method, in comparison to those with ZnO and GQD separately. It was found that the activity was markedly enhanced under UV photoirradiation as compared to that in ambient light. This was ascribed to the enhanced generation of reactive oxygen species under UV photoirradiation, with minor contributions from membrane damage, as manifested in electron paramagnetic resonance and fluorescence microscopic measurements. The results highlight the significance of functional nanocomposites based on semiconductor nanoparticles and graphene derivatives in the development of effective bactericidal agents.
- Published
- 2017
- Full Text
- View/download PDF
27. Rare Variant Analysis of Human and Rodent Obesity Genes in Individuals with Severe Childhood Obesity.
- Author
-
Hendricks AE, Bochukova EG, Marenne G, Keogh JM, Atanassova N, Bounds R, Wheeler E, Mistry V, Henning E, Körner A, Muddyman D, McCarthy S, Hinney A, Hebebrand J, Scott RA, Langenberg C, Wareham NJ, Surendran P, Howson JM, Butterworth AS, Danesh J, Nordestgaard BG, Nielsen SF, Afzal S, Papadia S, Ashford S, Garg S, Millhauser GL, Palomino RI, Kwasniewska A, Tachmazidou I, O'Rahilly S, Zeggini E, Barroso I, and Farooqi IS
- Subjects
- Animals, Case-Control Studies, Chromogranins chemistry, Chromogranins genetics, Chromogranins metabolism, Female, GTP-Binding Protein alpha Subunits, Gs chemistry, GTP-Binding Protein alpha Subunits, Gs genetics, GTP-Binding Protein alpha Subunits, Gs metabolism, Humans, Male, Mice, Models, Molecular, Mutation, Obesity, Morbid diagnosis, Odds Ratio, Pediatric Obesity diagnosis, Pedigree, Protein Conformation, Rodentia, Genetic Association Studies, Genetic Predisposition to Disease, Genetic Variation, Obesity, Morbid genetics, Pediatric Obesity genetics
- Abstract
Obesity is a genetically heterogeneous disorder. Using targeted and whole-exome sequencing, we studied 32 human and 87 rodent obesity genes in 2,548 severely obese children and 1,117 controls. We identified 52 variants contributing to obesity in 2% of cases including multiple novel variants in GNAS, which were sometimes found with accelerated growth rather than short stature as described previously. Nominally significant associations were found for rare functional variants in BBS1, BBS9, GNAS, MKKS, CLOCK and ANGPTL6. The p.S284X variant in ANGPTL6 drives the association signal (rs201622589, MAF~0.1%, odds ratio = 10.13, p-value = 0.042) and results in complete loss of secretion in cells. Further analysis including additional case-control studies and population controls (N = 260,642) did not support association of this variant with obesity (odds ratio = 2.34, p-value = 2.59 × 10
-3 ), highlighting the challenges of testing rare variant associations and the need for very large sample sizes. Further validation in cohorts with severe obesity and engineering the variants in model organisms will be needed to explore whether human variants in ANGPTL6 and other genes that lead to obesity when deleted in mice, do contribute to obesity. Such studies may yield druggable targets for weight loss therapies.- Published
- 2017
- Full Text
- View/download PDF
28. The N-terminus of the prion protein is a toxic effector regulated by the C-terminus.
- Author
-
Wu B, McDonald AJ, Markham K, Rich CB, McHugh KP, Tatzelt J, Colby DW, Millhauser GL, and Harris DA
- Subjects
- Animals, Dendrites pathology, Hippocampus pathology, Magnetic Resonance Spectroscopy, Mice, Neurons pathology, PrPC Proteins chemistry, Prion Proteins chemistry, Protein Conformation, Homeostasis, PrPC Proteins toxicity, Prion Proteins toxicity
- Abstract
PrP
C , the cellular isoform of the prion protein, serves to transduce the neurotoxic effects of PrPSc , the infectious isoform, but how this occurs is mysterious. Here, using a combination of electrophysiological, cellular, and biophysical techniques, we show that the flexible, N-terminal domain of PrPC functions as a powerful toxicity-transducing effector whose activity is tightly regulated in cis by the globular C-terminal domain. Ligands binding to the N-terminal domain abolish the spontaneous ionic currents associated with neurotoxic mutants of PrP, and the isolated N-terminal domain induces currents when expressed in the absence of the C-terminal domain. Anti-PrP antibodies targeting epitopes in the C-terminal domain induce currents, and cause degeneration of dendrites on murine hippocampal neurons, effects that entirely dependent on the effector function of the N-terminus. NMR experiments demonstrate intramolecular docking between N- and C-terminal domains of PrPC , revealing a novel auto-inhibitory mechanism that regulates the functional activity of PrPC .- Published
- 2017
- Full Text
- View/download PDF
29. The agouti-related peptide binds heparan sulfate through segments critical for its orexigenic effects.
- Author
-
Palomino R, Lee HW, and Millhauser GL
- Subjects
- Agouti-Related Protein metabolism, Heparitin Sulfate metabolism, Humans, Peptides metabolism, Protein Binding, Protein Domains, Agouti-Related Protein chemistry, Heparitin Sulfate chemistry, Peptides chemistry
- Abstract
Syndecans potently modulate agouti-related peptide (AgRP) signaling in the central melanocortin system. Through heparan sulfate moieties, syndecans are thought to anchor AgRP near its receptor, enhancing its orexigenic effects. Original work proposed that the N-terminal domain of AgRP facilitates this interaction. However, this is not compatible with evidence that this domain is posttranslationally cleaved. Addressing this long-standing incongruity, we used calorimetry and magnetic resonance to probe interactions of AgRP peptides with glycosaminoglycans, including heparan sulfate. We show that mature, cleaved, C-terminal AgRP, not the N-terminal domain, binds heparan sulfate. NMR shows that the binding site consists of regions distinct from the melanocortin receptor-binding site. Using a library of designed AgRP variants, we find that the strength of the syndecan interaction perfectly tracks orexigenic action. Our data provide compelling evidence that AgRP is a heparan sulfate-binding protein and localizes critical regions in the AgRP structure required for this interaction., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
30. Copper- and Zinc-Promoted Interdomain Structure in the Prion Protein: A Mechanism for Autoinhibition of the Neurotoxic N-Terminus.
- Author
-
Evans EGB and Millhauser GL
- Subjects
- Animals, Humans, Models, Molecular, Protein Domains, Copper pharmacology, Neurotoxins antagonists & inhibitors, Neurotoxins chemistry, Prion Proteins antagonists & inhibitors, Prion Proteins chemistry, Zinc pharmacology
- Abstract
The function of the cellular prion protein (PrP
C ), while still poorly understood, is increasingly linked to its ability to bind physiological metal ions at the cell surface. PrPC binds divalent forms of both copper and zinc through its unstructured N-terminal domain, modulating interactions between PrPC and various receptors at the cell surface and ultimately tuning downstream cellular processes. In this chapter, we briefly discuss the molecular features of copper and zinc uptake by PrPC and summarize evidence implicating these metal ions in PrP-mediated physiology. We then focus our review on recent biophysical evidence revealing a physical interaction between the flexible N-terminal and globular C-terminal domains of PrPC . This interdomain cis interaction is electrostatic in nature and is promoted by the binding of Cu2+ and Zn2+ to the N-terminal octarepeat domain. These findings, along with recent cellular studies, suggest a mechanism whereby NC interactions serve to regulate the activity and/or toxicity of the PrPC N-terminus. We discuss this potential mechanism in relation to familial prion disease mutations, lethal deletions of the PrPC central region, and neurotoxicity induced by certain globular domain ligands, including bona fide prions and toxic amyloid-β oligomers., (© 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
31. Interaction between Prion Protein's Copper-Bound Octarepeat Domain and a Charged C-Terminal Pocket Suggests a Mechanism for N-Terminal Regulation.
- Author
-
Evans EG, Pushie MJ, Markham KA, Lee HW, and Millhauser GL
- Subjects
- Animals, Binding Sites, Mice, Molecular Docking Simulation, Prion Proteins metabolism, Protein Binding, Copper metabolism, Prion Proteins chemistry
- Abstract
Copper plays a critical role in prion protein (PrP) physiology. Cu(2+) binds with high affinity to the PrP N-terminal octarepeat (OR) domain, and intracellular copper promotes PrP expression. The molecular details of copper coordination within the OR are now well characterized. Here we examine how Cu(2+) influences the interaction between the PrP N-terminal domain and the C-terminal globular domain. Using nuclear magnetic resonance and copper-nitroxide pulsed double electron-electron resonance, with molecular dynamics refinement, we localize the position of Cu(2+) in its high-affinity OR-bound state. Our results reveal an interdomain cis interaction that is stabilized by a conserved, negatively charged pocket of the globular domain. Interestingly, this interaction surface overlaps an epitope recognized by the POM1 antibody, the binding of which drives rapid cerebellar degeneration mediated by the PrP N terminus. The resulting structure suggests that the globular domain regulates the N-terminal domain by binding the Cu(2+)-occupied OR within a complementary pocket., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
32. Correction: Human β-D-3 Exacerbates MDA5 but Suppresses TLR3 Responses to the Viral Molecular Pattern Mimic Polyinosinic:Polycytidylic Acid.
- Author
-
Semple F, MacPherson H, Webb S, Kilanowski F, Lettice L, McGlasson SL, Wheeler AP, Chen V, Millhauser GL, Melrose L, Davidson DJ, and Dorin JR
- Abstract
[This corrects the article DOI: 10.1371/journal.pgen.1005673.].
- Published
- 2016
- Full Text
- View/download PDF
33. Human β-Defensin 3 [corrected] Exacerbates MDA5 but Suppresses TLR3 Responses to the Viral Molecular Pattern Mimic Polyinosinic:Polycytidylic Acid.
- Author
-
Semple F, MacPherson H, Webb S, Kilanowski F, Lettice L, McGlasson SL, Wheeler AP, Chen V, Millhauser GL, Melrose L, Davidson DJ, and Dorin JR
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Vesicular Transport metabolism, Animals, Bone Marrow, Chemokine CXCL10 genetics, DEAD-box RNA Helicases metabolism, Humans, Immunity, Innate genetics, Interferon-Induced Helicase, IFIH1, Liposomes metabolism, Macrophages metabolism, Macrophages pathology, Mice, Mice, Knockout, Poly I-C administration & dosage, Psoriasis pathology, Toll-Like Receptor 3 antagonists & inhibitors, beta-Defensins metabolism, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Vesicular Transport genetics, DEAD-box RNA Helicases genetics, Psoriasis genetics, Toll-Like Receptor 3 genetics, beta-Defensins genetics
- Abstract
Human β-defensin 3 (hBD3) is a cationic host defence peptide and is part of the innate immune response. HBD3 is present on a highly copy number variable block of six β-defensin genes, and increased copy number is associated with the autoimmune disease psoriasis. It is not known how this increase influences disease development, but psoriasis is a T cell-mediated disease and activation of the innate immune system is required for the initial trigger that leads to the amplification stage. We investigated the effect of hBD3 on the response of primary macrophages to various TLR agonists. HBD3 exacerbated the production of type I Interferon-β in response to the viral ligand mimic polyinosinic:polycytidylic acid (polyI:C) in both human and mouse primary cells, although production of the chemokine CXCL10 was suppressed. Compared to polyI:C alone, mice injected with both hBD3 peptide and polyI:C also showed an enhanced increase in Interferon-β. Mice expressing a transgene encoding hBD3 had elevated basal levels of Interferon-β, and challenge with polyI:C further increased this response. HBD3 peptide increased uptake of polyI:C by macrophages, however the cellular response and localisation of polyI:C in cells treated contemporaneously with hBD3 or cationic liposome differed. Immunohistochemistry showed that hBD3 and polyI:C do not co-localise, but in the presence of hBD3 less polyI:C localises to the early endosome. Using bone marrow derived macrophages from knockout mice we demonstrate that hBD3 suppresses the polyI:C-induced TLR3 response mediated by TICAM1 (TRIF), while exacerbating the cytoplasmic response through MDA5 (IFIH1) and MAVS (IPS1/CARDIF). Thus, hBD3, a highly copy number variable gene in human, influences cellular responses to the viral mimic polyI:C implying that copy number may have a significant phenotypic effect on the response to viral infection and development of autoimmunity in humans.
- Published
- 2015
- Full Text
- View/download PDF
34. Electrostatic Similarity Analysis of Human β-Defensin Binding in the Melanocortin System.
- Author
-
Nix MA, Kaelin CB, Palomino R, Miller JL, Barsh GS, and Millhauser GL
- Subjects
- Amino Acid Sequence, Binding, Competitive, Databases, Protein, Humans, Models, Chemical, Models, Molecular, Molecular Sequence Data, Peptides chemistry, Protein Binding, Protein Folding, Sequence Alignment, Sequence Homology, Amino Acid, beta-Defensins genetics, Receptor, Melanocortin, Type 1 chemistry, Static Electricity, beta-Defensins chemistry
- Abstract
The β-defensins are a class of small cationic proteins that serve as components of numerous systems in vertebrate biology, including the immune and melanocortin systems. Human β-defensin 3 (HBD3), which is produced in the skin, has been found to bind to melanocortin receptors 1 and 4 through complementary electrostatics, a unique mechanism of ligand-receptor interaction. This finding indicates that electrostatics alone, and not specific amino acid contact points, could be sufficient for function in this ligand-receptor system, and further suggests that other small peptide ligands could interact with these receptors in a similar fashion. Here, we conducted molecular-similarity analyses and functional studies of additional members of the human β-defensin family, examining their potential as ligands of melanocortin-1 receptor, through selection based on their electrostatic similarity to HBD3. Using Poisson-Boltzmann electrostatic calculations and molecular-similarity analysis, we identified members of the human β-defensin family that are both similar and dissimilar to HBD3 in terms of electrostatic potential. Synthesis and functional testing of a subset of these β-defensins showed that peptides with an HBD3-like electrostatic character bound to melanocortin receptors with high affinity, whereas those that were anticorrelated to HBD3 showed no binding affinity. These findings expand on the central role of electrostatics in the control of this ligand-receptor system and further demonstrate the utility of employing molecular-similarity analysis. Additionally, we identified several new potential ligands of melanocortin-1 receptor, which may have implications for our understanding of the role defensins play in melanocortin physiology., (Copyright © 2015 Biophysical Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. n-Dopants Based on Dimers of Benzimidazoline Radicals: Structures and Mechanism of Redox Reactions.
- Author
-
Zhang S, Naab BD, Jucov EV, Parkin S, Evans EG, Millhauser GL, Timofeeva TV, Risko C, Brédas JL, Bao Z, Barlow S, and Marder SR
- Subjects
- Benzene Derivatives chemistry, Crystallography, X-Ray, Dimerization, Electron Transport, Models, Molecular, Naphthacenes chemistry, Oxidation-Reduction, Quantum Theory, Thermodynamics, Free Radicals chemistry, Imidazolines chemistry, Semiconductors
- Abstract
Dimers of 2-substituted N,N'-dimethylbenzimidazoline radicals, (2-Y-DMBI)2 (Y=cyclohexyl (Cyc), ferrocenyl (Fc), ruthenocenyl (Rc)), have recently been reported as n-dopants for organic semiconductors. Here their structural and energetic characteristics are reported, along with the mechanisms by which they react with acceptors, A (PCBM, TIPS-pentacene), in solution. X-ray data and DFT calculations both indicate a longer C-C bond for (2-Cyc-DMBI)2 than (2-Fc-DMBI)2 , yet DFT and ESR data show that the latter dissociates more readily due to stabilization of the radical by Fc. Depending on the energetics of dimer (D2 ) dissociation and of D2 -to-A electron transfer, D2 reacts with A to form D(+) and A(-) by either of two mechanisms, differing in whether the first step is endergonic dissociation or endergonic electron transfer. However, the D(+) /0.5 D2 redox potentials-the effective reducing strengths of the dimers-vary little within the series (ca. -1.9 V vs. FeCp2 (+/0) ) (Cp=cyclopentadienyl) due to cancelation of trends in the D(+/0) potential and D2 dissociation energy. The implications of these findings for use of these dimers as n-dopants, and for future dopant design, are discussed., (© 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
36. G-protein-independent coupling of MC4R to Kir7.1 in hypothalamic neurons.
- Author
-
Ghamari-Langroudi M, Digby GJ, Sebag JA, Millhauser GL, Palomino R, Matthews R, Gillyard T, Panaro BL, Tough IR, Cox HM, Denton JS, and Cone RD
- Subjects
- Action Potentials, Agouti-Related Protein metabolism, Animals, Eating genetics, Energy Metabolism, Female, HEK293 Cells, Homeostasis genetics, Humans, Ligands, Male, Melanocortins metabolism, Mice, Receptor, Melanocortin, Type 4 genetics, Signal Transduction genetics, alpha-MSH metabolism, GTP-Binding Protein alpha Subunits, Gs, Neurons metabolism, Paraventricular Hypothalamic Nucleus cytology, Potassium Channels, Inwardly Rectifying metabolism, Receptor, Melanocortin, Type 4 metabolism
- Abstract
The regulated release of anorexigenic α-melanocyte stimulating hormone (α-MSH) and orexigenic Agouti-related protein (AgRP) from discrete hypothalamic arcuate neurons onto common target sites in the central nervous system has a fundamental role in the regulation of energy homeostasis. Both peptides bind with high affinity to the melanocortin-4 receptor (MC4R); existing data show that α-MSH is an agonist that couples the receptor to the Gαs signalling pathway, while AgRP binds competitively to block α-MSH binding and blocks the constitutive activity mediated by the ligand-mimetic amino-terminal domain of the receptor. Here we show that, in mice, regulation of firing activity of neurons from the paraventricular nucleus of the hypothalamus (PVN) by α-MSH and AgRP can be mediated independently of Gαs signalling by ligand-induced coupling of MC4R to closure of inwardly rectifying potassium channel, Kir7.1. Furthermore, AgRP is a biased agonist that hyperpolarizes neurons by binding to MC4R and opening Kir7.1, independently of its inhibition of α-MSH binding. Consequently, Kir7.1 signalling appears to be central to melanocortin-mediated regulation of energy homeostasis within the PVN. Coupling of MC4R to Kir7.1 may explain unusual aspects of the control of energy homeostasis by melanocortin signalling, including the gene dosage effect of MC4R and the sustained effects of AgRP on food intake.
- Published
- 2015
- Full Text
- View/download PDF
37. Octarepeat region flexibility impacts prion function, endoproteolysis and disease manifestation.
- Author
-
Lau A, McDonald A, Daude N, Mays CE, Walter ED, Aglietti R, Mercer RC, Wohlgemuth S, van der Merwe J, Yang J, Gapeshina H, Kim C, Grams J, Shi B, Wille H, Balachandran A, Schmitt-Ulms G, Safar JG, Millhauser GL, and Westaway D
- Subjects
- Animals, Cell Line, DNA Mutational Analysis, Disease Models, Animal, Histocytochemistry, Humans, Mice, Transgenic, Microscopy, Mutant Proteins chemistry, Mutant Proteins metabolism, Protein Conformation, Proteolysis, PrPC Proteins chemistry, PrPC Proteins metabolism, Prion Diseases pathology, Prion Diseases physiopathology, Protein Processing, Post-Translational
- Abstract
The cellular prion protein (PrP(C)) comprises a natively unstructured N-terminal domain, including a metal-binding octarepeat region (OR) and a linker, followed by a C-terminal domain that misfolds to form PrP(S) (c) in Creutzfeldt-Jakob disease. PrP(C) β-endoproteolysis to the C2 fragment allows PrP(S) (c) formation, while α-endoproteolysis blocks production. To examine the OR, we used structure-directed design to make novel alleles, 'S1' and 'S3', locking this region in extended or compact conformations, respectively. S1 and S3 PrP resembled WT PrP in supporting peripheral nerve myelination. Prion-infected S1 and S3 transgenic mice both accumulated similar low levels of PrP(S) (c) and infectious prion particles, but differed in their clinical presentation. Unexpectedly, S3 PrP overproduced C2 fragment in the brain by a mechanism distinct from metal-catalysed hydrolysis reported previously. OR flexibility is concluded to impact diverse biological endpoints; it is a salient variable in infectious disease paradigms and modulates how the levels of PrP(S) (c) and infectivity can either uncouple or engage to drive the onset of clinical disease., (© 2015 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2015
- Full Text
- View/download PDF
38. Genetic Incorporation of the Unnatural Amino Acid p-Acetyl Phenylalanine into Proteins for Site-Directed Spin Labeling.
- Author
-
Evans EG and Millhauser GL
- Subjects
- Amino Acids chemistry, Cysteine chemistry, Mutagenesis, Site-Directed, Phenylalanine analogs & derivatives, Phenylalanine chemistry, Protein Structure, Secondary, Electron Spin Resonance Spectroscopy methods, High-Throughput Nucleotide Sequencing methods, Prions chemistry, Spin Labels
- Abstract
Site-directed spin labeling (SDSL) is a powerful tool for the characterization of protein structure and dynamics; however, its application in many systems is hampered by the reliance on unique and benign cysteine substitutions for the site-specific attachment of the spin label. An elegant solution to this problem involves the use of genetically encoded unnatural amino acids (UAAs) containing reactive functional groups that are chemically orthogonal to those of the 20 amino acids found naturally in proteins. These unique functional groups can then be selectively reacted with an appropriately functionalized spin probe. In this chapter, we detail the genetic incorporation of the ketone-bearing amino acid p-acetyl phenylalanine (pAcPhe) into recombinant proteins expressed in E. coli. Incorporation of pAcPhe is followed by chemoselective reaction of the ketone side chain with a hydroxylamine-functionalized nitroxide to afford the spin-labeled side chain "K1," and we present two protocols for successful K1 labeling of proteins bearing site-specific pAcPhe. We outline the basic requirements for pAcPhe incorporation and labeling, with an emphasis on practical aspects that must be considered by the researcher if high yields of UAA incorporation and efficient labeling reactions are to be achieved. To this end, we highlight recent advances that have led to increased yields of pAcPhe incorporation, and discuss the use of aniline-based catalysts allowing for facile conjugation of the hydroxylamine spin label under mild reaction conditions. To illustrate the utility of K1 labeling in proteins where traditional cysteine-based SDSL methods are problematic, we site-specifically K1 label the cellular prion protein at two positions in the C-terminal domain and determine the interspin distance using double electron-electron resonance EPR. Recent advances in UAA incorporation and ketone-based bioconjugation, in combination with the commercial availability of all requisite reagents, should make K1 labeling an increasingly viable alternative to cysteine-based methods for SDSL in proteins., (© 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
39. Copper-zinc cross-modulation in prion protein binding.
- Author
-
Stellato F, Minicozzi V, Millhauser GL, Pascucci M, Proux O, Rossi GC, Spevacek A, and Morante S
- Subjects
- Amino Acid Sequence, Dose-Response Relationship, Drug, Molecular Sequence Data, Peptide Fragments chemistry, Peptide Fragments metabolism, Protein Binding drug effects, Copper pharmacology, Prions chemistry, Prions metabolism, Zinc metabolism
- Abstract
In this paper we report a systematic XAS study of a set of samples in which Cu(II) was progressively added to complexes in which Zn(II) was bound to the tetra-octarepeat portion of the prion protein. This work extends previous EPR and XAS analysis in which, in contrast, the effect of adding Zn(II) to Cu(II)-tetra-octarepeat complexes was investigated. Detailed structural analysis of the XAS spectra taken at both the Cu and Zn K-edge when the two metals are present at different relative concentrations revealed that Zn(II) and Cu(II) ions compete for binding to the tetra-octarepeat peptide by cross-regulating their relative binding modes. We show that the specific metal-peptide coordination mode depends not only, as expected, on the relative metal concentrations, but also on whether Zn(II) or Cu(II) was first bound to the peptide. In particular, it seems that the Zn(II) binding mode in the absence of Cu(II) is able to promote the formation of small peptide clusters in which triplets of tetra-octarepeats are bridged by pairs of Zn ions. When Cu(II) is added, it starts competing with Zn(II) for binding, disrupting the existing peptide cluster arrangement, despite the fact that Cu(II) is unable to completely displace Zn(II). These results may have a bearing on our understanding of peptide-aggregation processes and, with the delicate cross-regulation balancing we have revealed, seem to suggest the existence of an interesting, finely tuned interplay among metal ions affecting protein binding, capable of providing a mechanism for regulation of metal concentration in cells.
- Published
- 2014
- Full Text
- View/download PDF
40. Tyrosine nitration in peptides by peroxynitrite generated in situ in a light-controlled platform: effects of pH and thiols.
- Author
-
deBoer TR, Palomino RI, Idiga SO, Millhauser GL, and Mascharak PK
- Subjects
- Carbon Dioxide chemistry, Glutathione chemistry, Hydrogen-Ion Concentration, Light, Tyrosine chemical synthesis, Peroxynitrous Acid chemistry, Sulfhydryl Compounds chemistry, Tyrosine analogs & derivatives, Tyrosine chemistry
- Abstract
Peroxynitrite has been shown to play a critical role in inflammation and affords 3-nitrotyrosine as the hallmark product. The reported methods of generating this reactive nitrogen species in situ often fails to provide a high and steady flux of peroxynitrite resulting in poor yields of 3-nitrotyrosine. Herein we report a two-component peroxynitrite-generating platform in which this anion is produced in a biomimetic fashion and under the control of visible light. Incorporation of the nitric oxide- and superoxide-generating components in polymer matrices allows easy alterations of pH in the reaction wells of this platform. We have demonstrated very efficient nitration of tyrosine by peroxynitrite at different pH values and with varying concentrations of carbonate. In addition to tyrosine, a set of tyrosine-containing peptides was also studied. Presence of glutathione in the reaction wells increases the extent of tyrosine nitration in such peptide substrates presumably by raising the lifetime of nitric oxide in the reaction medium. When a cysteine residue was included in the sequence of the peptide, the extent of nitration of the tyrosine residue was found to depend on the position of the cysteine residue with respect to tyrosine. The extent of tyrosine nitration is strongly attenuated when the cysteine residue is directly adjacent to the tyrosine. This effect has been attributed to an intramolecular radical transfer mechanism. Taken together, results of this study demonstrate the potential of this light-controlled platform as a convenient bioanalytical tool in studying the reactions of peroxynitrite under widely varying conditions., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
41. Combined EXAFS and DFT structure calculations provide structural insights into the 1:1 multi-histidine complexes of Cu(II) , Cu(I) , and Zn(II) with the tandem octarepeats of the mammalian prion protein.
- Author
-
Pushie MJ, Nienaber KH, McDonald A, Millhauser GL, and George GN
- Subjects
- Amino Acid Sequence, Animals, Histidine chemistry, Humans, Mammals, Models, Molecular, Molecular Sequence Data, X-Ray Absorption Spectroscopy methods, Coordination Complexes chemistry, Copper chemistry, Histidine analogs & derivatives, Prions chemistry, Zinc chemistry
- Abstract
The metal-coordinating properties of the prion protein (PrP) have been the subject of intense focus and debate since the first reports of its interaction with copper just before the turn of the century. The picture of metal coordination to PrP has been improved and refined over the past decade, but structural details of the various metal coordination modes have not been fully elucidated in some cases. In the present study, we have employed X-ray absorption near-edge spectroscopy as well as extended X-ray absorption fine structure (EXAFS) spectroscopy to structurally characterize the dominant 1:1 coordination modes for Cu(II) , Cu(I) , and Zn(II) with an N-terminal fragment of PrP. The PrP fragment corresponds to four tandem repeats representative of the mammalian octarepeat domain, designated as OR4 , which is also the most studied PrP fragment for metal interactions, making our findings applicable to a large body of previous work. Density functional theory (DFT) calculations have provided additional structural and thermodynamic data, and candidate structures have been used to inform EXAFS data analysis. The optimized geometries from DFT calculations have been used to identify potential coordination complexes for multi-histidine coordination of Cu(II) , Cu(I) , and Zn(II) in an aqueous medium, modelled using 4-methylimidazole to represent the histidine side chain. Through a combination of in silico coordination chemistry as well as rigorous EXAFS curve-fitting, using full multiple scattering on candidate structures derived from DFT calculations, we have characterized the predominant coordination modes for the 1:1 complexes of Cu(II) , Cu(I) , and Zn(II) with the OR4 peptide at pH 7.4 at atomic resolution, which are best represented as square-planar [Cu(II) (His)4 ](2+) , digonal [Cu(I) (His)2 ](+) , and tetrahedral [Zn(II) (His)3 (OH2 )](2+) , respectively., (© 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2014
- Full Text
- View/download PDF
42. Adapter reagents for protein site specific dye labeling.
- Author
-
Thompson DA, Evans EG, Kasza T, Millhauser GL, and Dawson PE
- Subjects
- Amino Acid Sequence, Animals, Chromatography, High Pressure Liquid, Chromatography, Reverse-Phase, Electrophoresis, Polyacrylamide Gel, Indicators and Reagents, Mice, Molecular Sequence Data, Peptides chemistry, Coloring Agents metabolism, Prions metabolism, Staining and Labeling
- Abstract
Chemoselective protein labeling remains a significant challenge in chemical biology. Although many selective labeling chemistries have been reported, the practicalities of matching the reaction with appropriately functionalized proteins and labeling reagents is often a challenge. For example, we encountered the challenge of site specifically labeling the cellular form of the murine Prion protein with a fluorescent dye. To facilitate this labeling, a protein was expressed with site specific p-acetylphenylalanine. However, the utility of this acetophenone reactive group is hampered by the severe lack of commercially available aminooxy fluorophores. Here we outline a general strategy for the efficient solid phase synthesis of adapter reagents capable of converting maleimido-labels into aminooxy or azide functional groups that can be further tuned for desired length or solubility properties. The utility of the adapter strategy is demonstrated in the context of fluorescent labeling of the murine Prion protein through an adapted aminooxy-Alexa dye., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
43. PrP overdrive: does inhibition of α-cleavage contribute to PrP(C) toxicity and prion disease?
- Author
-
McDonald AJ and Millhauser GL
- Subjects
- Animals, Mice, Mice, Knockout, PrPC Proteins genetics, PrPC Proteins toxicity, Proteolysis, Zinc metabolism, PrPC Proteins physiology, Prion Diseases metabolism
- Abstract
Knockout of the cellular prion protein (PrP(C)) in mice is tolerated, as is complete elimination of the protein's N-terminal domain. However, deletion of select short segments between the N- and C-terminal domains is lethal. How can one reconcile this apparent paradox? Research over the last few years demonstrates that PrP(C) undergoes α-cleavage in the vicinity of residue 109 (mouse sequence) to release the bioactive N1 and C1 fragments. In biophysical studies, we recently characterized the action of relevant members of the ADAM (A Disintegrin And Metalloproteinase) enzyme family (ADAM8, 10, and 17) and found that they all produce α-cleavage, but at 3 distinct cleavage sites, with proteolytic efficiency modulated by the physiologic metals copper and zinc. Remarkably, the shortest lethal deletion segment in PrP(C) fully encompasses the 3 α-cleavage sites. Analysis of all reported PrP(C) deletion mutants suggests that elimination of α-cleavage, coupled with retention of the protein's N-terminal residues, segments 23-31 and longer, confers the lethal phenotype. Interestingly, these N-terminal residues are implicated in the activation of several membrane proteins, including synaptic glutamate receptors. We propose that α-cleavage is a general mechanism essential for downregulating PrP(C)'s intrinsic activity, and that blockage of proteolysis leads to constitutively active PrP(C) and consequent dyshomeostasis.
- Published
- 2014
- Full Text
- View/download PDF
44. A new paradigm for enzymatic control of α-cleavage and β-cleavage of the prion protein.
- Author
-
McDonald AJ, Dibble JP, Evans EG, and Millhauser GL
- Subjects
- ADAM Proteins genetics, ADAM10 Protein, ADAM17 Protein, Amyloid Precursor Protein Secretases genetics, Animals, Chromatography, Liquid, Copper chemistry, Copper metabolism, Humans, Mass Spectrometry, Membrane Proteins genetics, Mice, Models, Molecular, Oxidation-Reduction, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, PrPC Proteins chemistry, PrPC Proteins genetics, Protein Structure, Tertiary, Proteolysis, Reactive Oxygen Species metabolism, Recombinant Proteins metabolism, Zinc chemistry, Zinc metabolism, ADAM Proteins metabolism, Amyloid Precursor Protein Secretases metabolism, Membrane Proteins metabolism, PrPC Proteins metabolism
- Abstract
The cellular form of the prion protein (PrP(C)) is found in both full-length and several different cleaved forms in vivo. Although the precise functions of the PrP(C) proteolytic products are not known, cleavage between the unstructured N-terminal domain and the structured C-terminal domain at Lys-109↓His-110 (mouse sequence), termed α-cleavage, has been shown to produce the anti-apoptotic N1 and the scrapie-resistant C1 peptide fragments. β-Cleavage, residing adjacent to the octarepeat domain and N-terminal to the α-cleavage site, is thought to arise from the action of reactive oxygen species produced from redox cycling of coordinated copper. We sought to elucidate the role of key members of the ADAM (a disintegrin and metalloproteinase) enzyme family, as well as Cu(2+) redox cycling, in recombinant mouse PrP (MoPrP) cleavage through LC/MS analysis. Our findings show that although Cu(2+) redox-generated reactive oxygen species do produce fragmentation corresponding to β-cleavage, ADAM8 also cleaves MoPrP in the octarepeat domain in a Cu(2+)- and Zn(2+)-dependent manner. Additional cleavage by ADAM8 was observed at the previously proposed location of α-cleavage, Lys-109↓His-110 (MoPrP sequencing); however, upon addition of Cu(2+), the location of α-cleavage shifted by several amino acids toward the C terminus. ADAM10 and ADAM17 have also been implicated in α-cleavage at Lys-109↓His-110; however, we observed that they instead cleaved MoPrP at a novel location, Ala-119↓Val-120, with additional cleavage by ADAM10 at Gly-227↓Arg-228 near the C terminus. Together, our results show that MoPrP cleavage is far more complex than previously thought and suggest a mechanism by which PrP(C) fragmentation responds to Cu(2+) and Zn(2+).
- Published
- 2014
- Full Text
- View/download PDF
45. New insights into metal interactions with the prion protein: EXAFS analysis and structure calculations of copper binding to a single octarepeat from the prion protein.
- Author
-
McDonald A, Pushie MJ, Millhauser GL, and George GN
- Subjects
- Amino Acid Sequence, Copper chemistry, Electron Spin Resonance Spectroscopy, Hydrogen-Ion Concentration, Peptides chemistry, Peptides metabolism, Prions chemistry, Protein Binding, Protein Structure, Tertiary, Thermodynamics, X-Ray Absorption Spectroscopy, Copper metabolism, Prions metabolism
- Abstract
Copper coordination to the prion protein (PrP) has garnered considerable interest for almost 20 years, due in part to the possibility that this interaction may be part of the normal function of PrP. The most characterized form of copper binding to PrP has been Cu(2+) interaction with the conserved tandem repeats in the N-terminal domain of PrP, termed the octarepeats, with many studies focusing on single and multiple repeats of PHGGGWGQ. Extended X-ray absorption fine structure (EXAFS) spectroscopy has been used in several previous instances to characterize the solution structure of Cu(2+) binding into the peptide backbone in the HGGG portion of the octarepeats. All previous EXAFS studies, however, have benefitted from crystallographic structure information for [Cu(II) (Ac-HGGGW-NH2)(-2H)] but have not conclusively demonstrated that the complex EXAFS spectrum represents the same coordination environment for Cu(2+) bound to the peptide backbone. Density functional structure calculations as well as full multiple scattering EXAFS curve fitting analysis are brought to bear on the predominant coordination mode for Cu(2+) with the Ac-PHGGGWGQ-NH2 peptide at physiological pH, under high Cu(2+) occupancy conditions. In addition to the structure calculations, which provide a thermodynamic link to structural information, methods are also presented for extensive deconvolution of the EXAFS spectrum. We demonstrate how the EXAFS data can be analyzed to extract the maximum structural information and arrive at a structural model that is significantly improved over previous EXAFS characterizations. The EXAFS spectrum for the chemically reduced form of copper binding to the Ac-PHGGGWGQ-NH2 peptide is presented, which is best modeled as a linear two-coordinate species with a single His imidazole ligand and a water molecule. The extent of in situ photoreduction of the copper center during standard data collection is also presented, and EXAFS curve fitting of the photoreduced species reveals an intermediate structure that is similar to the Cu(2+) form with reduced coordination number.
- Published
- 2013
- Full Text
- View/download PDF
46. Mechanistic study on the solution-phase n-doping of 1,3-dimethyl-2-aryl-2,3-dihydro-1H-benzoimidazole derivatives.
- Author
-
Naab BD, Guo S, Olthof S, Evans EG, Wei P, Millhauser GL, Kahn A, Barlow S, Marder SR, and Bao Z
- Subjects
- Electron Transport, Kinetics, Solutions, Thermodynamics, Imidazoles chemistry
- Abstract
The discovery of air-stable n-dopants for organic semiconductor materials has been hindered by the necessity of high-energy HOMOs and the air sensitivity of compounds that satisfy this requirement. One strategy for circumventing this problem is to utilize stable precursor molecules that form the active doping complex in situ during the doping process or in a postdeposition thermal- or photo-activation step. Some of us have reported on the use of 1H-benzimidazole (DMBI) and benzimidazolium (DMBI-I) salts as solution- and vacuum-processable n-type dopant precursors, respectively. It was initially suggested that DMBI dopants function as single-electron radical donors wherein the active doping species, the imidazoline radical, is generated in a postdeposition thermal annealing step. Herein we report the results of extensive mechanistic studies on DMBI-doped fullerenes, the results of which suggest a more complicated doping mechanism is operative. Specifically, a reaction between the dopant and host that begins with either hydride or hydrogen atom transfer and which ultimately leads to the formation of host radical anions is responsible for the doping effect. The results of this research will be useful for identifying applications of current organic n-doping technology and will drive the design of next-generation n-type dopants that are air stable and capable of doping low-electron-affinity host materials in organic devices.
- Published
- 2013
- Full Text
- View/download PDF
47. Molecular and functional analysis of human β-defensin 3 action at melanocortin receptors.
- Author
-
Nix MA, Kaelin CB, Ta T, Weis A, Morton GJ, Barsh GS, and Millhauser GL
- Subjects
- Agouti Signaling Protein agonists, Agouti Signaling Protein genetics, Agouti Signaling Protein metabolism, Agouti-Related Protein agonists, Agouti-Related Protein metabolism, Amino Acid Sequence, Animals, Humans, Ligands, Mice, Mice, Inbred C57BL, Mice, Transgenic, Molecular Sequence Data, Mutation, Protein Binding, Protein Structure, Tertiary, Receptor, Melanocortin, Type 1 antagonists & inhibitors, Receptor, Melanocortin, Type 1 genetics, Receptor, Melanocortin, Type 4 chemistry, Receptor, Melanocortin, Type 4 genetics, Receptor, Melanocortin, Type 4 metabolism, Static Electricity, beta-Defensins genetics, Receptor, Melanocortin, Type 1 metabolism, beta-Defensins metabolism
- Abstract
The β-defensins are a class of small, cationic proteins first recognized as antimicrobial components of the innate and adaptive immune system. More recently, one of the major β-defensins produced in skin, β-defensin 3, has been discovered to function as a melanocortin receptor ligand in vivo and in vitro, but its biophysical and pharmacological basis of action has been enigmatic. Here, we report functional and biochemical studies focused on human β-defensin 3 (HBD3) and melanocortin receptors 1 and 4. Genetic and pharmacologic studies indicate that HBD3 acts as a neutral melanocortin receptor antagonist capable of blocking the action of either stimulatory agonists such as α-melanocyte stimulating hormone or inhibitory inverse agonists such as Agouti signaling protein (ASIP) and Agouti-related protein (AGRP). A comprehensive structure-function analysis demonstrates that two patches of positively charged residues, located on opposite poles of HBD3 and spatially organized by the compact β-defensin fold, are primarily responsible for high-affinity binding to melanocortin receptors. These findings identify a distinct mode of melanocortin receptor-ligand interactions based primarily on electrostatic complementarity, with implications for designing ligands that target melanocortin and potentially other seven transmembrane receptors., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
48. A novel α-synuclein missense mutation in Parkinson disease.
- Author
-
Proukakis C, Dudzik CG, Brier T, MacKay DS, Cooper JM, Millhauser GL, Houlden H, and Schapira AH
- Subjects
- Aged, 80 and over, Amino Acid Sequence, Amino Acid Substitution genetics, Fatal Outcome, Female, Humans, Molecular Sequence Data, Mutation, Missense genetics, Parkinson Disease diagnosis, Parkinson Disease genetics, alpha-Synuclein genetics
- Abstract
Alpha-synuclein (SNCA) is central to the pathogenesis of Parkinson disease (PD), with 3 missense mutations reported to date. We report a novel mutation (p.H50Q) in a pathologically proven case.
- Published
- 2013
- Full Text
- View/download PDF
49. Zinc drives a tertiary fold in the prion protein with familial disease mutation sites at the interface.
- Author
-
Spevacek AR, Evans EG, Miller JL, Meyer HC, Pelton JG, and Millhauser GL
- Subjects
- Amino Acid Sequence, Animals, Electron Spin Resonance Spectroscopy, Mice, Molecular Dynamics Simulation, Molecular Sequence Data, PrPC Proteins genetics, Protein Binding, Protein Interaction Domains and Motifs, Protein Structure, Secondary, Mutation, Missense, PrPC Proteins chemistry, Zinc chemistry
- Abstract
The cellular prion protein PrP(C) consists of two domains--a flexible N-terminal domain, which participates in copper and zinc regulation, and a largely helical C-terminal domain that converts to β sheet in the course of prion disease. These two domains are thought to be fully independent and noninteracting. Compelling cellular and biophysical studies, however, suggest a higher order structure that is relevant to both PrP(C) function and misfolding in disease. Here, we identify a Zn²⁺-driven N-terminal to C-terminal tertiary interaction in PrP(C). The C-terminal surface participating in this interaction carries the majority of the point mutations that confer familial prion disease. Investigation of mutant PrPs finds a systematic relationship between the type of mutation and the apparent strength of this domain structure. The structural features identified here suggest mechanisms by which physiologic metal ions trigger PrP(C) trafficking and control prion disease., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
50. The elevated copper binding strength of amyloid-β aggregates allows the sequestration of copper from albumin: a pathway to accumulation of copper in senile plaques.
- Author
-
Jiang D, Zhang L, Grant GP, Dudzik CG, Chen S, Patel S, Hao Y, Millhauser GL, and Zhou F
- Subjects
- Amino Acid Substitution, Amyloid beta-Peptides chemistry, Amyloid beta-Peptides genetics, Chromatography, Gel, Copper analysis, Copper chemistry, Electron Spin Resonance Spectroscopy, Histidine chemistry, Histidine metabolism, Humans, Kinetics, Microscopy, Atomic Force, Models, Molecular, Molecular Weight, Mutant Proteins chemistry, Mutant Proteins metabolism, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Plaque, Amyloid chemistry, Protein Denaturation, Serum Albumin, Human, Spectrometry, Fluorescence, Tryptophan chemistry, Tryptophan metabolism, Amyloid beta-Peptides metabolism, Copper metabolism, Plaque, Amyloid metabolism, Serum Albumin metabolism
- Abstract
Copper coexists with amyloid-β (Aβ) peptides at a high concentration in the senile plaques of Alzheimer's disease (AD) patients and has been linked to oxidative damage associated with AD pathology. However, the origin of copper and the driving force behind its accumulation are unknown. We designed a sensitive fluorescent probe, Aβ(1-16)(Y10W), by substituting the tyrosine residue at position 10 in the hydrophilic domain of Aβ(1-42) with tryptophan. Upon mixing Cu(II), Aβ(1-16)(Y10W), and aliquots of Aβ(1-42) taken from samples incubated for different lengths of time, we found that the Cu(II) binding strength of aggregated Aβ(1-42) has been elevated by more than 2 orders of magnitude with respect to that of monomeric Aβ(1-42). Electron paramagnetic spectroscopic measurements revealed that the Aβ(1-42) aggregates, unlike their monomeric form, can seize copper from human serum albumin, an abundant copper-containing protein in brain and cerebrospinal fluid. The significantly elevated binding strength of the Aβ(1-42) aggregates can be rationalized by a Cu(II) coordination sphere constituted by three histidines from two adjacent Aβ(1-42) molecules. Our work demonstrates that the copper binding affinity of Aβ(1-42) is dependent on its aggregation state and provides new insight into how and why senile plaques accumulate copper in vivo.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.