Network


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

Hotspot


Dive into the research topics where Peter A. Doucette is active.

Publication


Featured researches published by Peter A. Doucette.


Nature Structural & Molecular Biology | 2003

Amyloid-Like Filaments and Water-Filled Nanotubes Formed by Sod1 Mutant Proteins Linked to Familial Als

Jennifer Stine Elam; Alexander B. Taylor; Richard W. Strange; Svetlana V. Antonyuk; Peter A. Doucette; Jorge A. Rodriguez; S. Samar Hasnain; Lawrence J. Hayward; Joan Selverstone Valentine; Todd O. Yeates; P. John Hart

Mutations in the SOD1 gene cause the autosomal dominant, neurodegenerative disorder familial amyotrophic lateral sclerosis (FALS). In spinal cord neurons of human FALS patients and in transgenic mice expressing these mutant proteins, aggregates containing FALS SOD1 are observed. Accumulation of SOD1 aggregates is believed to interfere with axonal transport, protein degradation and anti-apoptotic functions of the neuronal cellular machinery. Here we show that metal-deficient, pathogenic SOD1 mutant proteins crystallize in three different crystal forms, all of which reveal higher-order assemblies of aligned β-sheets. Amyloid-like filaments and water-filled nanotubes arise through extensive interactions between loop and β-barrel elements of neighboring mutant SOD1 molecules. In all cases, non-native conformational changes permit a gain of interaction between dimers that leads to higher-order arrays. Normal β-sheet–containing proteins avoid such self-association by preventing their edge strands from making intermolecular interactions. Loss of this protection through conformational rearrangement in the metal-deficient enzyme could be a toxic property common to mutants of SOD1 linked to FALS.


PLOS ONE | 2009

Loss of Metal Ions, Disulfide Reduction and Mutations Related to Familial ALS Promote Formation of Amyloid-Like Aggregates from Superoxide Dismutase

Zeynep A. Oztug Durer; Jeffrey A. Cohlberg; Phong Dinh; Shelby Padua; Krista Ehrenclou; Sean Downes; James K. Tan; Yoko Nakano; Christopher J. Bowman; Jessica L. Hoskins; Chuhee Kwon; Andrew Z. Mason; Jorge A. Rodriguez; Peter A. Doucette; Bryan F. Shaw; Joan Selverstone Valentine

Mutations in the gene encoding Cu-Zn superoxide dismutase (SOD1) are one of the causes of familial amyotrophic lateral sclerosis (FALS). Fibrillar inclusions containing SOD1 and SOD1 inclusions that bind the amyloid-specific dye thioflavin S have been found in neurons of transgenic mice expressing mutant SOD1. Therefore, the formation of amyloid fibrils from human SOD1 was investigated. When agitated at acidic pH in the presence of low concentrations of guanidine or acetonitrile, metalated SOD1 formed fibrillar material which bound both thioflavin T and Congo red and had circular dichroism and infrared spectra characteristic of amyloid. While metalated SOD1 did not form amyloid-like aggregates at neutral pH, either removing metals from SOD1 with its intramolecular disulfide bond intact or reducing the intramolecular disulfide bond of metalated SOD1 was sufficient to promote formation of these aggregates. SOD1 formed amyloid-like aggregates both with and without intermolecular disulfide bonds, depending on the incubation conditions, and a mutant SOD1 lacking free sulfhydryl groups (AS-SOD1) formed amyloid-like aggregates at neutral pH under reducing conditions. ALS mutations enhanced the ability of disulfide-reduced SOD1 to form amyloid-like aggregates, and apo-AS-SOD1 formed amyloid-like aggregates at pH 7 only when an ALS mutation was also present. These results indicate that some mutations related to ALS promote formation of amyloid-like aggregates by facilitating the loss of metals and/or by making the intramolecular disulfide bond more susceptible to reduction, thus allowing the conversion of SOD1 to a form that aggregates to form resembling amyloid. Furthermore, the occurrence of amyloid-like aggregates per se does not depend on forming intermolecular disulfide bonds, and multiple forms of such aggregates can be produced from SOD1.


Journal of Biological Chemistry | 2008

Structures of the G85R Variant of SOD1 in Familial Amyotrophic Lateral Sclerosis.

Xiaohang Cao; Svetlana V. Antonyuk; Sai V. Seetharaman; Lisa J. Whitson; Alexander B. Taylor; Stephen P. Holloway; Richard W. Strange; Peter A. Doucette; Joan Selverstone Valentine; Ashutosh Tiwari; Lawrence J. Hayward; Shelby Padua; Jeffrey A. Cohlberg; S. Samar Hasnain; P. John Hart

Mutations in the gene encoding human copper-zinc superoxide dismutase (SOD1) cause a dominant form of the progressive neurodegenerative disease amyotrophic lateral sclerosis. Transgenic mice expressing the human G85R SOD1 variant develop paralytic symptoms concomitant with the appearance of SOD1-enriched proteinaceous inclusions in their neural tissues. The process(es) through which misfolding or aggregation of G85R SOD1 induces motor neuron toxicity is not understood. Here we present structures of the human G85R SOD1 variant determined by single crystal x-ray diffraction. Alterations in structure of the metal-binding loop elements relative to the wild type enzyme suggest a molecular basis for the metal ion deficiency of the G85R SOD1 protein observed in the central nervous system of transgenic mice and in purified recombinant G85R SOD1. These findings support the notion that metal-deficient and/or disulfide-reduced mutant SOD1 species contribute to toxicity in SOD1-linked amyotrophic lateral sclerosis.


Protein Science | 2005

Structural consequences of the familial amyotrophic lateral sclerosis SOD1 mutant His46Arg

Svetlana V. Antonyuk; Jennifer Stine Elam; Michael A. Hough; Richard W. Strange; Peter A. Doucette; Jorge A. Rodriguez; Lawrence J. Hayward; Joan Selverstone Valentine; P. John Hart; S. Samar Hasnain

The His46Arg (H46R) mutant of human copper‐zinc superoxide dismutase (SOD1) is associated with an unusual, slowly progressing form of familial amyotrophic lateral sclerosis (FALS). Here we describe in detail the crystal structures of pathogenic H46R SOD1 in the Zn‐loaded (Zn‐H46R) and metal‐free (apo‐H46R) forms. The Zn‐H46R structure demonstrates a novel zinc coordination that involves only three of the usual four liganding residues, His 63, His 80, and Asp 83 together with a water molecule. In addition, the Asp 124 “secondary bridge” between the copper‐ and zinc‐binding sites is disrupted, and the “electrostatic loop” and “zinc loop” elements are largely disordered. The apo‐H46R structure exhibits partial disorder in the electrostatic and zinc loop elements in three of the four dimers in the asymmetric unit, while the fourth has ordered loops due to crystal packing interactions. In both structures, nonnative SOD1–SOD1 interactions lead to the formation of higher‐order filamentous arrays. The disordered loop elements may increase the likelihood of protein aggregation in vivo, either with other H46R molecules or with other critical cellular components. Importantly, the binding of zinc is not sufficient to prevent the formation of nonnative interactions between pathogenic H46R molecules. The increased tendency to aggregate, even in the presence of Zn, arising from the loss of the secondary bridge is consistent with the observation of an increased abundance of hyaline inclusions in spinal motor neurons and supporting cells in H46R SOD1 transgenic rats.


Biochemistry | 2010

Disrupted zinc-binding sites in structures of pathogenic SOD1 variants D124V and H80R.

Sai V. Seetharaman; Duane D. Winkler; Alexander B. Taylor; Xiaohang Cao; Lisa J. Whitson; Peter A. Doucette; Joan Selverstone Valentine; Virgil Schirf; Borries Demeler; Mark C. Carroll; Valeria C. Culotta; P. John Hart

Mutations in human copper-zinc superoxide dismutase (SOD1) cause an inherited form of the fatal neurodegenerative disease amyotrophic lateral sclerosis (ALS). Here, we present structures of the pathogenic SOD1 variants D124V and H80R, both of which demonstrate compromised zinc-binding sites. The disruption of the zinc-binding sites in H80R SOD1 leads to conformational changes in loop elements, permitting non-native SOD1-SOD1 interactions that mediate the assembly of these proteins into higher-order filamentous arrays. Analytical ultracentrifugation sedimentation velocity experiments indicate that these SOD1 variants are more prone to monomerization than the wild-type enzyme. Although D124V and H80R SOD1 proteins appear to have fully functional copper-binding sites, inductively coupled plasma mass spectrometery (ICP-MS) and anomalous scattering X-ray diffraction analyses reveal that zinc (not copper) occupies the copper-binding sites in these variants. The absence of copper in these proteins, together with the results of covalent thiol modification experiments in yeast strains with and without the gene encoding the copper chaperone for SOD1 (CCS), suggests that CCS may not fully act on newly translated forms of these polypeptides. Overall, these findings lend support to the hypothesis that immature mutant SOD1 species contribute to toxicity in SOD1-linked ALS.


Journal of Inorganic Biochemistry | 2003

Misfolded superoxide dismutase and ALS

P. John Hart; Jennifer Stine Elam; Alexander B. Taylor; Richard W. Strange; S. Samar Hasnain; Peter A. Doucette; Joan Selverstone Valentine; Lawrence J. Hayward

P John Hart, University of Texas Health Science Center San Antonio, United States Jennifer Stine Elam, University of Texas Health Science Center San Antonio, UnitedStates Alexander B Taylor, University of Texas Health Science Center San Antonio, United States Richard Strange, CLRC Daresbury, United Kingdom S Samar Hasnain, CLRC Daresbzary, United Kingdom Peter A Doucette, University of California, Los Angeles, United States Joan S Valentine, Vniversi& of California, Los Angeles, United States Lawrence J Hayward, University of Massachusetts Medical School, United States


Annual Review of Biochemistry | 2005

COPPER-ZINC SUPEROXIDE DISMUTASE AND AMYOTROPHIC LATERAL SCLEROSIS

Joan Selverstone Valentine; Peter A. Doucette; Soshanna Zittin Potter


Proceedings of the National Academy of Sciences of the United States of America | 2004

Dimer destabilization in superoxide dismutase may result in disease-causing properties: Structures of motor neuron disease mutants

Michael A. Hough; J. Günter Grossmann; Svetlana V. Antonyuk; Richard W. Strange; Peter A. Doucette; Jorge A. Rodriguez; Lisa J. Whitson; P. John Hart; Lawrence J. Hayward; Joan Selverstone Valentine; S. Samar Hasnain


Journal of Molecular Biology | 2003

The Structure of Holo and Metal-Deficient Wild-Type Human Cu, Zn Superoxide Dismutase and its Relevance to Familial Amyotrophic Lateral Sclerosis

Richard W. Strange; Svetlana V. Antonyuk; Michael A. Hough; Peter A. Doucette; Jorge A. Rodriguez; P. John Hart; Lawrence J. Hayward; Joan Selverstone Valentine; S. Samar Hasnain


Proceedings of the National Academy of Sciences of the United States of America | 2005

Destabilization of apoprotein is insufficient to explain Cu,Zn-superoxide dismutase-linked ALS pathogenesis

Jorge A. Rodriguez; Bryan F. Shaw; Armando Durazo; Se Hui Sohn; Peter A. Doucette; Aram M. Nersissian; Kym F. Faull; Daryl K. Eggers; Ashutosh Tiwari; Lawrence J. Hayward; Joan Selverstone Valentine

Collaboration


Dive into the Peter A. Doucette's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

P. John Hart

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lawrence J. Hayward

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander B. Taylor

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Jennifer Stine Elam

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Lisa J. Whitson

University of Texas Health Science Center at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge