Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
MAbs ; 16(1): 2324801, 2024.
Article in English | MEDLINE | ID: mdl-38441119

ABSTRACT

Biologics have revolutionized disease management in many therapeutic areas by addressing unmet medical needs and overcoming resistance to standard-of-care treatment in numerous patients. However, the development of unwanted immune responses directed against these drugs, humoral and/or cellular, can hinder their efficacy and have safety consequences with various degrees of severity. Health authorities ask that a thorough immunogenicity risk assessment be conducted during drug development to incorporate an appropriate monitoring and mitigation plan in clinical studies. With the rapid diversification and complexification of biologics, which today include modalities such as multi-domain antibodies, cell-based products, AAV delivery vectors, and nucleic acids, developers are faced with the challenge of establishing a risk assessment strategy sometimes in the absence of specific regulatory guidelines. The European Immunogenicity Platform (EIP) Open Symposium on Immunogenicity of Biopharmaceuticals and its one-day training course gives experts and newcomers across academia, industry, and regulatory agencies an opportunity to share experience and knowledge to overcome these challenges. Here, we report the discussions that took place at the EIP's 14th Symposium, held in April 2023. The topics covered included immunogenicity monitoring and clinical relevance, non-clinical immunogenicity risk assessment, regulatory aspects of immunogenicity assessment and reporting, and the challenges associated with new modalities, which were discussed in a dedicated session.


Subject(s)
Biological Products , Humans , Antibodies , Drug Development , Risk Assessment
2.
MAbs ; 14(1): 2143009, 2022.
Article in English | MEDLINE | ID: mdl-36394299

ABSTRACT

ABBREVIATIONS: ADA Anti-Drug Antibodies; BCR B Cell Receptor; BId Idiotype-specific B Cell; BiTE Bispecific T cell Engager; BMC Bone Marrow Chimeric Mice; BSA Bovine Serum Albumin; CDR Complementary Determining Region; CEA Carcinoembryonic Antigen; CIT Cancer Immunotherapy; CitAbs Cancer Immunotherapy Antibodies; DC Dendritic Cell; ELISA Enzyme-Linked Immunosorbent Assay; FcRn Neonatal Fc Receptor; FcyR Fc gamma Receptor; GM-CSF Granulocyte-Macrophage Colony Stimulating Factor; gMFI Geometric Mean Fluorescence Intensity; H Heavy Chain; IC Immune Complex; Id Idiotype; IgA Immunoglobulin alpha; IgG1 Immunoglobulin gamma 1; IL-2 Interleukin 2; IL-2R Interleukin 2 Receptor; IL2v Interleukin 2 Variant; IVIG1 Intravenous Immunoglobulin 1; KLH Keyhole Limpet Hemocyanin; L Light Chain; MAPPs MHC-associated Peptide Proteomics; MHC Major Histocompatibility Complex; PBMC Peripheral Blood Mononuclear Cells; PBS Phosphate Buffered Saline; SHM Somatic Hypermutation; scFv Single-chain Variable Fragment; TCR T cell Receptor; TFc Fc-specific T cell; TId Id-specific T cell; UV Ultraviolet; V Variable.


Subject(s)
Immunoglobulin G , Neoplasms , Humans , Mice , Animals , Interleukin-2 , Mice, Transgenic , Leukocytes, Mononuclear , Immunotherapy
3.
AAPS J ; 24(3): 68, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35554731

ABSTRACT

The determination of a tailored anti-drug antibody (ADA) testing strategy is based on the immunogenicity risk assessment to allow a correlation of ADAs with changes to pharmacokinetics, efficacy, and safety. The clinical impact of ADA formation refines the immunogenicity risk assessment and defines appropriate risk mitigation strategies. Health agencies request for high-risk biotherapeutics to extend ADA monitoring for patients that developed an ADA response to the drug until ADAs return to baseline levels. However, there is no common understanding in which cases an extension of ADA follow-up sampling beyond the end of study (EOS) defined in the clinical study protocol is required. Here, the Immunogenicity Strategy Working Group of the European Immunogenicity Platform (EIP) provides recommendations on requirements for an extension of ADA follow-up sampling in clinical studies where there is a high risk of serious consequences from ADAs. The importance of ADA evaluation during a treatment-free period is recognized but the decision whether to extend ADA monitoring at a predefined EOS should be based on evaluation of ADA data in the context of corresponding clinical signals. If the clinical data set shows that safety consequences are minor, mitigated, or resolved, further ADA monitoring may not be required despite potentially detectable ADAs above baseline. Extended ADA monitoring should be centered on individual patient benefit.


Subject(s)
Antibodies , Humans
4.
Bioanalysis ; 14(11): 737-793, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35578991

ABSTRACT

The 15th edition of the Workshop on Recent Issues in Bioanalysis (15th WRIB) was held on 27 September to 1 October 2021. Even with a last-minute move from in-person to virtual, an overwhelmingly high number of nearly 900 professionals representing pharma and biotech companies, contract research organizations (CROs), and multiple regulatory agencies still eagerly convened to actively discuss the most current topics of interest in bioanalysis. The 15th WRIB included 3 Main Workshops and 7 Specialized Workshops that together spanned 1 week in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on biomarker assay development and validation (BAV) (focused on clarifying the confusion created by the increased use of the term "Context of Use - COU"); mass spectrometry of proteins (therapeutic, biomarker and transgene); state-of-the-art cytometry innovation and validation; and, critical reagent and positive control generation were the special features of the 15th edition. This 2021 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop, and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2021 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 3) covers the recommendations on TAb/NAb, Viral Vector CDx, Shedding Assays; CRISPR/Cas9 & CAR-T Immunogenicity; PCR & Vaccine Assay Performance; ADA Assay Comparability & Cut Point Appropriateness. Part 1A (Endogenous Compounds, Small Molecules, Complex Methods, Regulated Mass Spec of Large Molecules, Small Molecule, PoC), Part 1B (Regulatory Agencies' Inputs on Bioanalysis, Biomarkers, Immunogenicity, Gene & Cell Therapy and Vaccine) and Part 2 (ISR for Biomarkers, Liquid Biopsies, Spectral Cytometry, Inhalation/Oral & Multispecific Biotherapeutics, Accuracy/LLOQ for Flow Cytometry) are published in volume 14 of Bioanalysis, issues 9 and 10 (2022), respectively.


Subject(s)
Receptors, Chimeric Antigen , Vaccines , Biomarkers/analysis , CRISPR-Cas Systems , Cell- and Tissue-Based Therapy , Humans , Immunotherapy, Active , Polymerase Chain Reaction
5.
Bioanalysis ; 12(15): 1043-1048, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32803994

ABSTRACT

Given the expanding number of complex therapeutic protein drugs and advanced therapy medicinal products that are being developed, improving our ability to assess the potential immunogenicity of biologics is critical to ensuring treatment efficacy and patient safety. In this context, the European Immunogenicity Platform annual meeting provides opportunities for experts from industry and academia, regulators and clinicians to convene and discuss immunogenicity assessment methods and tools. This report summarizes the key messages on immunogenicity testing, prediction, clinical relevance and advanced therapy medicinal products discussed at the 11th Open Scientific European Immunogenicity Platform Symposium on Immunogenicity of Biopharmaceuticals, Lisbon, Portugal, 17-19 February 2020.


Subject(s)
Biopharmaceutics/methods , Immunogenetics/methods , Europe , Humans
6.
Methods Mol Biol ; 1017: 163-71, 2013.
Article in English | MEDLINE | ID: mdl-23719915

ABSTRACT

A pathological hallmark of many protein-misfolding diseases is the formation of insoluble aggregates. Quantitative methods are needed to better resolve and define the formation, aggregation, and temporal dynamics of soluble misfolded proteins in native settings. In this book chapter we describe simple and sensitive detection methods to characterize high ordered aggregates (AGERA) and subsets of distinct soluble aggregates (SEC-FRET) of mutant huntingtin protein in biological samples.


Subject(s)
Mutation , Nerve Tissue Proteins , Nuclear Proteins , Proteostasis Deficiencies , Animals , Disease Models, Animal , Humans , Huntingtin Protein , Immunoassay/methods , Mice , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology
7.
J Mol Med (Berl) ; 91(6): 653-64, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23529761

ABSTRACT

A common pathological hallmark in many neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease, is the formation of fibrillar protein aggregates referred to as amyloids. The amyloidogenic aggregates were long thought to be toxic, but mounting evidence supports the notion that a variety of amyloid aggregate intermediates to fibril formation, termed oligomers, may in fact be the primary culprit leading to neuronal dysfunction and cell death. While amyloid formation is a complex, heterogeneous process, aggregates formed by diverse, diseases-related proteins share many conformational similarities, suggesting common toxic mechanisms among these diseases. Ideally, similar therapeutic strategies may be applicable. This review focuses on the potential role of amyloidogenic oligomers in neurodegenerative disease, highlighting some promising therapeutic strategies.


Subject(s)
Amyloidogenic Proteins/metabolism , Neurodegenerative Diseases/metabolism , Amyloidogenic Proteins/chemistry , Animals , Humans , Membrane Lipids , Molecular Chaperones , Protein Conformation , RNA Interference
8.
Neurotherapeutics ; 10(3): 416-28, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23536253

ABSTRACT

A common pathological hallmark of protein-conformational brain diseases is the formation of disease-specific protein aggregates. In Alzheimer's disease, these are comprised of amyloid-ß and Tau as opposed to α-synuclein in Parkinson's disease and N-terminal fragments of mutant huntingtin in Huntington's disease. Most aggregates also sequester molecular chaperones, a protein family that assists in the folding, refolding, stabilization, and processing of client proteins, including misfolded proteins in brain diseases. Molecular chaperone modulation has achieved remarkable therapeutic effects in some cellular and preclinical animal models of protein-conformational diseases. This has raised hope for chaperone-based strategies to combat these diseases. Here, we review briefly the functional diversity and medical significance of molecular chaperones, their therapeutic potential, and common and specific challenges towards clinical application.


Subject(s)
Brain/metabolism , Molecular Chaperones/metabolism , Molecular Chaperones/therapeutic use , Proteostasis Deficiencies/therapy , Animals , Humans , Proteostasis Deficiencies/pathology
9.
PLoS One ; 7(9): e44457, 2012.
Article in English | MEDLINE | ID: mdl-22984513

ABSTRACT

Cleavage of the full-length mutant huntingtin (mhtt) protein into smaller, soluble aggregation-prone mhtt fragments appears to be a key process in the neuropathophysiology of Huntington's Disease (HD). Recent quantification studies using TR-FRET-based immunoassays showed decreasing levels of soluble mhtt correlating with an increased load of aggregated mhtt in the aging HdhQ150 mouse brain. To better characterize the nature of these changes at the level of native mhtt species, we developed a detection method that combines size exclusion chromatography (SEC) and time-resolved fluorescence resonance energy transfer (TR-FRET) that allowed us to resolve and define the formation, aggregation and temporal dynamics of native soluble mhtt species and insoluble aggregates in the brain of the HdhQ150 knock-in mouse. We found that mhtt fragments and not full-length mhtt form oligomers in the brains of one month-old mice long before disease phenotypes and mhtt aggregate histopathology occur. As the HdhQ150 mice age, brain levels of soluble full-length mhtt protein remain similar. In contrast, the soluble oligomeric pool of mhtt fragments slightly increases during the first two months before it declines between 3 and 8 months of age. This decline inversely correlates with the formation of insoluble mhtt aggregates. We also found that the pool-size of soluble mhtt oligomers is similar in age-matched heterozygous and homozygous HdhQ150 mouse brains whereas insoluble aggregate formation is greatly accelerated in the homozygous mutant brain. The capacity of the soluble mhtt oligomer pool therefore seems exhausted already in the heterozygous state and likely kept constant by changes in flux and, as a consequence, increased rate of insoluble aggregate formation. We demonstrate that our novel findings in mice translate to human HD brain but not HD patient fibroblasts.


Subject(s)
Aging , Huntington Disease/genetics , Mutation , Nerve Tissue Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Animals , Brain/metabolism , Brain/pathology , Chromatography/methods , Disease Models, Animal , Embryonic Stem Cells/cytology , Fibroblasts/metabolism , Fluorescence Resonance Energy Transfer/methods , Humans , Huntingtin Protein , Huntington Disease/metabolism , Mice , Models, Biological , Protein Binding , Serotonin Plasma Membrane Transport Proteins/physiology
10.
J Neurosci ; 32(32): 11109-19, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22875942

ABSTRACT

Huntington's disease (HD) is a devastating neurodegenerative disorder with no disease-modifying treatments available. The disease is caused by expansion of a CAG trinucleotide repeat and manifests with progressive motor abnormalities, psychiatric symptoms, and cognitive decline. Expression of an expanded polyglutamine repeat within the Huntingtin (Htt) protein impacts numerous cellular processes, including protein folding and clearance. A hallmark of the disease is the progressive formation of inclusions that represent the culmination of a complex aggregation process. Methylene blue (MB), has been shown to modulate aggregation of amyloidogenic disease proteins. We investigated whether MB could impact mutant Htt-mediated aggregation and neurotoxicity. MB inhibited recombinant protein aggregation in vitro, even when added to preformed oligomers and fibrils. MB also decreased oligomer number and size and decreased accumulation of insoluble mutant Htt in cells. In functional assays, MB increased survival of primary cortical neurons transduced with mutant Htt, reduced neurodegeneration and aggregation in a Drosophila melanogaster model of HD, and reduced disease phenotypes in R6/2 HD modeled mice. Furthermore, MB treatment also promoted an increase in levels of BDNF RNA and protein in vivo. Thus, MB, which is well tolerated and used in humans, has therapeutic potential for HD.


Subject(s)
Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Huntington Disease/drug therapy , Methylene Blue/pharmacology , Methylene Blue/therapeutic use , Nerve Tissue Proteins/metabolism , Analysis of Variance , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Drosophila , Embryo, Mammalian , Excitatory Amino Acid Antagonists/toxicity , Humans , Huntingtin Protein , Huntington Disease/genetics , Kynurenic Acid/toxicity , Mice , Mice, Inbred C57BL , Microscopy, Atomic Force , Mutation/genetics , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Neurons/drug effects , Neurons/metabolism , Psychomotor Performance , Rats , Rotarod Performance Test , Transfection , Trinucleotide Repeat Expansion/genetics
11.
J Biol Chem ; 287(40): 33691-705, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22843695

ABSTRACT

Familial Parkinson disease (PD) can result from α-synuclein gene multiplication, implicating the reduction of neuronal α-synuclein as a therapeutic target. Moreover, α-synuclein content in human cerebrospinal fluid (CSF) represents a PD biomarker candidate. However, capture-based assays for α-synuclein quantification in CSF (such as by ELISA) have shown discrepancies and have limited suitability for high-throughput screening. Here, we describe two sensitive, in-solution, time-resolved Förster's resonance energy transfer (TR-FRET)-based immunoassays for total and oligomeric α-synuclein quantification. CSF analysis showed strong concordance for total α-synuclein content between two TR-FRET assays and, in agreement with a previously characterized 36 h protocol-based ELISA, demonstrated lower α-synuclein levels in PD donors. Critically, the assay suitability for high-throughput screening of siRNA constructs and small molecules aimed at reducing endogenous α-synuclein levels was established and validated. In a small-scale proof of concept compound screen using 384 well plates, signals ranged from <30 to >120% of the mean of vehicle-treated cells for molecules known to lower and increase cellular α-synuclein, respectively. Furthermore, a reverse genetic screen of a kinase-directed siRNA library identified seven genes that modulated α-synuclein protein levels (five whose knockdown increased and two that decreased cellular α-synuclein protein). This provides critical new biological insight into cellular pathways regulating α-synuclein steady-state expression that may help guide further drug discovery efforts. Moreover, we describe an inherent limitation in current α-synuclein oligomer detection methodology, a finding that will direct improvement of future assay design. Our one-step TR-FRET-based platform for α-synuclein quantification provides a novel platform with superior performance parameters for the rapid screening of large biomarker cohorts and of compound and genetic libraries, both of which are essential to the development of PD therapies.


Subject(s)
Biomarkers/metabolism , Parkinson Disease/diagnosis , alpha-Synuclein/chemistry , Animals , Antibodies/metabolism , Biochemistry/methods , Cohort Studies , Drug Design , Female , Fluorescence Resonance Energy Transfer/methods , Gene Expression Regulation , Gene Library , Humans , Immunoassay/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , RNA, Small Interfering/metabolism , alpha-Synuclein/cerebrospinal fluid
12.
Chem Biol ; 19(2): 264-75, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22365609

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by the amplification of a polyglutamine stretch at the N terminus of the huntingtin protein. N-terminal fragments of the mutant huntingtin (mHtt) aggregate and form intracellular inclusions in brain and peripheral tissues. Aggregates are an important hallmark of the disease, translating into a high need to quantify them in vitro and in vivo. We developed a one-step TR-FRET-based immunoassay to quantify soluble and aggregated mHtt in cell and tissue homogenates. Strikingly, quantification revealed a decrease of soluble mHtt correlating with an increase of aggregated protein in primary neuronal cell cultures, transgenic R6/2, and HdhQ150 knock-in HD mice. These results emphasize the assay's efficiency for highly sensitive and quantitative detection of soluble and aggregated mHtt and its application in high-throughput screening and characterization of HD models.


Subject(s)
Huntington Disease/metabolism , Immunoassay , Nerve Tissue Proteins/analysis , Nuclear Proteins/analysis , Animals , Cells, Cultured , Fluorescence Resonance Energy Transfer , Gene Knock-In Techniques , Huntingtin Protein , Huntington Disease/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
13.
J Huntingtons Dis ; 1(1): 119-32, 2012.
Article in English | MEDLINE | ID: mdl-24086178

ABSTRACT

The Huntington's disease (HD) mutation leads to a complex process of Huntingtin (Htt) aggregation into multimeric species that eventually form visible inclusions in cytoplasm, nuclei and neuronal processes. One hypothesis is that smaller, soluble forms of amyloid proteins confer toxic effects and contribute to early cell dysfunction. However, analysis of mutant Htt aggregation intermediates to identify conformers that may represent toxic forms of the protein and represent potential drug targets remains difficult. We performed a detailed analysis of aggregation conformers in multiple in vitro, cell and ex vivo models of HD. Conformation-specific antibodies were used to identify and characterize aggregation species, allowing assessment of multiple conformers present during the aggregation process. Using a series of assays together with these antibodies, several forms could be identified. Fibrillar oligomers, defined as having a ß-sheet rich conformation, are observed in vitro using recombinant protein and in protein extracts from cells in culture or mouse brain and shown to be globular, soluble and non-sedimentable structures. Compounds previously described to modulate visible inclusion body formation and reduce toxicity in HD models were also tested and consistently found to alter the formation of fibrillar oligomers. Interestingly, these compounds did not alter the rate of visible inclusion formation, indicating that fibrillar oligomers are not necessarily the rate limiting step of inclusion body formation. Taken together, we provide insights into the structure and formation of mutant Htt fibrillar oligomers that are modulated by small molecules with protective potential in HD models.


Subject(s)
Amyloid/chemistry , Amyloid/genetics , Huntington Disease/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Animals , Brain Chemistry , Cell Line, Tumor , Humans , Huntingtin Protein , Mice , Mice, Transgenic , Models, Biological , Mutation/genetics
14.
Nat Chem Biol ; 7(12): 925-34, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-22037470

ABSTRACT

Polyglutamine (polyQ) stretches exceeding a threshold length confer a toxic function to proteins that contain them and cause at least nine neurological disorders. The basis for this toxicity threshold is unclear. Although polyQ expansions render proteins prone to aggregate into inclusion bodies, this may be a neuronal coping response to more toxic forms of polyQ. The exact structure of these more toxic forms is unknown. Here we show that the monoclonal antibody 3B5H10 recognizes a species of polyQ protein in situ that strongly predicts neuronal death. The epitope selectively appears among some of the many low-molecular-weight conformational states assumed by expanded polyQ and disappears in higher-molecular-weight aggregated forms, such as inclusion bodies. These results suggest that protein monomers and possibly small oligomers containing expanded polyQ stretches can adopt a conformation that is recognized by 3B5H10 and is toxic or closely related to a toxic species.


Subject(s)
Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Peptides/chemistry , Peptides/toxicity , Antibodies, Monoclonal/immunology , Antibody Specificity , Cell Death/drug effects , Cells, Cultured , Epitopes/chemistry , Epitopes/immunology , Epitopes/toxicity , HEK293 Cells , Humans , Inclusion Bodies/chemistry , Molecular Weight , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Peptides/immunology , Structure-Activity Relationship , Trinucleotide Repeat Expansion
15.
Chem Biol ; 18(9): 1113-25, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21944750

ABSTRACT

Polyglutamine(polyQ)-expanded proteins are potential therapeutic targets for the treatment of polyQ expansion disorders such as Huntington's disease (HD) and spinocerebellar ataxia type 3 (SCA3). Here, we used an amino-terminal fragment of a mutant Huntingtin protein (Htt-N-82Q) as bait in an unbiased screen of a 60,000 peptoid library. Peptoid HQP09 was selected from the isolated hits and confirmed as a specific ligand of Htt-N-82Q and Atxn3-77Q mutant proteins in biochemical experiments. We identified three critical residues in the HQP09 sequence that are important for its activity and generated a minimal derivative, HQP09_9, which maintains the specific polyQ-binding activity. We demonstrated that HQP09 and HQP09_9 inhibited aggregation of Htt-N-53Q in vitro and exerted Ca(2+)-stabilizing and neuroprotective effects in experiments with primary striatal neuronal cultures derived from HD mice. We further demonstrated that intracerebroventricular delivery of HQP09 to an HD mouse model resulted in reduced accumulation of mutant Huntingtin aggregates and improved motor behavioral outcomes. These results suggest that HQP09 and similar peptoids hold promise as novel therapeutics for developing treatments for HD, SCA3, and other polyglutamine expansion disorders.


Subject(s)
Huntington Disease/drug therapy , Neuroprotective Agents/chemistry , Peptides/metabolism , Peptoids/chemistry , Peptoids/therapeutic use , Animals , Cells, Cultured , Huntingtin Protein , Machado-Joseph Disease/drug therapy , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Peptides/chemistry , Peptoids/pharmacology , Protein Binding
16.
J Biol Chem ; 285(49): 38183-93, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20864533

ABSTRACT

Inclusion bodies of aggregated mutant huntingtin (htt) fragments are a neuropathological hallmark of Huntington disease (HD). The molecular chaperones Hsp70 and Hsp40 colocalize to inclusion bodies and are neuroprotective in HD animal models. How these chaperones suppress mutant htt toxicity is unclear but might involve direct effects on mutant htt misfolding and aggregation. Using size exclusion chromatography and atomic force microscopy, we found that mutant htt fragments assemble into soluble oligomeric species with a broad size distribution, some of which reacted with the conformation-specific antibody A11. Hsp70 associated with A11-reactive oligomers in an Hsp40- and ATP-dependent manner and inhibited their formation coincident with suppression of caspase 3 activity in PC12 cells. Thus, Hsp70 and Hsp40 (DNAJB1) dynamically target specific subsets of soluble oligomers in a classic ATP-dependent reaction cycle, supporting a pathogenic role for these structures in HD.


Subject(s)
Adenosine Triphosphate/metabolism , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Inclusion Bodies/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Multimerization , Adenosine Triphosphate/genetics , Animals , Cattle , HSP40 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , Humans , Huntingtin Protein , Inclusion Bodies/genetics , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , PC12 Cells , Rats , Solubility
17.
J Biol Chem ; 285(19): 14777-90, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20220138

ABSTRACT

Huntington disease (HD) is caused by an expansion of more than 35-40 polyglutamine (polyQ) repeats in the huntingtin (htt) protein, resulting in accumulation of inclusion bodies containing fibrillar deposits of mutant htt fragments. Intriguingly, polyQ length is directly proportional to the propensity for htt to form fibrils and the severity of HD and is inversely correlated with age of onset. Although the structural basis for htt toxicity is unclear, the formation, abundance, and/or persistence of toxic conformers mediating neuronal dysfunction and degeneration in HD must also depend on polyQ length. Here we used atomic force microscopy to demonstrate mutant htt fragments and synthetic polyQ peptides form oligomers in a polyQ length-dependent manner. By time-lapse atomic force microscopy, oligomers form before fibrils, are transient in nature, and are occasionally direct precursors to fibrils. However, the vast majority of fibrils appear to form by monomer addition coinciding with the disappearance of oligomers. Thus, oligomers must undergo a major structural transition preceding fibril formation. In an immortalized striatal cell line and in brain homogenates from a mouse model of HD, a mutant htt fragment formed oligomers in a polyQ length-dependent manner that were similar in size to those formed in vitro, although these structures accumulated over time in vivo. Finally, using immunoelectron microscopy, we detected oligomeric-like structures in human HD brains. These results demonstrate that oligomer formation by a mutant htt fragment is strongly polyQ length-dependent in vitro and in vivo, consistent with a causative role for these structures, or subsets of these structures, in HD pathogenesis.


Subject(s)
Mutant Proteins/chemistry , Mutation/genetics , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Peptides/metabolism , Animals , Brain/cytology , Brain/metabolism , Corpus Striatum/cytology , Corpus Striatum/metabolism , Humans , Huntingtin Protein , In Vitro Techniques , Mice , Microscopy, Atomic Force , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptides/chemistry , Trinucleotide Repeat Expansion
18.
J Neurosci ; 29(28): 9104-14, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19605647

ABSTRACT

Endogenous protein quality control machinery has long been suspected of influencing the onset and progression of neurodegenerative diseases characterized by accumulation of misfolded proteins. Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expansion of a polyglutamine (polyQ) tract in the protein huntingtin (htt), which leads to its aggregation and accumulation in inclusion bodies. Here, we demonstrate in a mouse model of HD that deletion of the molecular chaperones Hsp70.1 and Hsp70.3 significantly exacerbated numerous physical, behavioral and neuropathological outcome measures, including survival, body weight, tremor, limb clasping and open field activities. Deletion of Hsp70.1 and Hsp70.3 significantly increased the size of inclusion bodies formed by mutant htt exon 1, but surprisingly did not affect the levels of fibrillar aggregates. Moreover, the lack of Hsp70s significantly decreased levels of the calcium regulated protein c-Fos, a marker for neuronal activity. In contrast, deletion of Hsp70s did not accelerate disease in a mouse model of infectious prion-mediated neurodegeneration, ruling out the possibility that the Hsp70.1/70.3 mice are nonspecifically sensitized to all protein misfolding disorders. Thus, endogenous Hsp70s are a critical component of the cellular defense against the toxic effects of misfolded htt protein in neurons, but buffer toxicity by mechanisms independent of the deposition of fibrillar aggregates.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , HSP72 Heat-Shock Proteins/deficiency , Huntington Disease/genetics , Huntington Disease/pathology , Nerve Tissue Proteins/genetics , Age Factors , Analysis of Variance , Animals , Disease Models, Animal , Female , Gene Expression Regulation/genetics , HSP70 Heat-Shock Proteins/deficiency , HSP72 Heat-Shock Proteins/classification , Huntington Disease/complications , Huntington Disease/mortality , Inclusion Bodies/pathology , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Movement Disorders/etiology , Movement Disorders/genetics , Nerve Tissue Proteins/metabolism , Neurologic Examination/methods , Proto-Oncogene Proteins c-fos/metabolism , Trinucleotide Repeat Expansion/genetics , Weight Loss/genetics
19.
J Biol Chem ; 284(32): 21647-58, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19491400

ABSTRACT

Huntington disease (HD) is a neurodegenerative disorder caused by an expansion of a polyglutamine (polyQ) domain in the N-terminal region of huntingtin (htt). PolyQ expansion above 35-40 results in disease associated with htt aggregation into inclusion bodies. It has been hypothesized that expanded polyQ domains adopt multiple potentially toxic conformations that belong to different aggregation pathways. Here, we used atomic force microscopy to analyze the effect of a panel of anti-htt antibodies (MW1-MW5, MW7, MW8, and 3B5H10) on aggregate formation and the stability of a mutant htt-exon1 fragment. Two antibodies, MW7 (polyproline-specific) and 3B5H10 (polyQ-specific), completely inhibited fibril formation and disaggregated preformed fibrils, whereas other polyQ-specific antibodies had widely varying effects on aggregation. These results suggest that expanded polyQ domains adopt multiple conformations in solution that can be readily distinguished by monoclonal antibodies, which has important implications for understanding the structural basis for polyQ toxicity and the development of intrabody-based therapeutics for HD.


Subject(s)
Antibodies, Monoclonal/chemistry , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Peptides/chemistry , Amino Acid Sequence , Animals , Epitopes/chemistry , Huntingtin Protein , Huntington Disease/metabolism , Microscopy, Atomic Force/methods , Molecular Sequence Data , Mutation , Neurons/metabolism , Protein Conformation , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid
20.
J Cell Sci ; 121(Pt 5): 717-23, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18270269

ABSTRACT

Heat shock protein 90 (HSP90) is considered a specialized molecular chaperone that controls the folding of cell-regulatory proteins such as steroid receptors and kinases. However, its high abundance is suggestive of a more general function in other fundamental processes. Here, we show that HSP90 is required for vesicular protein transport in the cell. We have identified a novel chaperone complex comprising HSP90 and TPR1 that is recruited to the membrane protein VAP-33. Depletion of the TPR1 protein in mammalian cells inhibits transport of vesicular stomatitis virus glycoprotein (VSVG) and leads to accumulation of this cargo protein in the Golgi apparatus. Furthermore, trafficking of VSVG between Golgi stacks is dependent on the ATPase function of HSP90 and can be inhibited by drugs specific for HSP90. Our results identify a new role for HSP90 in protein sorting, pointing to a central role for this molecular chaperone in the cell.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Intracellular Fluid/metabolism , Transport Vesicles/metabolism , Vesicular Transport Proteins/metabolism , Adenosine Triphosphatases/metabolism , Animals , CHO Cells , Cell Membrane/metabolism , Cricetinae , Cricetulus , Endocytosis/physiology , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Humans , Macromolecular Substances/metabolism , Molecular Chaperones/metabolism , Protein Transport/physiology , Transport Vesicles/ultrastructure , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...