Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Int J Mol Sci ; 22(17)2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34502533

ABSTRACT

Various intrinsic and extrinsic factors can interfere with the process of protein folding, resulting in protein aggregates. Usually, cells prevent the formation of aggregates or degrade them to prevent the cytotoxic effects they may cause. However, during viral infection, the formation of aggregates may serve as a cellular defense mechanism. On the other hand, some viruses are able to exploit the process of aggregate formation and removal to promote their replication or evade the immune response. This review article summarizes the process of cellular protein aggregation and gives examples of how different viruses exploit it. Particular emphasis is placed on the ribonucleotide reductases of herpesviruses and how their additional non-canonical functions in viral immune evasion are closely linked to protein aggregation.


Subject(s)
Immune Evasion/immunology , Protein Aggregates , Protein Aggregation, Pathological/immunology , Virus Diseases/immunology , Viruses/immunology , Herpesviridae/immunology , Herpesviridae/physiology , Herpesviridae Infections/immunology , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Host-Pathogen Interactions/immunology , Humans , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/virology , Ribonucleotide Reductases/immunology , Ribonucleotide Reductases/metabolism , Virus Diseases/metabolism , Virus Diseases/virology
2.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166234, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34339840

ABSTRACT

TAR DNA-binding protein-43 (TDP-43) pathology, including fibrillar aggregates and mutations, develops in amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD) and limbic-predominant age-related TDP-43 encephalopathy (LATE). Hyperphosphorylation and aggregation of TDP-43 contribute to pathology and are viable therapeutic targets for ALS. In vivo inhibition of TDP-43 aggregation was evaluated using anti-TDP-43 antibodies with promising outcomes. However, the exact mechanism of antibody-based inhibition targeting TDP-43 is not well understood but may lead to the identification of viable immunotherapies. Herein, the mechanism of in vitro aggregation of phosphorylated TDP-43 was explored, and the anti-TDP-43 antibodies tested for their inhibitor efficacies. Specifically, the aggregation of phosphorylated full-length TDP-43 protein (pS410) was monitored by transmission electron microscopy (TEM), turbidity absorbance, and thioflavin (ThT) spectroscopy. The protein aggregates were insoluble, ThT-positive and characterized with heterogeneous morphologies (fibers, amorphous structures). Antibodies specific to epitopes 178-393 and 256-269, within the RRM2-CTD domain, reduced the formation of ß-sheets and insoluble aggregates, at low antibody loading (antibody: protein ratio = 1 µg/mL: 45 µg/mL). Inhibition outcomes were highly dependent on the type and loading of antibodies, indicating dual functionality of such inhibitors, as aggregation inhibitors or aggregation promoters. Anti-SOD1 and anti-tau antibodies were not effective inhibitors against TDP-43 aggregation, indicating selective inhibition.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Antibodies, Anti-Idiotypic/immunology , Brain Diseases/genetics , DNA-Binding Proteins/genetics , Frontotemporal Lobar Degeneration/genetics , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/therapy , Brain Diseases/immunology , Brain Diseases/pathology , Brain Diseases/therapy , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/immunology , Epitopes/immunology , Frontotemporal Lobar Degeneration/immunology , Frontotemporal Lobar Degeneration/pathology , Frontotemporal Lobar Degeneration/therapy , Humans , Microscopy, Electron, Transmission , Phosphorylation/genetics , Protein Aggregates/genetics , Protein Aggregates/immunology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/therapy , Protein Conformation, beta-Strand/genetics , Superoxide Dismutase-1/antagonists & inhibitors , Superoxide Dismutase-1/immunology , tau Proteins/antagonists & inhibitors , tau Proteins/immunology
3.
J Biol Chem ; 296: 100334, 2021.
Article in English | MEDLINE | ID: mdl-33508322

ABSTRACT

Systemic light chain (AL) amyloidosis is a fatal protein misfolding disease in which excessive secretion, misfolding, and subsequent aggregation of free antibody light chains eventually lead to deposition of amyloid plaques in various organs. Patient-specific mutations in the antibody VL domain are closely linked to the disease, but the molecular mechanisms by which certain mutations induce misfolding and amyloid aggregation of antibody domains are still poorly understood. Here, we compare a patient VL domain with its nonamyloidogenic germline counterpart and show that, out of the five mutations present, two of them strongly destabilize the protein and induce amyloid fibril formation. Surprisingly, the decisive, disease-causing mutations are located in the highly variable complementarity determining regions (CDRs) but exhibit a strong impact on the dynamics of conserved core regions of the patient VL domain. This effect seems to be based on a deviation from the canonical CDR structures of CDR2 and CDR3 induced by the substitutions. The amyloid-driving mutations are not necessarily involved in propagating fibril formation by providing specific side chain interactions within the fibril structure. Rather, they destabilize the VL domain in a specific way, increasing the dynamics of framework regions, which can then change their conformation to form the fibril core. These findings reveal unexpected influences of CDR-framework interactions on antibody architecture, stability, and amyloid propensity.


Subject(s)
Amyloid/ultrastructure , Complementarity Determining Regions/genetics , Immunoglobulin Light-chain Amyloidosis/genetics , Plaque, Amyloid/genetics , Amino Acid Sequence/genetics , Amyloid/genetics , Amyloid/immunology , Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/immunology , Amyloidogenic Proteins/ultrastructure , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/ultrastructure , Humans , Immunoglobulin Light-chain Amyloidosis/immunology , Immunoglobulin Light-chain Amyloidosis/metabolism , Mutation/genetics , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/pathology , Protein Conformation , Protein Folding
4.
J Biol Chem ; 296: 100241, 2021.
Article in English | MEDLINE | ID: mdl-33376140

ABSTRACT

Conformationally distinct aggregates of the amyloid ß (Aß) peptide accumulate in brains of patients with Alzheimer's disease (AD), but the roles of the different aggregates in disease progression are not clear. We previously isolated two single-chain variable domain antibody fragments (scFvs), C6T and A4, that selectively bind different toxic conformational variants of oligomeric Aß. Here, we utilize these scFvs to localize the presence of these Aß variants in human AD brain and to demonstrate their potential as therapeutic agents for treating AD. Both A4 and C6T label oligomeric Aß in extracellular amyloid plaques, whereas C6T also labels intracellular oligomeric Aß in human AD brain tissue and in an AD mouse model. For therapeutic studies, the A4 and C6T scFvs were expressed in the AD mice by viral infection of liver cells. The scFvs were administered at 2 months of age, and mice sacrificed at 9 months. The scFvs contained a peptide tag to facilitate transport across the blood brain barrier. While treatment with C6T only slightly decreased Aß deposits and plaque-associated inflammation, it restored neuronal integrity to WT levels, significantly promoted growth of new neurons, and impressively rescued survival rates to WT levels. Treatment with A4 on the other hand significantly decreased Aß deposits but did not significantly decrease neuroinflammation or promote neuronal integrity, neurogenesis, or survival rate. These results suggest that the specific Aß conformation targeted in therapeutic applications greatly affects the outcome, and the location of the targeted Aß variants may also play a critical factor.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Neurons/metabolism , Single-Chain Antibodies/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/ultrastructure , Animals , Brain/immunology , Brain/metabolism , Brain/ultrastructure , Disease Models, Animal , Disease Progression , Humans , Mice , Neurogenesis/genetics , Neurogenesis/immunology , Neurons/pathology , Neurons/ultrastructure , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/immunology , Protein Conformation , Single-Chain Antibodies/immunology , Single-Chain Antibodies/ultrastructure
5.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255488

ABSTRACT

The accumulation of amyloid protein aggregates in tissues is the basis for the onset of diseases known as amyloidoses. Intriguingly, many amyloidoses impact the central nervous system (CNS) and usually are devastating diseases. It is increasingly apparent that neurotoxic soluble oligomers formed by amyloidogenic proteins are the primary molecular drivers of these diseases, making them lucrative diagnostic and therapeutic targets. One promising diagnostic/therapeutic strategy has been the development of antibody fragments against amyloid oligomers. Antibody fragments, such as fragment antigen-binding (Fab), scFv (single chain variable fragments), and VHH (heavy chain variable domain or single-domain antibodies) are an alternative to full-length IgGs as diagnostics and therapeutics for a variety of diseases, mainly because of their increased tissue penetration (lower MW compared to IgG), decreased inflammatory potential (lack of Fc domain), and facile production (low structural complexity). Furthermore, through the use of in vitro-based ligand selection, it has been possible to identify antibody fragments presenting marked conformational selectivity. In this review, we summarize significant reports on antibody fragments selective for oligomers associated with prevalent CNS amyloidoses. We discuss promising results obtained using antibody fragments as both diagnostic and therapeutic agents against these diseases. In addition, the use of antibody fragments, particularly scFv and VHH, in the isolation of unique oligomeric assemblies is discussed as a strategy to unravel conformational moieties responsible for neurotoxicity. We envision that advances in this field may lead to the development of novel oligomer-selective antibody fragments with superior selectivity and, hopefully, good clinical outcomes.


Subject(s)
Amyloid/immunology , Amyloidosis/diagnosis , Neurotoxicity Syndromes/diagnosis , Protein Aggregation, Pathological/diagnosis , Amyloid/antagonists & inhibitors , Amyloidosis/immunology , Amyloidosis/pathology , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fragments/immunology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , Peptide Fragments/immunology , Protein Aggregation, Pathological/immunology , Single-Domain Antibodies , Structure-Activity Relationship
6.
J Mol Biol ; 432(23): 6187-6199, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33058870

ABSTRACT

In antibody light chain amyloidosis (AL), mutant light chains (LCs) or their variable domains (VLs) form fibrils, which accumulate in organs and lead to their failure. The molecular mechanism of this disease is still poorly understood. One of the key open issues is whether the mutant VLs and LCs differ in fibril formation. We addressed this question studying the effects of the VL mutations S20N and R61A within the isolated VL domain and in the full-length LC scaffold. Both VL variants readily form fibrils. Here, we find that in the LC context, the S20N variant is protected from fibril formation while for LC R61A fibril formation is even accelerated compared to VL R61A. Our analyses revealed that the partially unfolded state of the VL R61A domain destabilizes the CL domain by non-native interactions, in turn leading to a further unfolding of the VL domain. In contrast, the folded mutant VL S20N and VL wt form native interactions with CL. These are beneficial for LC stability and promote amyloid resistance. Thus the effects of specific mutations on the VL fold can have opposing effects on LC domain interactions, stability and amyloidogenicity.


Subject(s)
Amyloid/genetics , Amyloidogenic Proteins/genetics , Immunoglobulin Light Chains/immunology , Protein Aggregation, Pathological/genetics , Amino Acid Sequence/genetics , Amyloid/immunology , Amyloidogenic Proteins/immunology , Amyloidosis/genetics , Amyloidosis/immunology , Humans , Immunoglobulin Light Chains/genetics , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/immunology , Protein Aggregation, Pathological/immunology , Protein Conformation
7.
Front Immunol ; 11: 725, 2020.
Article in English | MEDLINE | ID: mdl-32431697

ABSTRACT

Patients treated with bioproducts (BPs) frequently develop anti-drug antibodies (ADAs) with potential neutralizing capacities leading to loss of clinical response or potential hypersensitivity reactions. Many factors can influence BP immunogenicity and could be related to the patient, the treatment, as well as to the product itself. Among these latter factors, it is now well accepted that BP aggregation is associated with an increased potential for immunogenicity, as aggregates seem to be correlated with ADA development. Moreover, the presence of high-affinity ADAs suggests a CD4 T-cell dependent adaptive immune response and therefore a pivotal role for antigen-presenting cells (APCs), such as dendritic cells (DCs). In this review, we address the in vitro methods developed to evaluate how monoclonal antibodies could trigger the immunization process by focusing on the role of aggregated antibodies in the establishment of this response. In particular, we will present the different cell-based assays that have been used to assess the potential of antibodies and their aggregates to modulate cellular mechanisms leading to activation and the biological parameters (cellular activation markers, proliferation and secreted molecules) that can be measured to evaluate the different cell activation stages and their consequences in the propagation of the immune response. Indeed, the use of such strategies could help evaluate the risk of BP immunogenicity and their role in mitigating this risk.


Subject(s)
Adaptive Immunity , Antibodies, Monoclonal/immunology , Biological Products/immunology , Models, Immunological , Protein Aggregates/immunology , Protein Aggregation, Pathological/immunology , Antibodies, Monoclonal/therapeutic use , Biological Products/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Humans , Immunization
8.
Neurobiol Dis ; 141: 104943, 2020 07.
Article in English | MEDLINE | ID: mdl-32407769

ABSTRACT

Huntington's disease (HD) is caused by a highly polymorphic CAG trinucleotide expansion in the gene encoding for the huntingtin protein (HTT). The resulting mutant huntingtin protein (mutHTT) is ubiquitously expressed but also exhibits the ability to propagate from cell-to-cell to disseminate pathology; a property which may serve as a new therapeutic focus. Accordingly, we set out to develop a monoclonal antibody (mAB) targeting a particularly exposed region close to the aa586 caspase-6 cleavage site of the HTT protein. This monoclonal antibody, designated C6-17, effectively binds mutHTT and is able to deplete the protein from cell culture supernatants. Using cell-based assays, we demonstrate that extracellular secretion of mutHTT into cell culture media and its subsequent uptake in recipient HeLa cells can be almost entirely blocked by mAB C6-17. Immunohistochemical stainings of post-mortem HD brain tissue confirmed the specificity of mAB C6-17 to human mutHTT aggregates. These findings demonstrate that mAB C6-17 not only successfully engages with its target, mutHTT, but also inhibits cell uptake suggesting that this antibody could interfere with the pathological processes of mutHTT spreading in vivo.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/immunology , Huntington Disease/metabolism , Animals , Biological Transport , Female , HEK293 Cells , HeLa Cells , Humans , Huntington Disease/prevention & control , Mice, Inbred BALB C , Mutation , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/prevention & control
9.
Eur J Pharm Biopharm ; 152: 164-174, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32416134

ABSTRACT

Amorphous aggregates of therapeutic proteins can provoke an unwanted immune response (anti-drug antibodies; ADAs), but counter-examples have led to some controversy. Amorphous aggregates can possess unique biophysical and biochemical attributes depending on both the way they are generated and the protein's biophysical/biochemical properties. Here, we examine the immunogenicity of an anti-EGFR single domain antibody (VHH) in four types of amorphous aggregates: two heat-aggregated VHH incubated at 65 °C (VHH-65) and 95 °C (VHH-95), a misfolded VHH isolated from the insoluble fraction of the E. coli lysate (VHH-Ins), and a low solubility misfolded VHH produced by miss-shuffling the SS bonds of the native VHH (VHH-Mis). Biophysical and biochemical measurements indicated that VHH was indeed natively folded, monomeric, and ß-sheeted; that VHH-65 was partially unfolded and formed aggregates with a Z-average (Zave) of 771 nm; whereas VHH-95 was unfolded and formed aggregates of 1722 nm; and that both VHH-Ins and VHH-Mis were misfolded with non-native intermolecular SS bonds and formed aggregates with a Zave of 1846 nm and 1951 nm, respectively. The IgG level generated in Jcl:ICR mice determined by ELISA showed that the native VHH was barely immunogenic, VHH-95 was not immunogenic, while VHH-65 was mildly immunogenic. By contrast, the misfolded aggregates, VHH-Ins and VHH-Mis, having a Zave and an aggregation propensity similar to that of VHH-95, were highly immunogenic. These findings indicate the critical role of the biochemical and biophysical attributes of the amorphous aggregates in generating an immune response against a protein, rather than just their sizes.


Subject(s)
Antibodies/immunology , Antibody Formation/immunology , ErbB Receptors/immunology , Protein Aggregates/immunology , Protein Aggregation, Pathological/immunology , Single-Domain Antibodies/immunology , Animals , Escherichia coli/immunology , Female , Hot Temperature , Mice , Mice, Inbred ICR , Protein Folding , Solubility
10.
Acta Neuropathol Commun ; 8(1): 13, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32019610

ABSTRACT

Tau deposition in the brain is a pathological hallmark of many neurodegenerative disorders, including Alzheimer's disease (AD). During the course of these tauopathies, tau spreads throughout the brain via synaptically-connected pathways. Such propagation of pathology is thought to be mediated by tau species ("seeds") containing the microtubule binding region (MTBR) composed of either three repeat (3R) or four repeat (4R) isoforms. The tau MTBR also forms the core of the neuropathological filaments identified in AD brain and other tauopathies. Multiple approaches are being taken to limit tau pathology, including immunotherapy with anti-tau antibodies. Given its key structural role within fibrils, specifically targetting the MTBR with a therapeutic antibody to inhibit tau seeding and aggregation may be a promising strategy to provide disease-modifying treatment for AD and other tauopathies. Therefore, a monoclonal antibody generating campaign was initiated with focus on the MTBR. Herein we describe the pre-clinical generation and characterisation of E2814, a humanised, high affinity, IgG1 antibody recognising the tau MTBR. E2814 and its murine precursor, 7G6, as revealed by epitope mapping, are antibodies bi-epitopic for 4R and mono-epitopic for 3R tau isoforms because they bind to sequence motif HVPGG. Functionally, both antibodies inhibited tau aggregation in vitro. They also immunodepleted a variety of MTBR-containing tau protein species. In an in vivo model of tau seeding and transmission, attenuation of deposition of sarkosyl-insoluble tau in brain could also be observed in response to antibody treatment. In AD brain, E2814 bound different types of tau filaments as shown by immunogold labelling and recognised pathological tau structures by immunohistochemical staining. Tau fragments containing HVPGG epitopes were also found to be elevated in AD brain compared to PSP or control. Taken together, the data reported here have led to E2814 being proposed for clinical development.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/therapy , Antibodies, Monoclonal/immunology , Immunization, Passive/methods , tau Proteins/genetics , tau Proteins/immunology , Alzheimer Disease/pathology , Animals , Antibodies, Monoclonal/pharmacology , Frontal Lobe/metabolism , Frontal Lobe/pathology , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Male , Mice, Transgenic , Protein Aggregation, Pathological/immunology , Protein Isoforms/immunology , Protein Isoforms/pharmacology
11.
J Immunol ; 204(5): 1345-1361, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31969389

ABSTRACT

Aggregation of α-synuclein (αSN) is an important histological feature of Parkinson disease. Recent studies showed that the release of misfolded αSN from human and rodent neurons is relevant to the progression and spread of αSN pathology. Little is known, however, about the mechanisms responsible for clearance of extracellular αSN. This study found that human complement receptor (CR) 4 selectively bound fibrillar αSN, but not monomeric species. αSN is an abundant protein in the CNS, which potentially could overwhelm clearance of cytotoxic αSN species. The selectivity of CR4 toward binding fibrillar αSN consequently adds an important αSN receptor function for maintenance of brain homeostasis. Based on the recently solved structures of αSN fibrils and the known ligand preference of CR4, we hypothesize that the parallel monomer stacking in fibrillar αSN creates a known danger-associated molecular pattern of stretches of anionic side chains strongly bound by CR4. Conformational change in the receptor regulated tightly clearance of fibrillar αSN by human monocytes. The induced change coupled concomitantly with phagolysosome formation. Data mining of the brain transcriptome in Parkinson disease patients supported CR4 as an active αSN clearance mechanism in this disease. Our results associate an important part of the innate immune system, namely complement receptors, with the central molecular mechanisms of CNS protein aggregation in neurodegenerative disorders.


Subject(s)
Integrin alphaXbeta2 , Macrophages , Parkinson Disease , Phagosomes , Protein Aggregation, Pathological , alpha-Synuclein , Humans , Integrin alphaXbeta2/chemistry , Integrin alphaXbeta2/genetics , Integrin alphaXbeta2/immunology , Macrophages/immunology , Macrophages/pathology , Parkinson Disease/genetics , Parkinson Disease/immunology , Parkinson Disease/pathology , Phagosomes/chemistry , Phagosomes/genetics , Phagosomes/immunology , Phagosomes/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/pathology , Protein Structure, Quaternary , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/immunology
12.
Med Chem ; 16(8): 1007-1021, 2020.
Article in English | MEDLINE | ID: mdl-31702507

ABSTRACT

Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, with approximately 29 million older people suffering from this disease worldwide. This number is expected to become triple by 2050. AD is a complex and multifactorial neurodegenerative condition, characterized by complex pathology including oxidative stress, formation of aggregates of amyloid and tau, enhanced immune responses, metal deposition and disturbances in cholinesterase enzymes. There is no effective pharmacological treatment for combating the disease to date. The ineffectiveness of current pharmacological interventions in AD has led scientists to search for more safe and effective alternative therapeutic agents. Thus, natural products have become an important avenue for drug discovery in AD research. In this regard, polyphenols are natural products that have been shown to be effective in the modulation of the type of neurodegenerative changes seen in AD, suggesting a possible therapeutic role. The present review focuses on the chemistry of polyphenols, clinical studies for evaluating polyphenols as effective alternatives in AD treatment, cellular and molecular aspects of polyphenols in improving cognitive deficits and the current challenges and futuristic approaches to use polyphenols as safe and effective therapeutic agents in AD treatment.


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/pharmacology , Polyphenols/pharmacology , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Humans , Neuroprotective Agents/chemistry , Oxidative Stress/drug effects , Polyphenols/chemistry , Protein Aggregates/drug effects , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/immunology , tau Proteins/antagonists & inhibitors , tau Proteins/immunology , tau Proteins/metabolism
13.
ACS Chem Neurosci ; 10(8): 3464-3478, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31313906

ABSTRACT

The formation of misfolded protein oligomers during early stages of amyloid aggregation and the activation of neuroinflammatory responses are two key events associated with neurodegenerative diseases. Although it has been established that misfolded oligomers are involved in the neuroinflammatory process, the links between their structural features and their functional effects on the immune response remain unknown. To explore such links, we took advantage of two structurally distinct soluble oligomers (type A and B) of protein HypF-N and compared the elicited microglial inflammatory responses. By using confocal microscopy, protein pull-down, and high-throughput mass spectrometry, we found that, even though both types bound to a common pool of microglial proteins, type B oligomers-with a lower solvent-exposed hydrophobicity-showed enhanced protein binding, correlating with the observed inflammatory response. Furthermore, the interactome associated with inflammatory-mediated neurodegeneration revealed previously unidentified receptors and signaling molecules likely to be involved in the oligomer-elicited innate immune response.


Subject(s)
Carboxyl and Carbamoyl Transferases/immunology , Escherichia coli Proteins/immunology , Immunity, Innate/immunology , Microglia/immunology , Protein Aggregation, Pathological/immunology , Animals , Cell Line , Cricetinae , Humans , Mice , Microglia/pathology , Protein Aggregation, Pathological/pathology , Protein Binding
14.
ACS Chem Biol ; 14(7): 1410-1417, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31194501

ABSTRACT

Amyloid cascade and neuroinflammation are hallmarks of neurodegenerative diseases, and pro-inflammatory S100A9 protein is central to both of them. Here, we have shown that NCAM1 peptide constructs carrying polycationic sequences derived from Aß peptide (KKLVFF) and PrP protein (KKRPKP) significantly promote the S100A9 amyloid self-assembly in a concentration-dependent manner by making transient interactions with individual S100A9 molecules, perturbing its native structure and acting as catalysts. Since the individual molecule misfolding is a rate-limiting step in S100A9 amyloid aggregation, the effects of the NCAM1 construct on the native S100A9 are so critical for its amyloid self-assembly. S100A9 rapid self-assembly into large aggregated clumps may prevent its amyloid tissue propagation, and by modulating S100A9 aggregation as a part of the amyloid cascade, the whole process may be effectively tuned.


Subject(s)
Amyloid/immunology , CD56 Antigen/immunology , Calgranulin B/immunology , Protein Aggregation, Pathological/immunology , Amino Acid Sequence , Amyloid/chemistry , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , CD56 Antigen/chemistry , Calgranulin B/chemistry , Humans , Inflammation/immunology , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/immunology , Prions/chemistry , Prions/immunology , Protein Aggregates
15.
J Neurochem ; 150(5): 612-625, 2019 09.
Article in English | MEDLINE | ID: mdl-31055836

ABSTRACT

Synucleinopathies including Parkinson's disease, dementia with Lewy bodies and multiple system atrophy are characterized by the abnormal accumulation and propagation of α-synuclein (α-syn) pathology in the central and peripheral nervous system as Lewy bodies or glial cytoplasmic inclusions. Several antibodies against α-syn have been developed since it was first detected as the major component of Lewy bodies and glial cytoplasmic inclusions. Over the years, researchers have generated specific antibodies that alleviate the accumulation of intracellular aggregated α-syn and associated pathology in cellular and preclinical models of synucleinopathies. So far, antibodies have been the first choice as tools for research and diagnosis and currently, a wide variety of antibody fragments have been developed as an alternative to full-length antibodies for increasing its therapeutic usefulness. Recently, conformation specific antibody-based approaches have been found to be promising as therapeutic strategies, both to block α-syn aggregation and ameliorate the resultant cytotoxicity, and as diagnostic tools. In this review, we summarize different α-syn specific antibodies and provide their usefulness in tackling synucleinopathies. This article is part of the Special Issue "Synuclein".


Subject(s)
Antibodies/immunology , Synucleinopathies/therapy , alpha-Synuclein/immunology , Antibodies/therapeutic use , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Biomarkers , Delayed Diagnosis , Epitopes/immunology , Humans , Immunoglobulin Fragments/immunology , Immunologic Tests/methods , Parkinson Disease/diagnosis , Parkinson Disease/immunology , Parkinson Disease/therapy , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/prevention & control , Protein Conformation , Protein Engineering , Recombinant Proteins/immunology , Single-Domain Antibodies/immunology , Synucleinopathies/diagnosis , Synucleinopathies/immunology , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/chemistry
16.
Immunity ; 50(4): 955-974, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995509

ABSTRACT

Neurodegenerative diseases of the central nervous system progressively rob patients of their memory, motor function, and ability to perform daily tasks. Advances in genetics and animal models are beginning to unearth an unexpected role of the immune system in disease onset and pathogenesis; however, the role of cytokines, growth factors, and other immune signaling pathways in disease pathogenesis is still being examined. Here we review recent genetic risk and genome-wide association studies and emerging mechanisms for three key immune pathways implicated in disease, the growth factor TGF-ß, the complement cascade, and the extracellular receptor TREM2. These immune signaling pathways are important under both healthy and neurodegenerative conditions, and recent work has highlighted new functional aspects of their signaling. Finally, we assess future directions for immune-related research in neurodegeneration and potential avenues for immune-related therapies.


Subject(s)
Neurodegenerative Diseases/immunology , Signal Transduction/immunology , Aging/immunology , Animals , Complement Activation , Disease Progression , Genetic Predisposition to Disease , Genome-Wide Association Study , Gliosis/immunology , Gliosis/pathology , Humans , Immunity, Innate , Inflammation/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Mice, Transgenic , Microglia/immunology , Models, Immunological , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy , Protein Aggregation, Pathological/immunology , Receptors, Immunologic/immunology , Transforming Growth Factor beta/immunology
17.
Nat Commun ; 10(1): 1269, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894521

ABSTRACT

Systemic light chain amyloidosis (AL)  is a life-threatening disease caused by aggregation and deposition of monoclonal immunoglobulin light chains (LC) in target organs. Severity of heart involvement is the most important factor determining prognosis. Here, we report the 4.0 Å resolution cryo-electron microscopy map and molecular model of amyloid fibrils extracted from the heart of an AL amyloidosis patient with severe amyloid cardiomyopathy. The helical fibrils are composed of a single protofilament, showing typical 4.9 Å stacking and cross-ß architecture. Two distinct polypeptide stretches (total of 77 residues) from the LC variable domain (Vl) fit the fibril density. Despite Vl high sequence variability, residues stabilizing the fibril core are conserved through different cardiotoxic Vl, highlighting structural motifs that may be common to misfolding-prone LCs. Our data shed light on the architecture of LC amyloids, correlate amino acid sequences with fibril assembly, providing the grounds for development of innovative medicines.


Subject(s)
Amyloid/ultrastructure , Immunoglobulin Light Chains/ultrastructure , Immunoglobulin Light-chain Amyloidosis/pathology , Myocardium/ultrastructure , Protein Aggregation, Pathological/pathology , Aged , Amino Acid Sequence , Amyloid/immunology , Amyloid/metabolism , Autopsy , Cryoelectron Microscopy , Humans , Immunoglobulin Light Chains/immunology , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light-chain Amyloidosis/diagnosis , Immunoglobulin Light-chain Amyloidosis/immunology , Immunoglobulin Light-chain Amyloidosis/metabolism , Male , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Protein Aggregation, Pathological/diagnosis , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/metabolism , Protein Conformation, beta-Strand , Protein Folding , Sequence Alignment , Sequence Homology, Amino Acid , Severity of Illness Index
18.
Neurobiol Dis ; 124: 202-217, 2019 04.
Article in English | MEDLINE | ID: mdl-30481547

ABSTRACT

It has been suggested that aggregation of α-synuclein (α-syn) into oligomers leads to neurodegeneration in Parkinson's disease (PD), but intravenous immunoglobulin (IVIG) which contains antibodies against α-syn monomers and oligomers fails to treat PD mouse model. The reason may be because IVIG contains much low level of antibodies against α-syn, and of which only a small part can penetrate the blood-brain barrier, resulting in an extremely low level of effective antibodies in the brain, and limiting the beneficial effect of IVIG on PD mice. Here, we first isolated naturally occurring autoantibodies against α-syn (NAbs-α-syn) from IVIG. Our further investigation results showed that NAbs-α-syn inhibited α-syn aggregation and attenuated α-syn-induced cytotoxicity in vitro. Compared with vehicles, NAbs-α-syn significantly attenuated the memory and motor deficits by reducing the levels of soluble α-syn, total human α-syn and α-syn oligomers, decreasing the intracellular p-α-synser129 deposits and axonal pathology, inhibiting the microgliosis and astrogliosis, as well as the production of proinflammatory cytokines, increasing the levels of PSD95, synaptophysin and TH in the brain of A53T transgenic mice. These findings suggest that NAbs-α-syn overcomes the deficiency of IVIG and exhibits a promising therapeutic potential for the treatment of PD.


Subject(s)
Autoantibodies/administration & dosage , Brain/immunology , Motor Activity , Parkinson Disease/immunology , Spatial Memory , alpha-Synuclein/immunology , Animals , Autoantibodies/isolation & purification , Brain/pathology , Disease Models, Animal , Immunization, Passive , Immunoglobulins, Intravenous/isolation & purification , Mice, Transgenic , Microglia/immunology , Parkinson Disease/pathology , Protein Aggregation, Pathological/immunology
19.
Neurobiol Dis ; 121: 131-137, 2019 01.
Article in English | MEDLINE | ID: mdl-30176351

ABSTRACT

Mutations in Cu/Zn superoxide dismutase (SOD1) are the cause of ~20% of cases of familial ALS (FALS), which comprise ~10% of the overall total number of cases of ALS. Mutant (mt) SOD1 is thought to cause FALS through a gain and not loss in function, perhaps as a result of the mutant protein's misfolding and aggregation. Previously we used a phage display library to raise single chain variable fragment antibodies (scFvs) against SOD1, which were found to decrease aggregation of mtSOD1 and toxicity in vitro. In the present study, we show that two scFvs directed against SOD1 ameliorate disease in G93A mtSOD1 transgenic mice and also decrease motor neuron loss, microgliosis, astrocytosis, as well as SOD1 burden and aggregation. The results suggest that the use of antibodies or antibody mimetics directed against SOD1 may be a useful therapeutic direction in mtSOD1-induced FALS. Since studies suggest that wild type SOD1 may be misfolded similar to that seen with mtSOD1, this therapeutic direction may be effective in sporadic as well as FALS.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Single-Chain Antibodies/administration & dosage , Superoxide Dismutase/immunology , Animals , Disease Models, Animal , Female , Gliosis/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/immunology , Protein Aggregation, Pathological/immunology , Spinal Cord/immunology , Spinal Cord/pathology , Superoxide Dismutase/genetics
20.
Int Immunol ; 31(1): 41-49, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30239735

ABSTRACT

A hapten is a small molecule that is not immunogenic on its own but can stimulate the production of antibodies at the sensitization phase when conjugated to carrier proteins. The hapten then reacts specifically with the antibodies generated against it to elicit an immune or allergic response at the challenge phase. Here, we compared various carrier proteins conjugated with the same hapten in their ability to induce hapten-specific IgE-mediated allergic responses in vitro and in vivo, and characterized the nature of carrier proteins that determines the magnitude of response at the challenge phase of allergic reactions. Hapten 2,4,6-trinitrophenol (TNP)-conjugated ovalbumin (TNP-OVA) and bovine serum albumin (TNP-BSA) elicited TNP-specific, mast cell-dependent, immediate-type allergic reactions at a comparable level in mice that had been passively sensitized with TNP-specific IgE. In contrast, TNP-OVA but not TNP-BSA efficiently induced a basophil-dependent, IgE-mediated chronic allergic inflammation (IgE-CAI), even though both proteins could stimulate basophils in vitro at a comparable level. By comparing different carrier proteins and structurally modifying them, we found that the formation of large aggregates is crucial for TNP-conjugated carrier proteins to efficiently elicit IgE-CAI, regardless of the type of protein. Thus, the aggregation status of carrier proteins appears to determine the magnitude of allergic response at the challenge phase of hapten-specific IgE-CAI. Our findings suggest that the allergenicity of substances is a matter of importance not only at the sensitization but also at the challenge phase in a certain type of allergy including a basophil-mediated allergic inflammation.


Subject(s)
Allergens/immunology , Basophils/immunology , Hypersensitivity/immunology , Protein Aggregates/immunology , Protein Aggregation, Pathological/immunology , Proteins/immunology , Allergens/chemistry , Animals , Basophils/metabolism , Disease Models, Animal , Haptens , Hypersensitivity/diagnosis , Hypersensitivity/metabolism , Immunoglobulin E/immunology , Macrophages/immunology , Macrophages/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mice , Molecular Weight , Proteins/chemistry , Proteins/metabolism , Skin/immunology , Skin/metabolism , Skin/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...