Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Curr Genet ; 67(6): 833-847, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34319422

ABSTRACT

The yeast prions (infectious proteins) [URE3] and [PSI+] are essentially non-functional (or even toxic) amyloid forms of Ure2p and Sup35p, whose normal function is in nitrogen catabolite repression and translation termination, respectively. Yeast has an array of systems working in normal cells that largely block infection with prions, block most prion formation, cure most nascent prions and mitigate the toxic effects of those prions that escape the first three types of systems. Here we review recent progress in defining these anti-prion systems, how they work and how they are regulated. Polymorphisms of the prion domains partially block infection with prions. Ribosome-associated chaperones ensure proper folding of nascent proteins, thus reducing [PSI+] prion formation and curing many [PSI+] variants that do form. Btn2p is a sequestering protein which gathers [URE3] amyloid filaments to one place in the cells so that the prion is often lost by progeny cells. Proteasome impairment produces massive overexpression of Btn2p and paralog Cur1p, resulting in [URE3] curing. Inversely, increased proteasome activity, by derepression of proteasome component gene transcription or by 60S ribosomal subunit gene mutation, prevents prion curing by Btn2p or Cur1p. The nonsense-mediated decay proteins (Upf1,2,3) cure many nascent [PSI+] variants by associating with Sup35p directly. Normal levels of the disaggregating chaperone Hsp104 can also cure many [PSI+] prion variants. By keeping the cellular levels of certain inositol polyphosphates / pyrophosphates low, Siw14p cures certain [PSI+] variants. It is hoped that exploration of the yeast innate immunity to prions will lead to discovery of similar systems in humans.


Subject(s)
Disease Resistance/immunology , Disease Susceptibility , Host-Pathogen Interactions/immunology , Immunity, Innate , Prion Diseases/etiology , Prions/immunology , Amyloid/chemistry , Amyloid/immunology , Amyloid/metabolism , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/immunology , Amyloidogenic Proteins/metabolism , Animals , Autophagy , Disease Susceptibility/immunology , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/immunology , Host-Pathogen Interactions/genetics , Humans , Molecular Chaperones/metabolism , Mutation , Nonsense Mediated mRNA Decay , Prion Diseases/metabolism , Prions/chemistry , Prions/genetics , Prions/metabolism , Protein Binding , Protein Conformation , Protein Folding , Ribosomes/metabolism
2.
Front Immunol ; 12: 638867, 2021.
Article in English | MEDLINE | ID: mdl-33717189

ABSTRACT

The human microbiota is the community of microorganisms that live upon or within their human host. The microbiota consists of various microorganisms including bacteria, fungi, viruses, and archaea; the gut microbiota is comprised mostly of bacteria. Many bacterial species within the gut microbiome grow as biofilms, which are multicellular communities embedded in an extracellular matrix. Studies have shown that the relative abundances of bacterial species, and therefore biofilms and bacterial byproducts, change during progression of a variety of human diseases including gastrointestinal, autoimmune, neurodegenerative, and cancer. Studies have shown the location and proximity of the biofilms within the gastrointestinal tract might impact disease outcome. Gram-negative enteric bacteria secrete the amyloid curli, which makes up as much as 85% of the extracellular matrix of enteric biofilms. Curli mediates cell-cell attachment and attachment to various surfaces including extracellular matrix components such as fibronectin and laminin. Structurally, curli is strikingly similar to pathological and immunomodulatory human amyloids such as amyloid-ß, which has been implicated in Alzheimer's disease, α-synuclein, which is involved in Parkinson's disease, and serum amyloid A, which is secreted during the acute phase of inflammation. The immune system recognizes both bacterial amyloid curli and human amyloids utilizing the same receptors, so curli also induces inflammation. Moreover, recent work indicates that curli can participate in the self-assembly process of pathological human amyloids. Curli is found within biofilms of commensal enteric bacteria as well as invasive pathogens; therefore, evidence suggests that curli contributes to complex human diseases. In this review, we summarize the recent findings on how bacterial biofilms containing curli participate in the pathological and immunological processes in gastrointestinal diseases, systemic autoimmune diseases, and neurodegenerative diseases.


Subject(s)
Bacterial Proteins/immunology , Biofilms , Gastrointestinal Microbiome/immunology , Amyloidogenic Proteins/immunology , Autoimmune Diseases/immunology , Gastrointestinal Diseases/immunology , Humans , Neurodegenerative Diseases/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 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
5.
Trends Immunol ; 40(8): 762-780, 2019 08.
Article in English | MEDLINE | ID: mdl-31320280

ABSTRACT

Amyloid formation contributes to the development of progressive metabolic and neurodegenerative diseases, while also serving functional roles in host defense. Emerging evidence suggests that as amyloidogenic peptides populate distinct aggregation states, they interact with different combinations of pattern recognition receptors (PRRs) to direct the phenotype and function of tissue-resident and infiltrating innate immune cells. We review recent evidence of innate immunomodulation by distinct forms of amyloidogenic peptides produced by mammals (humans, non-human primates), bacteria, and fungi, as well as the corresponding cell-surface and intracellular PRRs in these interactions, in human and mouse models. Our emerging understanding of peptide aggregate-innate immune cell interactions, and the factors regulating the balance between amyloid function and pathogenicity, might aid the development of anti-amyloid and immunomodulating therapies.


Subject(s)
Amyloidogenic Proteins/metabolism , Immunity, Innate , Immunomodulation , Peptides/metabolism , Amyloid/immunology , Amyloid/metabolism , Amyloidogenic Proteins/immunology , Amyloidosis/etiology , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Biomarkers , Disease Susceptibility/immunology , Disease Susceptibility/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Monocytes/immunology , Monocytes/metabolism , Peptides/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction
6.
Biochemistry ; 58(6): 763-775, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30513201

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disorder manifested via chronic inflammation, demyelination, and neurodegeneration inside the central nervous system. The progressive phase of MS is characterized by neurodegeneration, but unlike classical neurodegenerative diseases, amyloid-like aggregation of self-proteins has not been documented. There is evidence that citrullination protects an immunodominant peptide of human myelin oligodendrocyte glycoprotein (MOG34-56) against destructive processing in Epstein-Barr virus-infected B-lymphocytes (EBV-BLCs) in marmosets and causes exacerbation of ongoing MS-like encephalopathies in mice. Here we collected evidence that citrullination of MOG can also lead to amyloid-like behavior shifting the disease pathogenesis toward neurodegeneration. We observed that an immunodominant MOG peptide, MOG35-55, displays amyloid-like behavior upon site-specific citrullination at positions 41, 46, and/or 52. These amyloid aggregates are shown to be toxic to the EBV-BLCs and to dendritic cells at concentrations favored for antigen presentation, suggesting a role of amyloid-like aggregation in the pathogenesis of progressive MS.


Subject(s)
Amyloid/metabolism , Amyloidogenic Proteins/metabolism , B-Lymphocytes/metabolism , Myelin-Oligodendrocyte Glycoprotein/metabolism , Peptide Fragments/metabolism , Amino Acid Sequence , Amyloid/immunology , Amyloid/toxicity , Amyloidogenic Proteins/chemical synthesis , Amyloidogenic Proteins/immunology , Amyloidogenic Proteins/toxicity , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , B-Lymphocytes/virology , Benzothiazoles/chemistry , Callithrix , Cell Line , Citrullination/immunology , Dendritic Cells/metabolism , Herpesvirus 4, Human , Humans , Mice, Inbred C57BL , Multiple Sclerosis, Chronic Progressive/immunology , Multiple Sclerosis, Chronic Progressive/metabolism , Multiple Sclerosis, Chronic Progressive/virology , Myelin-Oligodendrocyte Glycoprotein/chemical synthesis , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Peptide Fragments/toxicity , Protein Aggregation, Pathological , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Front Immunol ; 9: 672, 2018.
Article in English | MEDLINE | ID: mdl-29686672

ABSTRACT

The post-injury inflammatory response is a key mediator in long-term recovery from traumatic brain injury (TBI). Moreover, the immune response to TBI, mediated by microglia and macrophages, is influenced by existing brain pathology and by secondary immune challenges. For example, recent evidence shows that the presence of beta-amyloid and phosphorylated tau protein, two hallmark features of AD that increase during normal aging, substantially alter the macrophage response to TBI. Additional data demonstrate that post-injury microglia are "primed" and become hyper-reactive following a subsequent acute immune challenge thereby worsening recovery. These alterations may increase the incidence of neuropsychiatric complications after TBI and may also increase the frequency of neurodegenerative pathology. Therefore, the purpose of this review is to summarize experimental studies examining the relationship between TBI and development of AD-like pathology with an emphasis on the acute and chronic microglial and macrophage response following injury. Furthermore, studies will be highlighted that examine the degree to which beta-amyloid and tau accumulation as well as pre- and post-injury immune stressors influence outcome after TBI. Collectively, the studies described in this review suggest that the brain's immune response to injury is a key mediator in recovery, and if compromised by previous, coincident, or subsequent immune stressors, post-injury pathology and behavioral recovery will be altered.


Subject(s)
Alzheimer Disease/immunology , Brain Injuries, Traumatic/immunology , Aging/immunology , Amyloidogenic Proteins/immunology , Animals , Humans , Inflammation/immunology , Macrophages/immunology , tau Proteins/immunology
8.
CNS Neurol Disord Drug Targets ; 17(9): 671-679, 2018.
Article in English | MEDLINE | ID: mdl-29546836

ABSTRACT

BACKGROUND & OBJECTIVE: Protein misfolding and aggregation have been considered the common pathological hallmarks for a number of neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). These abnormal proteins aggregates damage mitochondria and induce oxidative stress, resulting in neuronal cell death. Prolonged neuronal damage activates microglia and astrocytes, development of inflammation reaction and further promotes neurodegeneration. Thus, elimination of abnormal protein aggregates without eliciting any adverse effects are the main treatment strategies. To overcome this, recent studies have deployed single- chain fragment variable antibodies (scFvs) to target the pathological protein aggregates, such as amyloid-beta (Aß) peptides, α-synuclein (α-syn) and Huntingtin (Htt). To date scFv has been effective at inhibiting abnormal protein aggregates formation in both in vitro and in vivo model system of AD, PD and HD. CONCLUSION: Currently active research is still ongoing to improve the scFv gene delivery technology, to further enhance brain penetration, intracellular stability, solubility and efficacy of scFv intrabody.


Subject(s)
Neurodegenerative Diseases/drug therapy , Single-Chain Antibodies/therapeutic use , Amyloidogenic Proteins/immunology , Animals , Humans , Huntingtin Protein/immunology , alpha-Synuclein/immunology
9.
J Neuroinflammation ; 15(1): 98, 2018 Mar 28.
Article in English | MEDLINE | ID: mdl-29592816

ABSTRACT

BACKGROUND: Currently, several amyloid beta (Aß) antibodies, including the protofibril selective antibody BAN2401, are in clinical trials. The murine version of BAN2401, mAb158, has previously been shown to lower the levels of pathogenic Aß and prevent Aß deposition in animal models of Alzheimer's disease (AD). However, the cellular mechanisms of the antibody's action remain unknown. We have recently shown that astrocytes effectively engulf Aß42 protofibrils, but store rather than degrade the ingested Aß aggregates. In a co-culture set-up, the incomplete degradation of Aß42 protofibrils by astrocytes results in increased neuronal cell death, due to the release of extracellular vesicles, containing N-truncated, neurotoxic Aß. METHODS: The aim of the present study was to investigate if the accumulation of Aß in astrocytes can be affected by the Aß protofibril selective antibody mAb158. Co-cultures of astrocytes, neurons, and oligodendrocytes, derived from embryonic mouse cortex, were exposed to Aß42 protofibrils in the presence or absence of mAb158. RESULTS: Our results demonstrate that the presence of mAb158 almost abolished Aß accumulation in astrocytes. Consequently, mAb158 treatment rescued neurons from Aß-induced cell death. CONCLUSION: Based on these findings, we conclude that astrocytes may play a central mechanistic role in anti-Aß immunotherapy.


Subject(s)
Amyloidogenic Proteins , Antibodies, Monoclonal/pharmacology , Cell Death/drug effects , Neuroglia/drug effects , Neurons/drug effects , Amyloidogenic Proteins/immunology , Amyloidogenic Proteins/metabolism , Amyloidogenic Proteins/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred C57BL
10.
Med Hypotheses ; 110: 60-63, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29317070

ABSTRACT

The cause and therapy of neurodegenerative diseases remain unsolved puzzles. These diseases are correlated with presence of beta sheet-rich amyloid assemblies. Here, I derive and assemble puzzle pieces to obtain a loose end-tying hypothesis for cause with direct implications for therapy. I use the following extrapolations to find connectable puzzle pieces: (a) the traditional extrapolation that amyloid/amyloid precursors cause disease, (b) a recent extrapolation that amyloid-forming proteins, some of which are virus protein homologs, are components of an empirically obscure innate immune system that counters insults, including those by both viruses and bacteria, (c) a new extrapolation that various insults produce assemblies with structural features in common and that amyloid-forming, innate immune system proteins recognize these features and, then, counter insults by co-assembly, (d, 1) a second new extrapolation that beta sheet is a common structural feature and is extended during insult-neutralizing co-assembly and (d, 2) an appendix, derived from studies of phages T3 and T4, that most insult-produced assemblies are obscure to current biochemical analysis. The hypothesis is the following. One function of amyloid-forming proteins is non-classical innate immunity to biological insults. This immunity works via beta sheet-extending co-assembly of amyloid-forming proteins with beta sheet-containing insult products. For example, co-assembly with beta sheet-containing viral assembly intermediates inhibits virus production. Amyloid-forming proteins cause neurodegenerative disease when errant, typically overproduced. Other innate immunity systems sometimes exacerbate symptoms. This hypothesis suggests the following therapy, based on manipulating Nature's chemistry. First, conduct directed evolution to obtain low-pathogenicity, chronic symptom-producing viruses with assembly intermediates that co-assemble with and destabilize both amyloid and amyloid sub-assemblies. Then, infect patients with these viruses.


Subject(s)
Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/therapy , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/immunology , Animals , Biological Evolution , Humans , Immunity, Innate , Models, Neurological , Neurodegenerative Diseases/immunology , Protein Conformation, beta-Strand , Viral Proteins/immunology
11.
Sci Rep ; 7(1): 9881, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28852189

ABSTRACT

We describe a novel approach to produce conformational monoclonal antibodies selected to specifically react with the ß-sheet secondary structure of pathological oligomeric conformers, characteristic of many neurodegenerative diseases. Contrary to past and current efforts, we utilize a mammalian non-self-antigen as an immunogen. The small, non-self peptide selected was covalently polymerized with glutaraldehyde until it reached a high ß-sheet secondary structure content, and species between 10-100kDa that are immunogenic, stable and soluble (p13Bri). Inoculation of p13Bri in mice elicited antibodies to the peptide and the ß-sheet secondary structure conformation. Hybridomas were produced and clones selected for their reactivity with at least two different oligomeric conformers from Alzheimer's, Parkinson and/or Prion diseases. The resulting conformational monoclonals are able to detect pathological oligomeric forms in different human neurodegenerative diseases by ELISA, immunohistochemistry and immunoblots. This technological approach may be useful to develop tools for detection, monitoring and treatment of multiple misfolding disorders.


Subject(s)
Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/immunology , Antibodies, Monoclonal/immunology , Protein Conformation, beta-Strand , Protein Multimerization , Amyloidogenic Proteins/metabolism , Amyloidogenic Proteins/ultrastructure , Animals , Humans , Mice , Neurodegenerative Diseases/immunology , Protein Aggregation, Pathological
12.
Int Immunopharmacol ; 49: 155-160, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28595078

ABSTRACT

Neuroinflammation has been shown as an essential factor in the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and Multiple Sclerosis. Furthermore, activated microglia and increased pro-inflammatory cytokines are the major hallmarks in neurodegenerative diseases. A multimolecular complex named as inflammasome is involved in the process of inflammatory response, which can activate inflammatory caspases, leading to the cleavage and secretion of inflammatory cytokines, and finally generates a potent inflammatory response. In neurodegenerative diseases, it has been widely assumed that some types of amyloid proteins might be the triggers to activate the NLRP3 inflammasome. In this review, we summarize the current researches about the role of NLRP3 inflammasome, by reviewing the main studies in vitro and in vivo experiments and discuss the potential for new therapeutic interventions in neurodegenerative diseases.


Subject(s)
Amyloidogenic Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurodegenerative Diseases/immunology , Amyloidogenic Proteins/immunology , Animals , Cytokines/metabolism , Humans , Inflammation
13.
J Neurosci ; 37(20): 5155-5171, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28442538

ABSTRACT

Increasing evidence has suggested that systemic inflammation along with local brain inflammation can play a significant role in Alzheimer's disease (AD) pathogenesis. Identifying key molecules that regulate the crosstalk between the immune and the CNS can provide potential therapeutic targets. TNF-α is a proinflammatory cytokine implicated in the pathogenesis of systemic inflammatory and neurodegenerative diseases, such as rheumatoid arthritis (RA) and AD. Recent studies have reported that anti-TNF-α therapy or RA itself can modulate AD pathology, although the underlying mechanism is unclear. To investigate the role of peripheral TNF-α as a mediator of RA in the pathogenesis of AD, we generated double-transgenic 5XFAD/Tg197 AD/TNF mice that develop amyloid deposits and inflammatory arthritis induced by human TNF-α (huTNF-α) expression. We found that 5XFAD/Tg197 mice display decreased amyloid deposition, compromised neuronal integrity, and robust brain inflammation characterized by extensive gliosis and elevated blood-derived immune cell populations, including phagocytic macrophages and microglia. To evaluate the contribution of peripheral huTNF-α in the observed brain phenotype, we treated 5XFAD/Tg197 mice systemically with infliximab, an anti-huTNF-α antibody that does not penetrate the blood-brain barrier and prevents arthritis. Peripheral inhibition of huTNF-α increases amyloid deposition, rescues neuronal impairment, and suppresses gliosis and recruitment of blood-derived immune cells, without affecting brain huTNF-α levels. Our data report, for the first time, a distinctive role for peripheral TNF-α in the modulation of the amyloid phenotype in mice by regulating blood-derived and local brain inflammatory cell populations involved in ß-amyloid clearance.SIGNIFICANCE STATEMENT Mounting evidence supports the active involvement of systemic inflammation, in addition to local brain inflammation, in Alzheimer's disease (AD) progression. TNF-α is a pluripotent cytokine that has been independently involved in the pathogenesis of systemic inflammatory rheumatoid arthritis (RA) and AD. Here we first demonstrate that manipulation of peripheral TNF-α in the context of arthritis modulates the amyloid phenotype by regulating immune cell trafficking in the mouse brain. Our study suggests that additionally to its local actions in the AD brain, TNF-α can also indirectly modulate amyloid pathology as a regulator of peripheral inflammation. Our findings may have significant implications in the treatment of RA patients with anti-TNF-α drugs and in the potential use of TNF-targeted therapies for AD.


Subject(s)
Alzheimer Disease/immunology , Amyloidogenic Proteins/immunology , Arthritis, Rheumatoid/immunology , Brain/immunology , Macrophages/immunology , Neuroglia/immunology , Tumor Necrosis Factor-alpha/immunology , Alzheimer Disease/complications , Alzheimer Disease/pathology , Animals , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/pathology , Brain/pathology , Cytokines/immunology , Female , Immunologic Factors/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/pathology
15.
ACS Chem Neurosci ; 8(3): 454-459, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28292186

ABSTRACT

Clearance of amyloid ß (Aß) by immunotherapy is one of the fancy methods to treat Alzheimer's disease (AD). However, the failure of some clinical trials suggested that there may be something ignored in the past development of immunotherapy. Pyroglutamate-3 Aß (AßpE3-X), which was found to be abundant in the patients' brain, has attracted much attention after the report that AßpE3-42 could serve as a template to exacerbate the aggregation of Aß. In addition, AßpE3-X could not be recognized by the antibodies targeting the N-terminus of Aß, suggesting that AßpE3-X maybe the ignored one. Indeed, passive immunization targeting AßpE3-X has shown some beneficial results, while active immunotherapy has not been extensively studied. In the present study, we designed and synthesized a novel peptide vaccine targeting AßpE3-X, which contains AßpE3-15 as B cell epitope and P2 as T cell epitope. We showed that this vaccine could induce strong antibody response to AßpE3-X. We also showed that prophylactic immunization of AD model mice with our vaccine could reduce Aß plaques and rescue cognitive decline. This new kind of Aß vaccine will open up new directions for AD immunotherapy.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/immunology , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/therapeutic use , Cognition Disorders/prevention & control , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloidogenic Proteins/immunology , Animals , Antibodies/blood , Brain/metabolism , Brain/pathology , Cognition Disorders/pathology , Disease Models, Animal , Humans , Maze Learning/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/drug therapy , Presenilin-1/genetics , Reaction Time/drug effects , Reaction Time/physiology
16.
PLoS One ; 12(3): e0174152, 2017.
Article in English | MEDLINE | ID: mdl-28350808

ABSTRACT

BACKGROUND: Monoclonal free light chain (LC) proteins are present in the circulation of patients with immunoproliferative disorders such as light chain (AL) amyloidosis and multiple myeloma (MM). Light chain-associated amyloid is a complex pathology composed of proteinaceous fibrils and extracellular matrix proteins found in all patients with AL and in ~10-30% of patients who presented with MM. Amyloid deposits systemically in multiple organs and tissues leading to dysfunction and ultimately death. The overall survival of patients with amyloidosis is worse than for those with early stage MM. METHODS AND FINDINGS: We have developed a sensitive binding assay quantifying the recruitment of full length, patient-derived LC proteins by synthetic amyloid fibrils, as a method for studying their amyloidogenic potential. In a survey of eight urinary LC, both AL and MM-associated proteins were recruited by synthetic amyloid fibrils; however, AL-associated LC bound significantly more efficiently (p < 0.05) than did MM LCs. The LC proteins used in this study were isolated from urine and presumed to represent a surrogate of serum free light chains. CONCLUSION: The binding of LC to synthetic fibrils in this assay accurately differentiated LC with amyloidogenic propensity from MM LC that were not associated with clinical amyloid disease. Notably, the LC from a MM patient who subsequently developed amyloid behaved as an AL-associated protein in the assay, indicating the possibility for identifying MM patients at risk for developing amyloidosis based on the light chain recruitment efficacy. With this information, at risk patients can be monitored more closely for the development of amyloidosis, allowing timely administration of novel, amyloid-directed immunotherapies-this approach may improve the prognosis for these patients.


Subject(s)
Amyloid/immunology , Amyloidogenic Proteins/immunology , Amyloidosis/immunology , Immunoglobulin Light Chains/immunology , Multiple Myeloma/immunology , Adult , Aged , Aged, 80 and over , Amyloid/metabolism , Amyloid/ultrastructure , Amyloidogenic Proteins/metabolism , Amyloidosis/metabolism , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Humans , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light-chain Amyloidosis , Male , Microscopy, Immunoelectron , Middle Aged , Multiple Myeloma/metabolism , Prognosis , Protein Binding , Protein Multimerization/drug effects , Surface Plasmon Resonance , Thermodynamics , Urea/pharmacology
17.
PLoS One ; 11(11): e0165964, 2016.
Article in English | MEDLINE | ID: mdl-27824888

ABSTRACT

Misfolding, abnormal accumulation, and secretion of α-Synuclein (α-Syn) are closely associated with synucleinopathies, including Parkinson's disease (PD). VH14 is a human single domain intrabody selected against the non-amyloid component (NAC) hydrophobic interaction region of α-Syn, which is critical for initial aggregation. Using neuronal cell lines, we show that as a bifunctional nanobody fused to a proteasome targeting signal, VH14PEST can counteract heterologous proteostatic effects of mutant α-Syn on mutant huntingtin Exon1 and protect against α-Syn toxicity using propidium iodide or Annexin V readouts. We compared this anti-NAC candidate to NbSyn87, which binds to the C-terminus of α-Syn. NbSyn87PEST degrades α-Syn as well or better than VH14PEST. However, while both candidates reduced toxicity, VH14PEST appears more effective in both proteostatic stress and toxicity assays. These results show that the approach of reducing intracellular monomeric targets with novel antibody engineering technology should allow in vivo modulation of proteostatic pathologies.


Subject(s)
Amyloidogenic Proteins/immunology , alpha-Synuclein/chemistry , Animals , Blotting, Western , Cell Line , Flow Cytometry , Humans , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Rats , Single-Domain Antibodies , Stem Cells , Transfection , alpha-Synuclein/immunology , alpha-Synuclein/physiology
18.
Amyloid ; 23(3): 168-177, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27494229

ABSTRACT

Amyloid light chain (AL) amyloidosis is characterized by misfolded light chain (LC) (amyloid) deposition in various peripheral organs, leading to progressive dysfunction and death. There are no regulatory agency-approved treatments for AL amyloidosis, and none of the available standard of care approaches directly targets the LC protein that constitutes the amyloid. NEOD001, currently in late-stage clinical trials, is a conformation-specific, anti-LC antibody designed to specifically target misfolded LC aggregates and promote phagocytic clearance of AL amyloid deposits. The present study demonstrated that the monoclonal antibody 2A4, the murine form of NEOD001, binds to patient-derived soluble and insoluble LC aggregates and induces phagocytic clearance of AL amyloid in vitro. 2A4 specifically labeled all 21 fresh-frozen organ samples studied, which were derived from 10 patients representing both κ and λ LC amyloidosis subtypes. 2A4 immunoreactivity largely overlapped with thioflavin T-positive labeling, and 2A4 bound both soluble and insoluble LC aggregates extracted from patient tissue. Finally, 2A4 induced macrophage engagement and phagocytic clearance of AL amyloid deposits in vitro. These findings provide further evidence that 2A4/NEOD001 can effectively clear and remove human AL-amyloid from tissue and further support the rationale for the evaluation of NEOD001 in patients with AL amyloidosis.


Subject(s)
Amyloidogenic Proteins/immunology , Amyloidosis/immunology , Antibodies, Monoclonal/chemistry , Antigen-Antibody Complex/chemistry , Immunoglobulin Light Chains/chemistry , Phagocytosis , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/isolation & purification , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Antibodies, Monoclonal/biosynthesis , Benzothiazoles , Cell Line , Humans , Immunoglobulin Light Chains/isolation & purification , Mice , Monocytes/cytology , Monocytes/immunology , Protein Aggregates/immunology , Protein Binding , Staining and Labeling/methods , Thiazoles/chemistry
19.
Sci Rep ; 6: 22258, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26924748

ABSTRACT

Why do patients suffering from neurodegenerative diseases generate autoantibodies that selectively bind soluble aggregates of amyloidogenic proteins? Presently, molecular basis of interactions between the soluble aggregates and human immune system is unknown. By analyzing sequences of experimentally validated T-cell autoimmune epitopes, aggregating peptides, amyloidogenic proteins and randomly generated peptides, here we report overlapping regions that likely drive aggregation as well as generate autoantibodies against the aggregates. Sequence features, that make short peptides susceptible to aggregation, increase their incidence in human T-cell autoimmune epitopes by 4-6 times. Many epitopes are predicted to be significantly aggregation prone (aggregation propensities ≥10%) and the ones containing experimentally validated aggregating regions are enriched in hydrophobicity by 10-20%. Aggregate morphologies also influence Human Leukocyte Antigen (HLA)--types recognized by the aggregating regions containing epitopes. Most (88%) epitopes that contain amyloid fibril forming regions bind HLA-DR, while majority (63%) of those containing amorphous ß-aggregating regions bind HLA-DQ. More than two-thirds (70%) of human amyloidogenic proteins contain overlapping regions that are simultaneously aggregation prone and auto-immunogenic. Such regions help clear soluble aggregates by generating selective autoantibodies against them. This can be harnessed for early diagnosis of proteinopathies and for drug/vaccine design against them.


Subject(s)
Amyloidogenic Proteins/immunology , Amyloidogenic Proteins/metabolism , Autoimmunity , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , Protein Aggregates/immunology , Protein Aggregation, Pathological/immunology , Protein Aggregation, Pathological/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amyloidogenic Proteins/chemistry , Conserved Sequence , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Position-Specific Scoring Matrices , Protein Conformation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
20.
Prog Histochem Cytochem ; 51(1): 1-8, 2016 05.
Article in English | MEDLINE | ID: mdl-26851150

ABSTRACT

This review discussed the importance of mutated tau, amyloid and neuroinflammatory factors and microglia in Alzheimer disease. In particular tau, CD4 and TNF alpha were included in the review and the colocalizations of these factors were highlighted. It is important to realize the Alzheimer disease may result from the interactions of these factors. Some of these factors may coexist at the same region and at the same time e.g. mutated tau and amyloid in plaques. A summary scheme of etiology leading to the disease was included.


Subject(s)
Alzheimer Disease/genetics , Amyloidogenic Proteins/genetics , Mutation , Plaque, Amyloid/genetics , tau Proteins/genetics , Alzheimer Disease/diagnosis , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloidogenic Proteins/immunology , Animals , Brain/immunology , Brain/pathology , CD4 Antigens/genetics , CD4 Antigens/immunology , Cell Death , Gene Expression , Humans , Inflammation , Microglia/immunology , Microglia/pathology , Plaque, Amyloid/diagnosis , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , tau Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL