Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 253
1.
Brief Bioinform ; 23(5)2022 09 20.
Article En | MEDLINE | ID: mdl-35595541

Proteins/peptides have shown to be promising therapeutic agents for a variety of diseases. However, toxicity is one of the obstacles in protein/peptide-based therapy. The current study describes a web-based tool, ToxinPred2, developed for predicting the toxicity of proteins. This is an update of ToxinPred developed mainly for predicting toxicity of peptides and small proteins. The method has been trained, tested and evaluated on three datasets curated from the recent release of the SwissProt. To provide unbiased evaluation, we performed internal validation on 80% of the data and external validation on the remaining 20% of data. We have implemented the following techniques for predicting protein toxicity; (i) Basic Local Alignment Search Tool-based similarity, (ii) Motif-EmeRging and with Classes-Identification-based motif search and (iii) Prediction models. Similarity and motif-based techniques achieved a high probability of correct prediction with poor sensitivity/coverage, whereas models based on machine-learning techniques achieved balance sensitivity and specificity with reasonably high accuracy. Finally, we developed a hybrid method that combined all three approaches and achieved a maximum area under receiver operating characteristic curve around 0.99 with Matthews correlation coefficient 0.91 on the validation dataset. In addition, we developed models on alternate and realistic datasets. The best machine learning models have been implemented in the web server named 'ToxinPred2', which is available at https://webs.iiitd.edu.in/raghava/toxinpred2/ and a standalone version at https://github.com/raghavagps/toxinpred2. This is a general method developed for predicting the toxicity of proteins regardless of their source of origin.


Proteins , Software , Databases, Protein , Machine Learning , Peptides , Proteins/toxicity
2.
Molecules ; 26(8)2021 Apr 09.
Article En | MEDLINE | ID: mdl-33918763

Three-finger toxins are naturally occurring proteins in Elapidae snake venoms. Nowadays, they are gaining popularity because of their therapeutic potential. On the other hand, these proteins may cause undesirable reactions inside the body's cells. A full assessment of the safety of Naja ashei venom components for human cell application is still unknown. The aim of the study was to determine the effect of the exogenous application of three-finger toxins on the cells of monocytes (U-937) and promyelocytes (HL-60), with particular emphasis on the modification of their membranes under the influence of various doses of 3FTx protein fraction (0-120 ng/mL). The fraction exhibiting the highest proportion of 3FTx proteins after size exclusion chromatography (SEC) separation was used in the experiments. The structural response of cell membranes was described on the basis of single-component and multi-component Langmuir monolayers that mimicked the native membranes. The results show that the mechanism of protein-lipid interactions depends on both the presence of lipid polar parts (especially zwitterionic type of lipids) and the degree of membrane saturation (the greatest-for unsaturated lipids). The biochemical indicators reflecting the tested cells (MDA, LDH, cell survival, induction of inflammation, LD50) proved the results that were obtained for the model.


Elapid Venoms/chemistry , Elapid Venoms/toxicity , Membranes, Artificial , Naja/metabolism , Proteins/toxicity , Animals , Chemical Fractionation , Chromatography, Gel , Female , HL-60 Cells , Humans , L-Lactate Dehydrogenase/metabolism , Lethal Dose 50 , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Membranes , Pressure , Temperature , U937 Cells
3.
Nat Cell Biol ; 23(2): 127-135, 2021 02.
Article En | MEDLINE | ID: mdl-33495632

Ribosomes are multicomponent molecular machines that synthesize all of the proteins of living cells. Most of the genes that encode the protein components of ribosomes are therefore essential. A reduction in gene dosage is often viable albeit deleterious and is associated with human syndromes, which are collectively known as ribosomopathies1-3. The cell biological basis of these pathologies has remained unclear. Here, we model human ribosomopathies in Drosophila and find widespread apoptosis and cellular stress in the resulting animals. This is not caused by insufficient protein synthesis, as reasonably expected. Instead, ribosomal protein deficiency elicits proteotoxic stress, which we suggest is caused by the accumulation of misfolded proteins that overwhelm the protein degradation machinery. We find that dampening the integrated stress response4 or autophagy increases the harm inflicted by ribosomal protein deficiency, suggesting that these activities could be cytoprotective. Inhibition of TOR activity-which decreases ribosomal protein production, slows down protein synthesis and stimulates autophagy5-reduces proteotoxic stress in our ribosomopathy model. Interventions that stimulate autophagy, combined with means of boosting protein quality control, could form the basis of a therapeutic strategy for this class of diseases.


Mutation/genetics , Proteins/toxicity , Ribosomes/genetics , Ribosomes/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Alleles , Animals , Apoptosis/drug effects , Autophagy/drug effects , Drosophila melanogaster/drug effects , Drosophila melanogaster/metabolism , HEK293 Cells , Heterozygote , Humans , Imaginal Discs/drug effects , Imaginal Discs/metabolism , Protein Aggregates/drug effects , Protein Biosynthesis/drug effects , Proteomics , Ribosomal Proteins/biosynthesis , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Wings, Animal/drug effects , Wings, Animal/metabolism
4.
Nat Cell Biol ; 23(2): 136-146, 2021 02.
Article En | MEDLINE | ID: mdl-33495633

Cell competition allows winner cells to eliminate less fit loser cells in tissues. In Minute cell competition, cells with a heterozygous mutation in ribosome genes, such as RpS3+/- cells, are eliminated by wild-type cells. How cells are primed as losers is partially understood and it has been proposed that reduced translation underpins the loser status of ribosome mutant, or Minute, cells. Here, using Drosophila, we show that reduced translation does not cause cell competition. Instead, we identify proteotoxic stress as the underlying cause of the loser status for Minute competition and competition induced by mahjong, an unrelated loser gene. RpS3+/- cells exhibit reduced autophagic and proteasomal flux, accumulate protein aggregates and can be rescued from competition by improving their proteostasis. Conversely, inducing proteotoxic stress is sufficient to turn otherwise wild-type cells into losers. Thus, we propose that tissues may preserve their health through a proteostasis-based mechanism of cell competition and cell selection.


Cell Competition , Drosophila melanogaster/cytology , Proteins/toxicity , Stress, Physiological , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Competition/drug effects , Drosophila melanogaster/drug effects , Drosophila melanogaster/ultrastructure , Green Fluorescent Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Aggregates , Protein Biosynthesis/drug effects , Proteostasis/drug effects , Ribosomal Proteins/metabolism , Stress, Physiological/drug effects
5.
J Mater Chem B ; 8(40): 9258-9268, 2020 10 21.
Article En | MEDLINE | ID: mdl-32969459

Herein, the synthesis of an amino-acid-based di-block copolymer (di-BCP) in-between an l-glutamic acid-5-benzyl ester and l-aspartic acid-4-benzyl ester [(l-GluA-5-BE)-b-(l-AspA-4-BE)] has been reported. However, the synthesis of di-BCP of [(l-GluA-5-BE)-b-(l-AspA-4-BE)] was carried out through the facile modified ring-opening polymerization (ROP) without using any surfactants and harmful chemicals. Interestingly, the synthesized [(l-GluA-5-BE)-b-(l-AspA-4-BE)] has been used to design nanoflower capsules (NFCs) with surface-functionalized nanoflakes and petals. Notably, the simple solvent propanol has been used as a dispersing medium for the di-BCP-based powder to observe morphology of NFCs. Moreover, these amino-acid-based NFCs are biocompatible, biodegradable, and bio-safe for mankind usage. Consequently, di-BCP-based NFCs show changes in morphology with different temperature conditions, i.e., at ∼10 °C, ∼25 °C (RT), and ∼37 °C (body temperature). Furthermore, the average thickness of the surface-functionalized nanopetals has been calculated as ∼324 nm (in diameter). Similarly, the average distance between petals is calculated as 3.6 µm and the pore depth is ∼21 nm. Additionally, the porosity throughout the surface of capsules in-between nanopetals is an advantageous characteristic feature to improve the drug/paclitaxel (PTX) loading capacity. It is a unique and novel approach to design NFCs, which are a potential payload for nanomedicine and cancer therapy. Furthermore, NFCs were used to evaluate the loading efficacy of drugs and showed ∼78% (wt/wt%) of the PTX loading. Moreover, NFCs showed ∼74% drug release at physiological body temperature. Thus, NFCs showed remarkable release at acidic pH medium. However, PTX released from NFCs showed greater cell inhibition (i.e., ∼79%) with an increase of the PTX concentration after 24 h incubation over HeLa (human epithelial cervical cancer) cells. Besides, PTX released from NFC showed significant (∼34%) cell killing capacity. Such promising NFCs are recommended for breast, liver, and lung cancer therapeutics.


Antineoplastic Agents, Phytogenic/therapeutic use , Drug Carriers/chemistry , Nanostructures/chemistry , Neoplasms/drug therapy , Paclitaxel/therapeutic use , Proteins/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Drug Carriers/toxicity , Drug Liberation , HEK293 Cells , HeLa Cells , Humans , Hydrogen-Ion Concentration , Nanostructures/toxicity , Paclitaxel/chemistry , Porosity , Proteins/toxicity , Rats
6.
Int J Mol Sci ; 21(16)2020 Aug 06.
Article En | MEDLINE | ID: mdl-32781742

Neurodegenerative diseases are a major burden for our society, affecting millions of people worldwide. A main goal of past and current research is to enhance our understanding of the mechanisms underlying proteotoxicity, a common theme among these incurable and debilitating conditions. Cell proteome alteration is considered to be one of the main driving forces that triggers neurodegeneration, and unraveling the biological complexity behind the affected molecular pathways constitutes a daunting challenge. This review summarizes the current state on key processes that lead to cellular proteotoxicity in Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, providing a comprehensive landscape of recent literature. A foundational understanding of how proteotoxicity affects disease etiology and progression may provide essential insight towards potential targets amenable of therapeutic intervention.


Neurodegenerative Diseases/pathology , Proteins/toxicity , Animals , Humans , Neurodegenerative Diseases/drug therapy , Protein Aggregates
7.
Nucleic Acids Res ; 48(10): 5235-5253, 2020 06 04.
Article En | MEDLINE | ID: mdl-32356888

Antisense oligonucleotides (ASOs) interact with target RNAs via hybridization to modulate gene expression through different mechanisms. ASO therapeutics are chemically modified and include phosphorothioate (PS) backbone modifications and different ribose and base modifications to improve pharmacological properties. Modified PS ASOs display better binding affinity to the target RNAs and increased binding to proteins. Moreover, PS ASO protein interactions can affect many aspects of their performance, including distribution and tissue delivery, cellular uptake, intracellular trafficking, potency and toxicity. In this review, we summarize recent progress in understanding PS ASO protein interactions, highlighting the proteins with which PS ASOs interact, the influence of PS ASO protein interactions on ASO performance, and the structure activity relationships of PS ASO modification and protein interactions. A detailed understanding of these interactions can aid in the design of safer and more potent ASO drugs, as illustrated by recent findings that altering ASO chemical modifications dramatically improves therapeutic index.


Phosphorothioate Oligonucleotides/chemistry , Proteins/chemistry , Cell Membrane/chemistry , Cell Membrane/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Intracellular Space/chemistry , Intracellular Space/metabolism , Ligands , Phosphorothioate Oligonucleotides/metabolism , Phosphorothioate Oligonucleotides/pharmacology , Phosphorothioate Oligonucleotides/toxicity , Protein Binding , Protein Domains , Proteins/metabolism , Proteins/toxicity , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonuclease H/chemistry , Ribonuclease H/metabolism , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/metabolism
8.
EMBO J ; 39(4): e100574, 2020 02 17.
Article En | MEDLINE | ID: mdl-31930538

Expansion of G4C2 repeats within the C9ORF72 gene is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Such repeats lead to decreased expression of the autophagy regulator C9ORF72 protein. Furthermore, sense and antisense repeats are translated into toxic dipeptide repeat (DPR) proteins. It is unclear how these repeats are translated, and in which way their translation and the reduced expression of C9ORF72 modulate repeat toxicity. Here, we found that sense and antisense repeats are translated upon initiation at canonical AUG or near-cognate start codons, resulting in polyGA-, polyPG-, and to a lesser degree polyGR-DPR proteins. However, accumulation of these proteins is prevented by autophagy. Importantly, reduced C9ORF72 levels lead to suboptimal autophagy, thereby impairing clearance of DPR proteins and causing their toxic accumulation, ultimately resulting in neuronal cell death. Of clinical importance, pharmacological compounds activating autophagy can prevent neuronal cell death caused by DPR proteins accumulation. These results suggest the existence of a double-hit pathogenic mechanism in ALS/FTD, whereby reduced expression of C9ORF72 synergizes with DPR protein accumulation and toxicity.


Amyotrophic Lateral Sclerosis/genetics , Autophagy , C9orf72 Protein/genetics , Dipeptides/toxicity , Frontotemporal Dementia/genetics , Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/metabolism , Cell Death , DNA Repeat Expansion , Dipeptides/genetics , Frontotemporal Dementia/pathology , HEK293 Cells , Humans , Neurons/pathology , Proteins/genetics , Proteins/toxicity
9.
Chemosphere ; 245: 125624, 2020 Apr.
Article En | MEDLINE | ID: mdl-31864050

Once nanoparticles (NPs) contact with the biological fluids, the proteins immediately adsorb onto their surface, forming a layer called protein corona (PC), which bestows the biological identity on NPs. Importantly, the NPs-PC complex is the true identity of NPs in physiological environment. Based on the affinity and the binding and dissociation rate, PC is classified into soft protein corona, hard protein corona, and interfacial protein corona. Especially, the hard PC, a protein layer relatively stable and closer to their surface, plays particularly important role in the biological effects of the complex. However, the abundant corona proteins rarely correspond to the most abundant proteins found in biological fluids. The composition profile, formation and conformational change of PC can be affected by many factors. Here, the influence factors, not only the nature of NPs, but also surface chemistry and biological medium, are discussed. Likewise, the formed PC influences the interaction between NPs and cells, and the associated subsequent cellular uptake and cytotoxicity. The uncontrolled PC formation may induce undesirable and sometimes opposite results: increasing or inhibiting cellular uptake, hindering active targeting or contributing to passive targeting, mitigating or aggravating cytotoxicity, and stimulating or mitigating the immune response. In the present review, we discuss these aspects and hope to provide a valuable reference for controlling protein adsorption, predicting their behavior in vivo experiments and designing lower toxicity and enhanced targeting nanomedical materials for nanomedicine.


Cells/chemistry , Nanoparticles/chemistry , Protein Corona/chemistry , Adsorption , Animals , Cells/drug effects , Hardness , Humans , Nanoparticles/toxicity , Proteins/chemistry , Proteins/toxicity
10.
Sci Rep ; 9(1): 17923, 2019 11 29.
Article En | MEDLINE | ID: mdl-31784686

With advancements in synthetic biology, the cost and the time needed for designing and synthesizing customized gene products have been steadily decreasing. Many research laboratories in academia as well as industry routinely create genetically engineered proteins as a part of their research activities. However, manipulation of protein sequences could result in unintentional production of toxic proteins. Therefore, being able to identify the toxicity of a protein before the synthesis would reduce the risk of potential hazards. Existing methods are too specific, which limits their application. Here, we extended general function prediction methods for predicting the toxicity of proteins. Protein function prediction methods have been actively studied in the bioinformatics community and have shown significant improvement over the last decade. We have previously developed successful function prediction methods, which were shown to be among top-performing methods in the community-wide functional annotation experiment, CAFA. Based on our function prediction method, we developed a neural network model, named NNTox, which uses predicted GO terms for a target protein to further predict the possibility of the protein being toxic. We have also developed a multi-label model, which can predict the specific toxicity type of the query sequence. Together, this work analyses the relationship between GO terms and protein toxicity and builds predictor models of protein toxicity.


Neural Networks, Computer , Proteins/chemistry , Sequence Analysis, Protein/methods , Toxins, Biological/chemistry , Animals , Gene Ontology , Humans , Proteins/genetics , Proteins/toxicity , Software , Toxins, Biological/genetics , Toxins, Biological/toxicity
11.
Toxicol Lett ; 317: 120-129, 2019 Dec 15.
Article En | MEDLINE | ID: mdl-31580884

PEGylation is considered a safe mechanism to enhance the pharmacokinetics (PK) and pharmacodynamics (PD) of biotherapeutics. Previous studies using PEGylation as a PK enhancement tool have reported benign PEG-related vacuolation in multiple tissues. This paper establishes a threshold for PEG burden beyond which there are alterations in tissue architecture that could potentially lead to dysfunction. As part of the nonclinical safety assessment of Compound A, a 12 kDa protein conjugated to a 40 kDa branched PEG molecule, monkeys were dosed subcutaneously twice weekly for 3 months at protein doses resulting in weekly PEG doses of 8, 24, 120, or 160 mg/kg. Consistent with previous reports with PEGylated biomolecules, Compound A administration resulted in intracellular vacuoles attributed to the PEG moiety in macrophages in numerous tissues and epithelial cells in the choroid plexus and kidney. Vacuolation occurred at all doses with dose-dependent severity and no evidence of recovery up to 2 months after dosing cessation. The vacuolation was considered nonadverse at PEG doses ≤120 mg/kg/week. However, at 160 mg/kg/week PEG, the vacuolation in choroid plexus, pituitary gland, kidney, and choroid of the eye was considered adverse due to significant alterations of tissue architecture that raised concern for the possibility of compromised tissue function. To our knowledge, this is the first report of potentially adverse cellular consequences of PEG accumulation in tissues other than kidney. Furthermore, the lack of reversibility of vacuolation coupled with the lack of a biomarker for intracellular PEG accumulation highlights a potential risk that should be weighed against the benefits of PK/PD enhancement for long-term administration of PEGylated compounds at high doses.


Epithelial Cells/drug effects , Macrophages/drug effects , Polyethylene Glycols/toxicity , Proteins/toxicity , Vacuoles/drug effects , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Compounding , Epithelial Cells/pathology , Female , Injections, Subcutaneous , Macaca fascicularis , Macrophages/pathology , Male , Polyethylene Glycols/administration & dosage , Proteins/administration & dosage , Risk Assessment , Time Factors , Vacuoles/pathology
12.
Food Chem Toxicol ; 132: 110666, 2019 Oct.
Article En | MEDLINE | ID: mdl-31288052

Recent studies suggest human-derived intestinal epithelial cell (IEC) lines cultured as polarized monolayers on permeable Transwell® filters are effective at differentiating between hazardous and non-hazardous proteins following a single exposure. In this study, IEC polarized monolayers were subjected to hazardous or non-hazardous proteins in nine exposures over 30 days and compared to a single exposure of the same protein. The objective was to evaluate whether repeated exposures to a protein differently alter barrier integrity or compromise cell viability compared to single exposures. Proteins tested included Clostridium difficile toxin A, Streptolysin O, Wheat Germ Agglutinin, Phaseolus vulgaris Hemagglutinin-E, bovine serum albumin, porcine serum albumin, and fibronectin. Evidence of diminished barrier integrity and/or cell viability following exposure to hazardous proteins was more pronounced in magnitude when IECs were subjected to multiple rather than single exposures. In some cases, an effect on IEC monolayers was observed only with repeated exposures. In general, IEC responses to non-hazardous proteins following either single or repeated exposures were minimal. Results from these studies support the utility of using cultured human IEC polarized monolayers to differentiate between hazardous and non-hazardous proteins and suggest that repeated exposures may reveal a greater magnitude of response when compared to single exposures.


Intestinal Mucosa/pathology , Proteins/toxicity , Cell Line, Tumor , Epithelial Cells/metabolism , Humans , In Vitro Techniques , Intestinal Mucosa/metabolism
13.
Autophagy ; 15(2): 368-371, 2019 02.
Article En | MEDLINE | ID: mdl-30153072

In its third edition, the Vancouver Autophagy Symposium presented a platform for vibrant discussion on the differential roles of macroautophagy/autophagy in disease. This one-day symposium was held at the BC Cancer Research Centre in Vancouver, BC, bringing together experts in cell biology, protein biochemistry and medicinal chemistry across several different disease models and model organisms. The Vancouver Autophagy Symposium featured 2 keynote speakers that are well known for their seminal contributions to autophagy research, Dr. David Rubinsztein (Cambridge Institute for Medical Research) and Dr. Kay F. Macleod (University of Chicago). Key discussions included the context-dependent roles and mechanisms of dysregulation of autophagy in diseases and the corresponding need to consider context-dependent autophagy modulation strategies. Additional highlights included the differential roles of bulk autophagy versus selective autophagy, novel autophagy regulators, and emerging chemical tools to study autophagy inhibition. Interdisciplinary discussions focused on addressing questions such as which stage of disease to target, which type of autophagy to target and which component to target for autophagy modulation. Abbreviations: AD: Alzheimer disease; AMFR/Gp78: autocrine motility factor receptor; CCCP: carbonyl cyanide m-chlorophenylhydrazone; CML: chronic myeloid leukemia; CVB3: coxsackievirus B3; DRPLA: dentatorubral-pallidoluysian atrophy; ER: endoplasmic reticulum; ERAD: ER-associated degradation; FA: focal adhesion; HCQ: hydroxychloroquine; HD: Huntingtin disease; HIF1A/Hif1α: hypoxia inducible factor 1 subunit alpha; HTT: huntingtin; IM: imatinib mesylate; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; NBR1: neighbour of BRCA1; OGA: O-GlcNAcase; PDAC: pancreatic ductal adenocarcinoma; PLEKHM1: pleckstrin homology and RUN domain containing M1; polyQ: poly-glutamine; ROS: reactive oxygen species; RP: retinitis pigmentosa; SNAP29: synaptosome associated protein 29; SPCA3: spinocerebellar ataxia type 3; TNBC: triple-negative breast cancer.


Autophagy , Animals , Apoptosis/drug effects , Autophagy/drug effects , Humans , Mitophagy/drug effects , Neoplasms/pathology , Proteins/toxicity
14.
Food Chem Toxicol ; 115: 451-459, 2018 May.
Article En | MEDLINE | ID: mdl-29614382

Recent studies suggest that human derived intestinal epithelial cells (IECs) cultured as polarized monolayers on Transwell® filters may respond differently when exposed to hazardous and non-hazardous proteins. This experimental platform was based on apical exposure of IEC monolayers to test proteins for 24 h followed by assessment of barrier integrity and cell viability. In this study, Caco-2 and T84 IEC polarized monolayers were evaluated for barrier integrity and cytotoxicity following exposure to hazardous and non-hazardous proteins for 24, 48 and 72 h. Hazardous proteins included Clostridium difficile toxin A (ToxA), Streptolysin O (SLO), Wheat Germ Agglutinin (WGA), and Phaseolus vulgaris haemagglutinin-E (PHA-E). Non-hazardous proteins included bovine serum albumin (BSA), porcine serum albumin (PSA), and fibronectin (Fbn). In general, evidence of diminished barrier integrity or cell viability observed following exposure to hazardous proteins for 24 h was more pronounced after 48 and 72 h for both IEC monolayers. Non-hazardous proteins exhibiting no impact following 24 h of exposure elicited minimal effects over longer exposure durations. These results support the utility of using cultured human IEC polarized monolayers to differentiate between hazardous and non-hazardous proteins and suggest that longer durations of exposure may further improve the ability to distinguish between them.


Intestinal Mucosa/drug effects , Proteins/pharmacology , Proteins/toxicity , Caco-2 Cells , Cell Membrane Permeability/drug effects , Humans
15.
Toxicol In Vitro ; 46: 155-162, 2018 Feb.
Article En | MEDLINE | ID: mdl-29017774

Many natural and industrial proteins are known to have properties that can result in type I hypersensitivity, however, to date, no validated test system exists that can predict the sensitizing potential of these allergens. Thus, the objective of this study was to develop a protocol based on the myeloid cell-based Genomic Allergen Rapid Detection (GARD) assay that can be used to assess and predict the capacity of protein allergens known to induce sensitization in the respiratory tract. Cellular responses induced by eight selected proteins were assessed using transcriptional profiling, flow cytometry and multiplex cytokine analysis. 391 potential biomarkers were identified as a predictive signature and a series of cross-validations supported the validity of the model. These results together with biological pathway analysis of the transcriptomic data indicate that the investigated cell system is able to capture relevant events linked to type I hypersensitization.


Allergens/toxicity , Proteins/toxicity , Respiratory System/drug effects , Toxicity Tests/methods , Animal Testing Alternatives , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Biomarkers , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/drug effects , Dermatitis, Allergic Contact/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Respiratory System/immunology
16.
J Mol Cell Biol ; 9(6): 516-532, 2017 12 01.
Article En | MEDLINE | ID: mdl-28992183

High temperature requirement A1 (HtrA1) belongs to an ancient protein family that is linked to various human disorders. The precise role of exon 1-encoded N-terminal domains and how these influence the biological functions of human HtrA1 remain elusive. In this study, we traced the evolutionary origins of these N-terminal domains to a single gene fusion event in the most recent common ancestor of vertebrates. We hypothesized that human HtrA1 is implicated in unfolded protein response. In highly secretory cells of the retinal pigmented epithelia, endoplasmic reticulum (ER) stress upregulated HtrA1. HtrA1 co-localized with vimentin intermediate filaments in highly arborized fashion. Upon ER stress, HtrA1 tracked along intermediate filaments, which collapsed and bundled in an aggresome at the microtubule organizing center. Gene silencing of HtrA1 altered the schedule and amplitude of adaptive signaling and concomitantly resulted in apoptosis. Restoration of wild-type HtrA1, but not its protease inactive mutant, was necessary and sufficient to protect from apoptosis. A variant of HtrA1 that harbored exon 1 substitutions displayed reduced efficacy in rescuing cells from proteotoxicity. Our results illuminate the integration of HtrA1 in the toolkit of mammalian cells against protein misfolding and the implications of defects in HtrA1 in proteostasis.


Endoplasmic Reticulum Stress , Protective Agents/metabolism , Proteins/toxicity , Cell Line , Endoplasmic Reticulum Stress/drug effects , Evolution, Molecular , Gene Knockdown Techniques , Humans , Intermediate Filaments/drug effects , Intermediate Filaments/metabolism , Microtubule-Organizing Center/metabolism , Phylogeny , Proteasome Endopeptidase Complex/metabolism , Protein Folding/drug effects , Protein Transport/drug effects , Retinal Pigment Epithelium/metabolism , Ubiquitin/metabolism , Unfolded Protein Response/drug effects , Up-Regulation/drug effects , Vimentin/metabolism
17.
Toxicol In Vitro ; 44: 85-93, 2017 Oct.
Article En | MEDLINE | ID: mdl-28652201

Relatively few proteins in nature produce adverse effects following oral exposure. Of those that do, effects are often observed in the gut, particularly on intestinal epithelial cells (IEC). Previous studies reported that addition of protein toxins to IEC lines disrupted monolayer integrity but innocuous dietary proteins did not. Studies presented here investigated the effects of innocuous (bovine serum albumin, ß-lactoglobulin, RuBisCO, fibronectin) or hazardous (phytohaemagglutinin-E, concanavalin A, wheat germ agglutinin, melittin) proteins that either were untreated or exposed to digestive enzymes prior to addition to Caco-2 human IEC line monolayers. At high concentrations intact fibronectin caused an increase in monolayer permeability but other innocuous proteins did not whether exposed to digestive enzymes or not. In contrast, all untreated hazardous proteins and those that were resistant to digestion (ex. wheat germ agglutinin) disrupted monolayer integrity. However, proteins sensitive to degradation by digestive enzymes (ex. melittin) did not adversely affect monolayers when exposed to these enzymes prior to addition to IEC line monolayers. These results indicate that in vitro exposure of proteins to digestive enzymes can assist in differentiating between innocuous and hazardous proteins as another component to consider in the overall weight of evidence approach in protein hazard assessment.


Epithelial Cells/drug effects , Pancreatin/pharmacology , Pepsin A/pharmacology , Proteins/toxicity , Caco-2 Cells , Digestion , Gastrointestinal Tract/metabolism , Humans , Intestines/cytology , Tight Junctions/drug effects
18.
BMC Biol ; 15(1): 29, 2017 04 06.
Article En | MEDLINE | ID: mdl-28385158

BACKGROUND: Animals exhibit astonishingly complex behaviors. Studying the subtle features of these behaviors requires quantitative, high-throughput, and accurate systems that can cope with the often rich perplexing data. RESULTS: Here, we present a Multi-Animal Tracker (MAT) that provides a user-friendly, end-to-end solution for imaging, tracking, and analyzing complex behaviors of multiple animals simultaneously. At the core of the tracker is a machine learning algorithm that provides immense flexibility to image various animals (e.g., worms, flies, zebrafish, etc.) under different experimental setups and conditions. Focusing on C. elegans worms, we demonstrate the vast advantages of using this MAT in studying complex behaviors. Beginning with chemotaxis, we show that approximately 100 animals can be tracked simultaneously, providing rich behavioral data. Interestingly, we reveal that worms' directional changes are biased, rather than random - a strategy that significantly enhances chemotaxis performance. Next, we show that worms can integrate environmental information and that directional changes mediate the enhanced chemotaxis towards richer environments. Finally, offering high-throughput and accurate tracking, we show that the system is highly suitable for longitudinal studies of aging- and proteotoxicity-associated locomotion deficits, enabling large-scale drug and genetic screens. CONCLUSIONS: Together, our tracker provides a powerful and simple system to study complex behaviors in a quantitative, high-throughput, and accurate manner.


Behavior, Animal , Caenorhabditis elegans/physiology , Ethology/methods , Aging/physiology , Algorithms , Animals , Chemotaxis , Machine Learning , Nerve Degeneration/pathology , Proteins/toxicity , Software , Time Factors , Video Recording
19.
Int J Biol Macromol ; 100: 89-96, 2017 Jul.
Article En | MEDLINE | ID: mdl-27327907

Curcumin, a diarylheptanoid compound, found in spice turmeric is known to alter the aggregation of proteins and reduce the toxicity of the aggregates. This review looks at the molecular basis of modulating protein aggregation and toxicity of the aggregates. Foremost, we identify the interaction of curcumin and its derivatives with proteins/peptides and the effect of their interaction on the conformational stability and unfolding/folding pathway(s). The unfolding/folding processes generate partially folded/unfolded intermediate, which serve as aggregation precursor state. Secondly, we discuss the effect of curcumin binding on the kinetics parameters of the aggregation process, which give information about the mechanism of the aggregation inhibition. We describe, in addition, that curcumin can accelerate/promote fibril formation by binding to oligomeric intermediate(s) accumulated in the aggregation pathway. Finally, we discuss the correlation of curcumin-induced monomeric and/or oligomeric precursor states with aggregate structure and toxicity. On the basis of these discussions, we propose a model describing curcumin-induced inhibition/promotion of formation of amyloid-like fibrils.


Curcumin/pharmacology , Protein Aggregates , Proteins/chemistry , Amyloid/chemistry , Animals , Humans , Peptides/chemistry , Peptides/toxicity , Proteins/toxicity
20.
Curr Pharm Biotechnol ; 18(2): 108-120, 2017.
Article En | MEDLINE | ID: mdl-27924724

Antimicrobial peptides (AMPs) and proteins are produced by a wide range of organisms as important elements of their defense mechanisms, forming a large number of antimicrobial compounds that can be used to treat several human infections. The potential for the use of AMPs and antimicrobial proteins in therapeutics is growing, but their application is often limited, due to their poor physical and/or chemical properties. In recent years, several drug delivery systems have been proposed to carry such molecules, in an attempt to overcome the difficulties regarding their properties. However, no review has yet systematized the most relevant information on this subject. Therefore, this review summarizes the work that has been conducted to develop delivery systems for the transport and protection of AMPs and antimicrobial proteins, including their description and potential applications, while highlighting the opportunities for future research in this field.


Anti-Infective Agents/administration & dosage , Drug Delivery Systems , Peptides/administration & dosage , Proteins/administration & dosage , Anti-Infective Agents/pharmacokinetics , Anti-Infective Agents/toxicity , Biological Availability , Humans , Peptides/pharmacokinetics , Peptides/toxicity , Proteins/pharmacokinetics , Proteins/toxicity
...