239 results on '"Adzhubei, A A"'
Search Results
2. DeMAG predicts the effects of variants in clinically actionable genes by integrating structural and evolutionary epistatic features
- Author
-
Luppino, Federica, Adzhubei, Ivan A., Cassa, Christopher A., and Toth-Petroczy, Agnes
- Published
- 2023
- Full Text
- View/download PDF
3. Small open reading frames: a comparative genetics approach to validation
- Author
-
Jain, Niyati, Richter, Felix, Adzhubei, Ivan, Sharp, Andrew J., and Gelb, Bruce D.
- Published
- 2023
- Full Text
- View/download PDF
4. Small open reading frames: a comparative genetics approach to validation
- Author
-
Niyati Jain, Felix Richter, Ivan Adzhubei, Andrew J. Sharp, and Bruce D. Gelb
- Subjects
Micropeptides ,Small open reading frames ,Human genetic variation ,Evolutionary conservation ,Comparative genetics ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Open reading frames (ORFs) with fewer than 100 codons are generally not annotated in genomes, although bona fide genes of that size are known. Newer biochemical studies have suggested that thousands of small protein-coding ORFs (smORFs) may exist in the human genome, but the true number and the biological significance of the micropeptides they encode remain uncertain. Here, we used a comparative genomics approach to identify high-confidence smORFs that are likely protein-coding. We identified 3,326 high-confidence smORFs using constraint within human populations and evolutionary conservation as additional lines of evidence. Next, we validated that, as a group, our high-confidence smORFs are conserved at the amino-acid level rather than merely residing in highly conserved non-coding regions. Finally, we found that high-confidence smORFs are enriched among disease-associated variants from GWAS. Overall, our results highlight that smORF-encoded peptides likely have important functional roles in human disease.
- Published
- 2023
- Full Text
- View/download PDF
5. DeMAG predicts the effects of variants in clinically actionable genes by integrating structural and evolutionary epistatic features
- Author
-
Federica Luppino, Ivan A. Adzhubei, Christopher A. Cassa, and Agnes Toth-Petroczy
- Subjects
Science - Abstract
Abstract Despite the increasing use of genomic sequencing in clinical practice, the interpretation of rare genetic variants remains challenging even in well-studied disease genes, resulting in many patients with Variants of Uncertain Significance (VUSs). Computational Variant Effect Predictors (VEPs) provide valuable evidence in variant assessment, but they are prone to misclassifying benign variants, contributing to false positives. Here, we develop Deciphering Mutations in Actionable Genes (DeMAG), a supervised classifier for missense variants trained using extensive diagnostic data available in 59 actionable disease genes (American College of Medical Genetics and Genomics Secondary Findings v2.0, ACMG SF v2.0). DeMAG improves performance over existing VEPs by reaching balanced specificity (82%) and sensitivity (94%) on clinical data, and includes a novel epistatic feature, the ‘partners score’, which leverages evolutionary and structural partnerships of residues. The ‘partners score’ provides a general framework for modeling epistatic interactions, integrating both clinical and functional information. We provide our tool and predictions for all missense variants in 316 clinically actionable disease genes (demag.org) to facilitate the interpretation of variants and improve clinical decision-making.
- Published
- 2023
- Full Text
- View/download PDF
6. Inhibition of HIV Replication by Apolipoprotein A-I Binding Protein Targeting the Lipid Rafts.
- Author
-
Pushkarsky, Tatiana, Brichacek, Beda, Vanpouille, Christophe, Adzhubei, Alexei, Mukhamedova, Nigora, Sviridov, Dmitri, Margolis, Leonid, Jones, Richard, Miller, Yury, Bukrinsky, Michael, Dubrovsky, Larisa, Ward, Adam, and Choi, Soo-Ho
- Subjects
AIBP ,HIV ,HLA ,Nef ,exosomes ,extracellular vesicles ,fusion ,lipid rafts - Abstract
Apolipoprotein A-I binding protein (AIBP) is a protein involved in regulation of lipid rafts and cholesterol efflux. AIBP has been suggested to function as a protective factor under several sets of pathological conditions associated with increased abundance of lipid rafts, such as atherosclerosis and acute lung injury. Here, we show that exogenously added AIBP reduced the abundance of lipid rafts and inhibited HIV replication in vitro as well as in HIV-infected humanized mice, whereas knockdown of endogenous AIBP increased HIV replication. Endogenous AIBP was much more abundant in activated T cells than in monocyte-derived macrophages (MDMs), and exogenous AIBP was much less effective in T cells than in MDMs. AIBP inhibited virus-cell fusion, specifically targeting cells with lipid rafts mobilized by cell activation or Nef-containing exosomes. MDM-HIV fusion was sensitive to AIBP only in the presence of Nef provided by the virus or exosomes. Peripheral blood mononuclear cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, bound less AIBP than cells from donors with other HLA genotypes and were not protected by AIBP from rapid HIV-1 replication. These results provide the first evidence for the role of Nef exosomes in regulating HIV-cell fusion by modifying lipid rafts and suggest that AIBP is an innate factor that restricts HIV replication by targeting lipid rafts.IMPORTANCE Apolipoprotein A-I binding protein (AIBP) is a recently identified innate anti-inflammatory factor. Here, we show that AIBP inhibited HIV replication by targeting lipid rafts and reducing virus-cell fusion. Importantly, AIBP selectively reduced levels of rafts on cells stimulated by an inflammatory stimulus or treated with extracellular vesicles containing HIV-1 protein Nef without affecting rafts on nonactivated cells. Accordingly, fusion of monocyte-derived macrophages with HIV was sensitive to AIBP only in the presence of Nef. Silencing of endogenous AIBP significantly upregulated HIV-1 replication. Interestingly, HIV-1 replication in cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, was not inhibited by AIBP. These results suggest that AIBP is an innate anti-HIV factor that targets virus-cell fusion.
- Published
- 2020
7. Carriers of Heterozygous Loss-of-Function ACE Mutations Are at Risk for Alzheimer’s Disease
- Author
-
Sergei M. Danilov, Ivan A. Adzhubei, Alexander J. Kozuch, Pavel A. Petukhov, Isolda A. Popova, Ananyo Choudhury, Dhriti Sengupta, and Steven M. Dudek
- Subjects
angiotensin I-converting enzyme ,mutations ,conformational changes ,plasma ACE ,screening ,Biology (General) ,QH301-705.5 - Abstract
We hypothesized that subjects with heterozygous loss-of-function (LoF) ACE mutations are at risk for Alzheimer’s disease because amyloid Aβ42, a primary component of the protein aggregates that accumulate in the brains of AD patients, is cleaved by ACE (angiotensin I-converting enzyme). Thus, decreased ACE activity in the brain, either due to genetic mutation or the effects of ACE inhibitors, could be a risk factor for AD. To explore this hypothesis in the current study, existing SNP databases were analyzed for LoF ACE mutations using four predicting tools, including PolyPhen-2, and compared with the topology of known ACE mutations already associated with AD. The combined frequency of >400 of these LoF-damaging ACE mutations in the general population is quite significant—up to 5%—comparable to the frequency of AD in the population > 70 y.o., which indicates that the contribution of low ACE in the development of AD could be under appreciated. Our analysis suggests several mechanisms by which ACE mutations may be associated with Alzheimer’s disease. Systematic analysis of blood ACE levels in patients with all ACE mutations is likely to have clinical significance because available sequencing data will help detect persons with increased risk of late-onset Alzheimer’s disease. Patients with transport-deficient ACE mutations (about 20% of damaging ACE mutations) may benefit from preventive or therapeutic treatment with a combination of chemical and pharmacological (e.g., centrally acting ACE inhibitors) chaperones and proteosome inhibitors to restore impaired surface ACE expression, as was shown previously by our group for another transport-deficient ACE mutation-Q1069R.
- Published
- 2024
- Full Text
- View/download PDF
8. Direct interaction between ABCA1 and HIV-1 Nef: Molecular modeling and virtual screening for inhibitors
- Author
-
Adzhubei, Alexei A., Kulkarni, Amol, Tolstova, Anna P., Anashkina, Anastasia A., Sviridov, Dmitri, Makarov, Alexander A., and Bukrinsky, Michael I.
- Published
- 2021
- Full Text
- View/download PDF
9. Direct interaction between ABCA1 and HIV-1 Nef: Molecular modeling and virtual screening for inhibitors
- Author
-
Alexei A. Adzhubei, Amol Kulkarni, Anna P. Tolstova, Anastasia A. Anashkina, Dmitri Sviridov, Alexander A. Makarov, and Michael I. Bukrinsky
- Subjects
HIV-1 ,Nef ,ABCA1 ,Molecular modeling ,Molecular dynamics ,Virtual screening ,Biotechnology ,TP248.13-248.65 - Abstract
HIV-1 infection impairs cellular cholesterol efflux by downmodulating the cholesterol transporter ABCA1, leading to metabolic co-morbidities like cardio-vascular disease. The main mechanism of this effect is impairment by the HIV-1 protein Nef of the ABCA1 interaction with the endoplasmic reticulum chaperone calnexin, which leads to a block in ABCA1 maturation followed by its degradation. However, ABCA1 is also downmodulated by Nef delivered with the extracellular vesicles, suggesting involvement of a direct Nef:ABCA1 interaction at the plasma membrane. Here, we present an optimized model of the Nef:ABCA1 interaction, which identifies interaction sites and provides an opportunity to perform a virtual screening for potential inhibitors. Interestingly, the predicted sites on Nef involved in the ABCA1 interaction overlap with those involved in the interaction with calnexin. The compounds previously shown to block Nef:calnexin interaction were among the top ranking ligands in docking simulations with ABCA1-interacting sites on Nef, suggesting the possibility that both interactions can be inhibited by the same chemical compounds. This study identifies a series of compounds for potential development as inhibitors of Nef-mediated co-morbidities of HIV infection.
- Published
- 2021
- Full Text
- View/download PDF
10. Carriers of Heterozygous Loss-of-Function ACE Mutations Are at Risk for Alzheimer’s Disease
- Author
-
Danilov, Sergei M., primary, Adzhubei, Ivan A., additional, Kozuch, Alexander J., additional, Petukhov, Pavel A., additional, Popova, Isolda A., additional, Choudhury, Ananyo, additional, Sengupta, Dhriti, additional, and Dudek, Steven M., additional
- Published
- 2024
- Full Text
- View/download PDF
11. No evidence that natural selection has been less effective at removing deleterious mutations in Europeans than in West Africans
- Author
-
Do, Ron, Balick, Daniel, Li, Heng, Adzhubei, Ivan, Sunyaev, Shamil, and Reich, David
- Subjects
Quantitative Biology - Populations and Evolution - Abstract
Non-African populations have experienced major bottlenecks in the time since their split from West Africans, which has led to the hypothesis that natural selection to remove weakly deleterious mutations may have been less effective in non-Africans. To directly test this hypothesis, we measure the per-genome accumulation of deleterious mutations across diverse humans. We fail to detect any significant differences, but find that archaic Denisovans accumulated non-synonymous mutations at a higher rate than modern humans, consistent with the longer separation time of modern and archaic humans. We also revisit the empirical patterns that have been interpreted as evidence for less effective removal of deleterious mutations in non-Africans than in West Africans, and show they are not driven by differences in selection after population separation, but by neutral evolution., Comment: 53 pages (22 page manuscript and 31 supplemental information)
- Published
- 2014
12. Switching On/Off Amyloid Plaque Formation in Transgenic Animal Models of Alzheimer’s Disease
- Author
-
Kozin, Sergey A., primary, Kechko, Olga I., additional, Adzhubei, Alexei A., additional, Makarov, Alexander A., additional, and Mitkevich, Vladimir A., additional
- Published
- 2023
- Full Text
- View/download PDF
13. Na,K-ATPase Acts as a Beta-Amyloid Receptor Triggering Src Kinase Activation
- Author
-
Irina Yu. Petrushanko, Artem M. Tverskoi, Evgeny P. Barykin, Aleksandra V. Petrovskaya, Maria A. Strelkova, Olga G. Leonova, Anastasia A. Anashkina, Anna P. Tolstova, Alexei A. Adzhubei, Anna Yu. Bogdanova, Alexander A. Makarov, and Vladimir A. Mitkevich
- Subjects
Na,K-ATPase ,beta-amyloid ,Src kinase ,hypoxia ,receptor function ,Cytology ,QH573-671 - Abstract
Beta-amyloid (Aβ) has a dual role, both as an important factor in the pathology of Alzheimer’s disease and as a regulator in brain physiology. The inhibitory effect of Aβ42 oligomers on Na,K-ATPase contributes to neuronal dysfunction in Alzheimer’s disease. Still, the physiological role of the monomeric form of Aβ42 interaction with Na,K-ATPase remains unclear. We report that Na,K-ATPase serves as a receptor for Aβ42 monomer, triggering Src kinase activation. The co-localization of Aβ42 with α1- and β1-subunits of Na,K-ATPase, and Na,K-ATPase with Src kinase in SH-SY5Y neuroblastoma cells, was observed. Treatment of cells with 100 nM Aβ42 causes Src kinase activation, but does not alter Na,K-ATPase transport activity. The interaction of Aβ42 with α1β1 Na,K-ATPase isozyme leads to activation of Src kinase associated with the enzyme. Notably, prevention of Na,K-ATPase:Src kinase interaction by a specific inhibitor pNaKtide disrupts the Aβ-induced Src kinase activation. Stimulatory effect of Aβ42 on Src kinase was lost under hypoxic conditions, which was similar to the effect of specific Na,K-ATPase ligands, the cardiotonic steroids. Our findings identify Na,K-ATPase as a Aβ42 receptor, thus opening a prospect on exploring the physiological and pathological Src kinase activation caused by Aβ42 in the nervous system.
- Published
- 2022
- Full Text
- View/download PDF
14. Interaction Interface of Aβ42 with Human Na,K-ATPase Studied by MD and ITC and Inhibitor Screening by MD
- Author
-
Alexei A. Adzhubei, Anna P. Tolstova, Maria A. Strelkova, Vladimir A. Mitkevich, Irina Yu. Petrushanko, and Alexander A. Makarov
- Subjects
Alzheimer’s disease ,Na,K-ATPase ,beta amyloid ,interaction interface ,interaction inhibitors screening ,conformations of Na,K-ATPase ,Biology (General) ,QH301-705.5 - Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease accompanied by progressive cognitive and memory dysfunction due to disruption of normal electrotonic properties of neurons and neuronal loss. The Na,K-ATPase interaction with beta amyloid (Aβ) plays an important role in AD pathogenesis. It has been shown that Na,K-ATPase activity in the AD brain was significantly lower than those in age-matched control brain. The interaction of Aβ42 with Na,K-ATPase and subsequent oligomerization leads to inhibition of the enzyme activity. In this study interaction interfaces between three common Aβ42 isoforms, and different conformations of human Na,K-ATPase (α1β1) have been obtained using molecular modeling, including docking and molecular dynamics (MD). Interaction sites of Na,K-ATPase with Aβ42 are localized between extracellular parts of α- and β- subunits and are practically identical for Na,K-ATPase at different conformations. Thermodynamic parameters for the formation of Na,K-ATPase:Aβ42 complex at different conformations acquired by isothermal titration calorimetry (ITC) are similar, which is in line with the data of molecular modeling. Similarity of Na,K-ATPase interaction interfaces with Aβ in all conformations allowed us to cross-screen potential inhibitors for this interaction and find pharmaceutical compounds that could block it.
- Published
- 2022
- Full Text
- View/download PDF
15. Informing variant assessment using structured evidence from prior classifications (PS1, PM5, and PVS1 sequence variant interpretation criteria)
- Author
-
Vineel Bhat, Ivan A. Adzhubei, James D. Fife, Matthew Lebo, and Christopher A. Cassa
- Subjects
Article ,Genetics (clinical) - Abstract
PurposeTo explore whether evidence of pathogenicity from prior variant classifications in ClinVar could be used to inform variant interpretation using the ACMG/AMP clinical guidelines.MethodsWe identify distinct SNVs which are either similar in location or in functional consequence to pathogenic variants in ClinVar, and analyze evidence in support of pathogenicity using three interpretation criteria.ResultsThousands of variants, including many in clinically actionable disease genes (ACMG SFv3.0), have evidence of pathogenicity from existing variant classifications, accounting for 2.5% of non-synonymous SNVs within ClinVar. Notably, there are many variants with uncertain or conflicting classifications which cause the same amino acid substitution as other pathogenic variants (PS1, N=323), variants which are predicted to cause different amino acid substitutions in the same codon as pathogenic variants (PM5, N=7,692), and LOF variants which are present in genes where many LOF variants are classified as pathogenic (PVS1, N=3,635). The majority of these variants have similar computational predictions of pathogenicity and splicing impact as their associated pathogenic variants.ConclusionBroadly, over 1.4 million SNVs exome-wide could make use of information from previously classified pathogenic variants. We have developed a pipeline to identify variants meeting these criteria, which may inform interpretation efforts.
- Published
- 2023
- Full Text
- View/download PDF
16. Erratum for Dubrovsky et al., 'Inhibition of HIV Replication by Apolipoprotein A-I Binding Protein Targeting the Lipid Rafts'
- Author
-
Larisa Dubrovsky, Adam Ward, Soo-Ho Choi, Tatiana Pushkarsky, Beda Brichacek, Christophe Vanpouille, Alexei A. Adzhubei, Nigora Mukhamedova, Dmitri Sviridov, Leonid Margolis, Richard B. Jones, Yury I. Miller, and Michael Bukrinsky
- Subjects
Microbiology ,QR1-502 - Published
- 2020
- Full Text
- View/download PDF
17. Inhibition of HIV Replication by Apolipoprotein A-I Binding Protein Targeting the Lipid Rafts
- Author
-
Larisa Dubrovsky, Adam Ward, Soo-Ho Choi, Tatiana Pushkarsky, Beda Brichacek, Christophe Vanpouille, Alexei A. Adzhubei, Nigora Mukhamedova, Dmitri Sviridov, Leonid Margolis, Richard B. Jones, Yury I. Miller, and Michael Bukrinsky
- Subjects
HIV ,AIBP ,Nef ,extracellular vesicles ,exosomes ,lipid rafts ,Microbiology ,QR1-502 - Abstract
ABSTRACT Apolipoprotein A-I binding protein (AIBP) is a protein involved in regulation of lipid rafts and cholesterol efflux. AIBP has been suggested to function as a protective factor under several sets of pathological conditions associated with increased abundance of lipid rafts, such as atherosclerosis and acute lung injury. Here, we show that exogenously added AIBP reduced the abundance of lipid rafts and inhibited HIV replication in vitro as well as in HIV-infected humanized mice, whereas knockdown of endogenous AIBP increased HIV replication. Endogenous AIBP was much more abundant in activated T cells than in monocyte-derived macrophages (MDMs), and exogenous AIBP was much less effective in T cells than in MDMs. AIBP inhibited virus-cell fusion, specifically targeting cells with lipid rafts mobilized by cell activation or Nef-containing exosomes. MDM-HIV fusion was sensitive to AIBP only in the presence of Nef provided by the virus or exosomes. Peripheral blood mononuclear cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, bound less AIBP than cells from donors with other HLA genotypes and were not protected by AIBP from rapid HIV-1 replication. These results provide the first evidence for the role of Nef exosomes in regulating HIV-cell fusion by modifying lipid rafts and suggest that AIBP is an innate factor that restricts HIV replication by targeting lipid rafts. IMPORTANCE Apolipoprotein A-I binding protein (AIBP) is a recently identified innate anti-inflammatory factor. Here, we show that AIBP inhibited HIV replication by targeting lipid rafts and reducing virus-cell fusion. Importantly, AIBP selectively reduced levels of rafts on cells stimulated by an inflammatory stimulus or treated with extracellular vesicles containing HIV-1 protein Nef without affecting rafts on nonactivated cells. Accordingly, fusion of monocyte-derived macrophages with HIV was sensitive to AIBP only in the presence of Nef. Silencing of endogenous AIBP significantly upregulated HIV-1 replication. Interestingly, HIV-1 replication in cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, was not inhibited by AIBP. These results suggest that AIBP is an innate anti-HIV factor that targets virus-cell fusion.
- Published
- 2020
- Full Text
- View/download PDF
18. Switching On/Off Amyloid Plaque Formation in Transgenic Animal Models of Alzheimer's Disease.
- Author
-
Kozin, Sergey A., Kechko, Olga I., Adzhubei, Alexei A., Makarov, Alexander A., and Mitkevich, Vladimir A.
- Subjects
ALZHEIMER'S disease ,TRANSGENIC animals ,AMYLOID plaque ,NICOTINIC receptors ,NICOTINIC acetylcholine receptors ,ANIMAL models in research ,CHOLINERGIC receptors ,AMYLOID beta-protein - Abstract
A hallmark of Alzheimer's disease (AD) are the proteinaceous aggregates formed by the amyloid-beta peptide (Aβ) that is deposited inside the brain as amyloid plaques. The accumulation of aggregated Aβ may initiate or enhance pathologic processes in AD. According to the amyloid hypothesis, any agent that has the capability to inhibit Aβ aggregation and/or destroy amyloid plaques represents a potential disease-modifying drug. In 2023, a humanized IgG1 monoclonal antibody (lecanemab) against the Aβ-soluble protofibrils was approved by the US FDA for AD therapy, thus providing compelling support to the amyloid hypothesis. To acquire a deeper insight on the in vivo Aβ aggregation, various animal models, including aged herbivores and carnivores, non-human primates, transgenic rodents, fish and worms were widely exploited. This review is based on the recent data obtained using transgenic animal AD models and presents experimental verification of the critical role in Aβ aggregation seeding of the interactions between zinc ions, Aβ with the isomerized Asp7 (isoD7-Aβ) and the α4β2 nicotinic acetylcholine receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Comorbidities of HIV infection: role of Nef-induced impairment of cholesterol metabolism and lipid raft functionality
- Author
-
Sviridov, Dmitri, Mukhamedova, Nigora, Makarov, Alexander A., Adzhubei, Alexei, and Bukrinsky, Michael
- Published
- 2020
- Full Text
- View/download PDF
20. Molecular Mechanism of Zinc-Dependent Oligomerization of Alzheimer’s Amyloid-β with Taiwan (D7H) Mutation
- Author
-
Kozin, Olga I. Kechko, Alexei A. Adzhubei, Anna P. Tolstova, Maria I. Indeykina, Igor A. Popov, Sergey S. Zhokhov, Nikolay V. Gnuchev, Vladimir A. Mitkevich, Alexander A. Makarov, and Sergey A.
- Subjects
Alzheimer’s disease ,amyloid-beta ,familial Taiwan mutation D7H ,zinc ,metal binding domain ,oligomerization ,aggregation seeding ,drug target ,amyloid plaque formation - Abstract
Amyloid-β (Aβ) is a peptide formed by 39–43 amino acids, heterogenous by the length of its C-terminus. Aβ constitutes a subnanomolar monomeric component of human biological fluids; however, in sporadic variants of Alzheimer’s disease (AD), it forms soluble neurotoxic oligomers and accumulates as insoluble extracellular polymeric aggregates (amyloid plaques) in the brain tissues. The plaque formation is controlled by zinc ions; therefore, abnormal interactions between the ions and Aβ seem to take part in the triggering of sporadic AD. The amyloid plaques contain various Aβ isoforms, among which the most common is Aβ with an isoaspartate in position 7 (isoD7). The spontaneous conversion of D7 to isoD7 is associated with Aβ aging. Aβ molecules with isoD7 (isoD7-Aβ) easily undergo zinc-dependent oligomerization, and upon administration to transgenic animals (mice, nematodes) used for AD modeling, act as zinc-dependent seeds of the pathological aggregation of Aβ. The formation of zinc-bound homo- and hetero-oligomers with the participation of isoD7-Aβ is based on the rigidly structured segment 11-EVHH-14, located in the Aβ metal binding domain (Aβ16). Some hereditary variants of AD are associated with familial mutations within the domain. Among these, the most susceptible to zinc-dependent oligomerization is Aβ with Taiwan (D7H) mutation (D7H-Aβ). In this study, the D7H-Aβ metal binding domain (D7H-Aβ16) has been used as a model to establish the molecular mechanism of zinc-induced D7H-Aβ oligomerization through turbidimetry, dynamic light scattering, isothermal titration calorimetry, mass spectrometry, and computer modelling. Additionally, the modeling data showed that a molecule of D7H-Aβ, as well as isoD7-Aβ in combination with two Aβ molecules, renders a stable zinc-induced heterotrimer. The trimers are held together by intermolecular interfaces via zinc ions, with the primary interfaces formed by 11-EVHH-14 sites of the interacting trimer subunits. In summary, the obtained results confirm the role of the 11-EVHH-14 region as a structure and function determinant for the zinc-dependent oligomerization of all known Aβ species (including various chemically modified isoforms and AD-associated mutants) and point at this region as a potent target for drugs aimed to stop amyloid plaque formation in both sporadic and hereditary variants of AD.
- Published
- 2023
- Full Text
- View/download PDF
21. Co-morbidities of HIV infection: role of Nef-induced impairment of cholesterol metabolism and lipid raft functionality
- Author
-
Sviridov, Dmitri, Mukhamedova, Nigora, Makarov, Alexander A., Adzhubei, Alexei, and Bukrinsky, Michael
- Published
- 2019
- Full Text
- View/download PDF
22. A literature review at genome scale: improving clinical variant assessment
- Author
-
Cassa, Christopher A, Jordan, Daniel M, Adzhubei, Ivan, and Sunyaev, Shamil
- Published
- 2018
- Full Text
- View/download PDF
23. Pharmacokinetics and Molecular Modeling Indicate nAChRα4-Derived Peptide HAEE Goes through the Blood–Brain Barrier
- Author
-
Yurii A. Zolotarev, Vladimir A. Mitkevich, Stanislav I. Shram, Alexei A. Adzhubei, Anna P. Tolstova, Oleg B. Talibov, Alexander K. Dadayan, Nikolai F. Myasoyedov, Alexander A. Makarov, and Sergey A. Kozin
- Subjects
Alzheimer’s disease ,beta-amyloid ,α4β2 nicotinic acetylcholine receptor ,peptide drug ,blood–brain barrier ,receptor-mediated transcytosis ,Microbiology ,QR1-502 - Abstract
One of the treatment strategies for Alzheimer’s disease (AD) is based on the use of pharmacological agents capable of binding to beta-amyloid (Aβ) and blocking its aggregation in the brain. Previously, we found that intravenous administration of the synthetic tetrapeptide Acetyl-His-Ala-Glu-Glu-Amide (HAEE), which is an analogue of the 35–38 region of the α4 subunit of α4β2 nicotinic acetylcholine receptor and specifically binds to the 11–14 site of Aβ, reduced the development of cerebral amyloidogenesis in a mouse model of AD. In the current study on three types of laboratory animals, we determined the biodistribution and tissue localization patterns of HAEE peptide after single intravenous bolus administration. The pharmacokinetic parameters of HAEE were established using uniformly tritium-labeled HAEE. Pharmacokinetic data provided evidence that HAEE goes through the blood–brain barrier. Based on molecular modeling, a role of LRP1 in receptor-mediated transcytosis of HAEE was proposed. Altogether, the results obtained indicate that the anti-amyloid effect of HAEE, previously found in a mouse model of AD, most likely occurs due to its interaction with Aβ species directly in the brain.
- Published
- 2021
- Full Text
- View/download PDF
24. Informing variant assessment using structured evidence from prior classifications (PS1, PM5, and PVS1 sequence variant interpretation criteria)
- Author
-
Bhat, Vineel, primary, Adzhubei, Ivan A., additional, Fife, James D., additional, Lebo, Matthew, additional, and Cassa, Christopher A., additional
- Published
- 2023
- Full Text
- View/download PDF
25. Carriers of heterozygous loss-of-function ACE mutations are at risk for Alzheimer’s disease
- Author
-
Sergei M. Danilov, Ivan A. Adzhubei, Alex J. Kozuch, Pavel A. Petukhov, Isolda A. Popova, Ananyo Choudhury, Dhriti Sengupta, and Steven M. Dudek
- Abstract
Amyloid Aβ42 (constituents of the protein aggregates in the brains of patients with Alzheimer’s disease (AD) cleaved by ACE, and thus, a decrease in tissue ACE activity (constitutive or ACE inhibitor-induced) could be risk factor for AD. We hypothesized that subjects with heterozygous Loss-of-Function (LoF) ACE mutations are at risk for Alzheimer’s disease. Existing SNP databases were analyzed for LoF ACE mutations using PolyPhen-2 scores and compared with the topology of known ACE mutations already associated with AD. The combined frequency of >400 of these LoF-damaging ACE mutations in the general population is quite significant – up to 5 % – comparable with the frequency of AD in the population >70 years old. Our analysis suggests several mechanisms by which ACE mutations may be associated with Alzheimer’s disease. Systematic analysis of blood ACE levels in patients with all ACE mutations is likely to have clinical significance because available sequencing data will help detect persons with increased risk of late-onset Alzheimer’s disease. Patients with transport-deficient ACE mutations (about 20 % of damaging ACE mutations) may benefit from preventive or therapeutic treatment with a combination of chemical and pharmacological (e.g., centrally acting ACE inhibitors) chaperones and proteosome inhibitors to restore impaired surface ACE expression.
- Published
- 2023
- Full Text
- View/download PDF
26. Intravenously Injected Amyloid-β Peptide With Isomerized Asp7 and Phosphorylated Ser8 Residues Inhibits Cerebral β-Amyloidosis in AβPP/PS1 Transgenic Mice Model of Alzheimer’s Disease
- Author
-
Sergey A. Kozin, Evgeny P. Barykin, Georgy B. Telegin, Alexander S. Chernov, Alexei A. Adzhubei, Sergey P. Radko, Vladimir A. Mitkevich, and Alexander A. Makarov
- Subjects
Alzheimer’s disease ,amyloid-β peptide ,zinc ,isoaspartate ,serine phosphorylation ,transgenic mice ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Cerebral β-amyloidosis, an accumulation in the patient’s brain of aggregated amyloid-β (Aβ) peptides abnormally saturated by divalent biometal ions, is one of the hallmarks of Alzheimer’s disease (AD). Earlier, we found that exogenously administrated synthetic Aβ with isomerized Asp7 (isoD7-Aβ) induces Aβ fibrillar aggregation in the transgenic mice model of AD. IsoD7-Aβ molecules have been implied to act as seeds enforcing endogenous Aβ to undergo pathological aggregation through zinc-mediated interactions. On the basis of our findings on zinc-induced oligomerization of the metal-binding domain of various Aβ species, we hypothesize that upon phosphorylation of Ser8, isoD7-Aβ loses its ability to form zinc-bound oligomeric seeds. In this work, we found that (i) in vitro isoD7-Aβ with phosphorylated Ser8 (isoD7-pS8-Aβ) is less prone to spontaneous and zinc-induced aggregation in comparison with isoD7-Aβ and intact Aβ as shown by thioflavin T fluorimetry and dynamic light scattering data, and (ii) intravenous injections of isoD7-pS8-Aβ significantly slow down the progression of institutional β-amyloidosis in AβPP/PS1 transgenic mice as shown by the reduction of the congophilic amyloid plaques’ number in the hippocampus. The results support the role of the zinc-mediated oligomerization of Aβ species in the modulation of cerebral β-amyloidosis and demonstrate that isoD7-pS8-Aβ can serve as a potential molecular tool to block the aggregation of endogenous Aβ in AD.
- Published
- 2018
- Full Text
- View/download PDF
27. Docking and Molecular Dynamics-Based Identification of Interaction between Various Beta-Amyloid Isoforms and RAGE Receptor
- Author
-
Tolstova, Anna P., primary, Adzhubei, Alexei A., additional, Mitkevich, Vladimir A., additional, Petrushanko, Irina Yu., additional, and Makarov, Alexander A., additional
- Published
- 2022
- Full Text
- View/download PDF
28. Modelling interaction between HIV-1 Nef and calnexin
- Author
-
Adzhubei, Alexei A., Anashkina, Anastasia A., Tkachev, Yaroslav V., Kravatsky, Yury V., Pushkarsky, Tatiana, Kulkarni, Amol, Makarov, Alexander A., and Bukrinsky, Michael I.
- Published
- 2018
- Full Text
- View/download PDF
29. Molecular Mechanism of Zinc-Dependent Oligomerization of Alzheimer's Amyloid-β with Taiwan (D7H) Mutation.
- Author
-
Kechko, Olga I., Adzhubei, Alexei A., Tolstova, Anna P., Indeykina, Maria I., Popov, Igor A., Zhokhov, Sergey S., Gnuchev, Nikolay V., Mitkevich, Vladimir A., Makarov, Alexander A., and Kozin, Sergey A.
- Subjects
- *
ALZHEIMER'S disease , *AMYLOID beta-protein , *OLIGOMERIZATION , *ISOTHERMAL titration calorimetry , *AMYLOID plaque , *ZINC ions - Abstract
Amyloid-β (Aβ) is a peptide formed by 39–43 amino acids, heterogenous by the length of its C-terminus. Aβ constitutes a subnanomolar monomeric component of human biological fluids; however, in sporadic variants of Alzheimer's disease (AD), it forms soluble neurotoxic oligomers and accumulates as insoluble extracellular polymeric aggregates (amyloid plaques) in the brain tissues. The plaque formation is controlled by zinc ions; therefore, abnormal interactions between the ions and Aβ seem to take part in the triggering of sporadic AD. The amyloid plaques contain various Aβ isoforms, among which the most common is Aβ with an isoaspartate in position 7 (isoD7). The spontaneous conversion of D7 to isoD7 is associated with Aβ aging. Aβ molecules with isoD7 (isoD7-Aβ) easily undergo zinc-dependent oligomerization, and upon administration to transgenic animals (mice, nematodes) used for AD modeling, act as zinc-dependent seeds of the pathological aggregation of Aβ. The formation of zinc-bound homo- and hetero-oligomers with the participation of isoD7-Aβ is based on the rigidly structured segment 11-EVHH-14, located in the Aβ metal binding domain (Aβ16). Some hereditary variants of AD are associated with familial mutations within the domain. Among these, the most susceptible to zinc-dependent oligomerization is Aβ with Taiwan (D7H) mutation (D7H-Aβ). In this study, the D7H-Aβ metal binding domain (D7H-Aβ16) has been used as a model to establish the molecular mechanism of zinc-induced D7H-Aβ oligomerization through turbidimetry, dynamic light scattering, isothermal titration calorimetry, mass spectrometry, and computer modelling. Additionally, the modeling data showed that a molecule of D7H-Aβ, as well as isoD7-Aβ in combination with two Aβ molecules, renders a stable zinc-induced heterotrimer. The trimers are held together by intermolecular interfaces via zinc ions, with the primary interfaces formed by 11-EVHH-14 sites of the interacting trimer subunits. In summary, the obtained results confirm the role of the 11-EVHH-14 region as a structure and function determinant for the zinc-dependent oligomerization of all known Aβ species (including various chemically modified isoforms and AD-associated mutants) and point at this region as a potent target for drugs aimed to stop amyloid plaque formation in both sporadic and hereditary variants of AD. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. Isomerization of Asp7 in Beta-Amyloid Enhances Inhibition of the α7 Nicotinic Receptor and Promotes Neurotoxicity
- Author
-
Evgeny P. Barykin, Alexandra I. Garifulina, Elena V. Kruykova, Ekaterina N. Spirova, Anastasia A. Anashkina, Alexei A. Adzhubei, Irina V. Shelukhina, Igor E. Kasheverov, Vladimir A. Mitkevich, Sergey A. Kozin, Michael Hollmann, Victor I. Tsetlin, and Alexander A. Makarov
- Subjects
amyloid-beta ,nicotinic acetylcholine receptor ,modifications ,Alzheimer’s disease ,neurotoxicity ,calcium imaging ,radioligand analysis ,aspartate isomerization ,Cytology ,QH573-671 - Abstract
Cholinergic dysfunction in Alzheimer’s disease (AD) can be mediated by the neuronal α7 nicotinic acetylcholine receptor (α7nAChR). Beta-amyloid peptide (Aβ) binds to the α7nAChR, disrupting the receptor’s function and causing neurotoxicity. In vivo not only Aβ but also its modified forms can drive AD pathogenesis. One of these forms, iso-Aβ (containing an isomerized Asp7 residue), shows an increased neurotoxicity in vitro and stimulates amyloidogenesis in vivo. We suggested that such effects of iso-Aβ are α7nAChR-dependent. Here, using calcium imaging and electrophysiology, we found that iso-Aβ is a more potent inhibitor of the α7nAChR-mediated calcium current than unmodified Aβ. However, Asp7 isomerization eliminated the ability of Aβ to decrease the α7nAChR levels. These data indicate differences in the interaction of the peptides with the α7nAChR, which we demonstrated using computer modeling. Neither Aβ nor iso-Aβ competed with 125I-α-bungarotoxin for binding to the orthosteric site of the receptor, suggesting the allosteric binging mode of the peptides. Further we found that increased neurotoxicity of iso-Aβ was mediated by the α7nAChR. Thus, the isomerization of Asp7 enhances the inhibitory effect of Aβ on the functional activity of the α7nAChR, which may be an important factor in the disruption of the cholinergic system in AD.
- Published
- 2019
- Full Text
- View/download PDF
31. Na,K-ATPase Acts as a Beta-Amyloid Receptor Triggering Src Kinase Activation
- Author
-
Petrushanko, Irina Yu., primary, Tverskoi, Artem M., additional, Barykin, Evgeny P., additional, Petrovskaya, Aleksandra V., additional, Strelkova, Maria A., additional, Leonova, Olga G., additional, Anashkina, Anastasia A., additional, Tolstova, Anna P., additional, Adzhubei, Alexei A., additional, Bogdanova, Anna Yu., additional, Makarov, Alexander A., additional, and Mitkevich, Vladimir A., additional
- Published
- 2022
- Full Text
- View/download PDF
32. Interaction Interface of Aβ42 with Human Na,K-ATPase Studied by MD and ITC and Inhibitor Screening by MD
- Author
-
Adzhubei, Alexei, primary, Tolstova, Anna, additional, Strelkova, Maria, additional, Mitkevich, Vladimir, additional, Petrushanko, Irina, additional, and Makarov, Alexander, additional
- Published
- 2022
- Full Text
- View/download PDF
33. Direct interaction between ABCA1 and HIV-1 Nef: Molecular modeling and virtual screening for inhibitors
- Author
-
Amol A. Kulkarni, Alexander A. Makarov, Alexei A. Adzhubei, Michael Bukrinsky, Anastasia A. Anashkina, Dmitri Sviridov, and Anna P. Tolstova
- Subjects
Virtual screening ,viruses ,Biophysics ,ABCA1 ,Molecular modeling ,Molecular dynamics ,Biochemistry ,Docking (dog) ,Structural Biology ,Calnexin ,polycyclic compounds ,Genetics ,cardiovascular diseases ,ComputingMethodologies_COMPUTERGRAPHICS ,Nef ,biology ,Chemistry ,Endoplasmic reticulum ,virus diseases ,Transporter ,Computer Science Applications ,Cell biology ,Chaperone (protein) ,HIV-1 ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Efflux ,TP248.13-248.65 ,Research Article ,Biotechnology - Abstract
Graphical abstract, HIV-1 infection impairs cellular cholesterol efflux by downmodulating the cholesterol transporter ABCA1, leading to metabolic co-morbidities like cardio-vascular disease. The main mechanism of this effect is impairment by the HIV-1 protein Nef of the ABCA1 interaction with the endoplasmic reticulum chaperone calnexin, which leads to a block in ABCA1 maturation followed by its degradation. However, ABCA1 is also downmodulated by Nef delivered with the extracellular vesicles, suggesting involvement of a direct Nef:ABCA1 interaction at the plasma membrane. Here, we present an optimized model of the Nef:ABCA1 interaction, which identifies interaction sites and provides an opportunity to perform a virtual screening for potential inhibitors. Interestingly, the predicted sites on Nef involved in the ABCA1 interaction overlap with those involved in the interaction with calnexin. The compounds previously shown to block Nef:calnexin interaction were among the top ranking ligands in docking simulations with ABCA1-interacting sites on Nef, suggesting the possibility that both interactions can be inhibited by the same chemical compounds. This study identifies a series of compounds for potential development as inhibitors of Nef-mediated co-morbidities of HIV infection.
- Published
- 2021
- Full Text
- View/download PDF
34. DeMAG predicts the effects of variants in clinically actionable genes by integrating structural and evolutionary epistatic features
- Author
-
Federica Luppino, Ivan A. Adzhubei, Christopher A. Cassa, and Agnes Toth-Petroczy
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
Despite an increasing use of genomic sequencing in clinical practice, interpretation of rare genetic variants remains challenging even in well-studied disease genes, resulting in many patients with Variants of Uncertain Significance (VUSs). Computational Variant Effect Predictors (VEPs) are currently used to provide valuable evidence in variant classifications, but they often misclassify benign variants, contributing to potential misdiagnoses. Here, we developed Deciphering Mutations in Actionable Genes (DeMAG), a supervised classifier for interpreting missense variants in actionable disease genes with improved performance over existing VEPs (20% decrease of false positive rate). Our tool has balanced specificity (82%) and sensitivity (94%) on clinical data, and the lowest misclassification rate on putatively benign variants among evaluated tools. DeMAG takes advantage of a novel epistatic feature, the ‘partners score’, which is based on evolutionary and structural partnerships of residues as estimated by evolutionary information and AlphaFold2 structural models. The ‘partners score’ as a general framework of epistatic interactions, can integrate not only clinical but functional information. We anticipate that our tool (demag.org) will facilitate the interpretation of variants and improve clinical decision-making.
- Published
- 2022
- Full Text
- View/download PDF
35. Comorbidities of HIV infection
- Author
-
Michael Bukrinsky, Nigora Mukhamedova, Alexander A. Makarov, D D Sviridov, and Alexei A. Adzhubei
- Subjects
0301 basic medicine ,Editorial Review ,Anti-HIV Agents ,Immunology ,Human immunodeficiency virus (HIV) ,HIV Infections ,Comorbidity ,comorbidities ,medicine.disease_cause ,Bioinformatics ,Virus ,Pathogenesis ,03 medical and health sciences ,Membrane Microdomains ,0302 clinical medicine ,Metabolic Diseases ,medicine ,Humans ,Immunology and Allergy ,Cognitive Dysfunction ,nef Gene Products, Human Immunodeficiency Virus ,030212 general & internal medicine ,Cholesterol metabolism ,Lipid raft ,Pathological ,lipid rafts ,Nef ,business.industry ,Macrophages ,HIV ,Biological Transport ,Antiretroviral therapy ,3. Good health ,Cholesterol ,030104 developmental biology ,Infectious Diseases ,Chronic disease ,Cardiovascular Diseases ,cholesterol metabolism ,extracellular vesicles ,business - Abstract
Combination antiretroviral therapy has dramatically changed the outcome of HIV infection, turning it from a death sentence to a manageable chronic disease. However, comorbidities accompanying HIV infection, such as metabolic and cardio-vascular diseases, as well as cognitive impairment, persist despite successful virus control by combination antiretroviral therapy and pose considerable challenges to clinical management of people living with HIV. These comorbidities involve a number of pathological processes affecting a variety of different tissues and cells, making it challenging to identify a common cause(s) that would link these different diseases to HIV infection. In this article, we will present evidence that impairment of cellular cholesterol metabolism may be a common factor driving pathogenesis of HIV-associated comorbidities. Potential implications for therapeutic approaches are discussed.
- Published
- 2020
- Full Text
- View/download PDF
36. Zn-dependent β-amyloid Aggregation and its Reversal by the Tetrapeptide HAEE
- Author
-
Evgeny Nudler, Alexander A. Makarov, Alexander S. Mironov, Sergey A. Kozin, Alexei A. Adzhubei, Vladimir I. Polshakov, Olga Katkova-Zhukotskaya, Bibhusita Pani, Svetlana Eremina, Evgeny P. Barykin, and Vladimir A. Mitkevich
- Subjects
Cell Biology ,Neurology (clinical) ,Geriatrics and Gerontology ,Pathology and Forensic Medicine - Published
- 2022
- Full Text
- View/download PDF
37. Zn-dependent β-amyloid Aggregation and its Reversal by the Tetrapeptide HAEE
- Author
-
Mitkevich, Vladimir A., primary, Barykin, Evgeny P., primary, Eremina, Svetlana, primary, Pani, Bibhusita, primary, Katkova-Zhukotskaya, Olga, primary, Polshakov, Vladimir I., primary, Adzhubei, Alexei A., primary, Kozin, Sergey A., primary, Mironov, Alexander S., primary, Makarov, Alexander A., primary, and Nudler, Evgeny, primary
- Published
- 2022
- Full Text
- View/download PDF
38. Zinc Induced Aβ16 Aggregation Modeled by Molecular Dynamics
- Author
-
Alexei A. Adzhubei, Alexander A. Makarov, and Anna P. Tolstova
- Subjects
Aβ16 ,QH301-705.5 ,chemistry.chemical_element ,Metal Binding Site ,Peptide ,Zinc ,Molecular Dynamics Simulation ,Catalysis ,Article ,Inorganic Chemistry ,chemistry.chemical_compound ,Residue (chemistry) ,Molecular dynamics ,Protein Aggregates ,Humans ,Physical and Theoretical Chemistry ,Biology (General) ,Molecular Biology ,QD1-999 ,Spectroscopy ,chemistry.chemical_classification ,Amyloid beta-Peptides ,metal binding site ,Zinc ion ,MD ,Organic Chemistry ,beta-amyloid ,aggregation ,General Medicine ,Peptide Fragments ,Computer Science Applications ,Chemistry ,Monomer ,zinc ion ,chemistry ,Polymerization ,Biophysics ,Protein Multimerization - Abstract
It is widely accepted that the addition of zinc leads to the formation of neurotoxic nonfibrillar aggregates of beta-amyloid peptides Aβ40 and Aβ42 and at the same time destabilizes amyloid fibrils. However, the mechanism of the effect of zinc on beta-amyloid is not fully understood. In this study, a fast zinc-induced aggregation of Aβ16 (as compared to a system without zinc) via the formation of Aβ16 dimers with one zinc ion coordinated in the metal-binding site 11EVHH14, followed by their polymerization, has been studied by molecular dynamics. The best aggregation was shown by the system composed of Aβ16 dimers bound by one zinc ion, with no additional zinc in solution. The presence of Aβ16 dimers was a major condition, sufficient for fast aggregation into larger complexes. It has been shown that the addition of zinc to a system with already formed dimers does not substantially affect the characteristics and rate of aggregation. At the same time, an excessive concentration of zinc at the early stages of the formation of conglomerates can negatively affect aggregation, since in systems where zinc ions occupied the 11EVHH14 coordination center and the His6 residue of every Aβ16 monomer, the aggregation proceeded more slowly and the resulting complexes were not as large as in the zinc-free Aβ system. Thus, this study has shown that the formation of Aβ16 dimers bound through zinc ions at the 11EVHH14 sites of the peptides plays an important role in the formation of neurotoxic non-fibrillar aggregates of beta-amyloid peptide Aβ16. The best energetically favorable structure has been obtained for the complex of two Aβ16 dimers with two zinc ions.
- Published
- 2021
39. Zn-dependent β-amyloid Aggregation and its Reversal by the Tetrapeptide HAEE.
- Author
-
Mitkevich, Vladimir A., Barykin, Evgeny P., Eremina, Svetlana, Pani, Bibhusita, Katkova-Zhukotskaya, Olga, Polshakov, Vladimir I., Adzhubei, Alexei A., Kozin, Sergey A., Mironov, Alexander S., Makarov, Alexander A., and Nudler, Evgeny
- Subjects
ALZHEIMER'S disease ,CEREBRAL amyloid angiopathy ,OLIGOMERS - Abstract
The pathogenesis of Alzheimer's disease (AD) is associated with the formation of cerebral amyloid plaques, the main components of which are the modified Aß molecules as well as the metal ions. Aß isomerized at Asp7 residue (isoD7-Aß) is the most abundant isoform in amyloid plaques. We hypothesized that the pathogenic effect of isoD7-Aß is due to the formation of zinc-dependent oligomers, and that this interaction can be disrupted by the rationally designed tetrapeptide (HAEE). Here, we utilized surface plasmon resonance, nuclear magnetic resonance, and molecular dynamics simulation to demonstrate Zn2+-dependent oligomerization of isoD7-Aß and the formation of a stable isoD7-Aß:Zn2+:HAEE complex incapable of forming oligomers. To demonstrate the physiological importance of zinc-dependent isoD7-Aß oligomerization and the ability of HAEE to interfere with this process at the organismal level, we employed transgenic nematodes overexpressing human Aß. We show that the presence of isoD7-Aß in the medium triggers extensive amyloidosis that occurs in a Zn2+-dependent manner, enhances paralysis, and shortens the animals' lifespan. Exogenous HAEE completely reverses these pathological effects of isoD7-Aß. We conclude that the synergistic action of isoD7-Aß and Zn2+ promotes Aß aggregation and that the selected small molecules capable of interrupting this process, such as HAEE, can potentially serve as anti-amyloid therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. Docking and Molecular Dynamics-Based Identification of Interaction between Various Beta-Amyloid Isoforms and RAGE Receptor
- Author
-
Anna P. Tolstova, Alexei A. Adzhubei, Vladimir A. Mitkevich, Irina Yu. Petrushanko, and Alexander A. Makarov
- Subjects
Amyloid beta-Peptides ,Alzheimer’s disease ,beta-amyloid ,RAGE ,interaction interface ,blood–brain barrier ,transcytosis ,molecular dynamics ,macromolecular docking ,Receptor for Advanced Glycation End Products ,Organic Chemistry ,General Medicine ,Molecular Dynamics Simulation ,Ligands ,Peptide Fragments ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Alzheimer Disease ,Blood-Brain Barrier ,Humans ,Protein Isoforms ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy - Abstract
Beta-amyloid peptide (Aβ) is a ligand associated with RAGE (Advanced glycosylation end product-specific receptor). Aβ is translocated in complexes with RAGE from the blood to brain across the blood–brain barrier (BBB) by transcytosis. Aβ and its isoforms are important factors in the Alzheimer’s disease (AD) pathogenesis. However, interaction with RAGE was previously studied for Aβ but not for its isoforms. The present study has been directed at identifying the key interaction interfaces between RAGE and Aβ isoforms (Aβ40, Aβ42, phosphorylated and isomerized isoforms pS8-Aβ42, isoD7-Aβ42). Two interfaces have been identified by docking: they are represented by an extended area at the junction of RAGE domains V and C1 and a smaller area linking C1 and C2 domains. Molecular dynamics (MD) simulations have shown that all Aβ isoforms form stable and tightly bound complexes. This indicates that all Aβ isoforms potentially can be transported through the cell as part of a complex with RAGE. Modeling of RAGE interaction interfaces with Aβ indicates which chemical compounds can potentially be capable of blocking this interaction, and impair the associated pathogenic cascades. The ability of three RAGE inhibitors (RAP, FPS-ZM1 and RP-1) to disrupt the RAGE:Aβ interaction has been probed by docking and subsequently the complexes’ stability verified by MD. The RP-1 and Aβ interaction areas coincide and therefore this inhibitor is very promising for the RAGE:Aβ interaction inhibition.
- Published
- 2022
- Full Text
- View/download PDF
41. Zinc Induced Aβ16 Aggregation Modeled by Molecular Dynamics
- Author
-
Tolstova, Anna P., primary, Makarov, Alexander A., additional, and Adzhubei, Alexei A., additional
- Published
- 2021
- Full Text
- View/download PDF
42. Beta-amyloid induces apoptosis of neuronal cells by inhibition of the Arg/N-end rule pathway proteolytic activity
- Author
-
Olga I. Kechko, Konstantin I. Piatkov, Vladimir A. Mitkevich, Alexei A. Adzhubei, Christopher S. Brower, Alexander A. Makarov, Alexey Moskalev, and Irina Yu. Petrushanko
- Subjects
Aging ,Programmed cell death ,Proteasome Endopeptidase Complex ,Amyloid ,Arginyltransferase ,N-end rule ,Peptide ,Protein degradation ,Mice ,Cell Line, Tumor ,medicine ,Animals ,chemistry.chemical_classification ,Neurons ,age-related disease ,Amyloid beta-Peptides ,Chemistry ,Neurodegeneration ,apoptosis ,Cell Biology ,medicine.disease ,Aminoacyltransferases ,Cell biology ,Apoptosis ,Proteolysis ,protein degradation ,Ate1 ,Alzheimer’s disease ,Research Paper - Abstract
Alzheimer's disease (AD) is accompanied by the dysfunction of intracellular protein homeostasis systems, in particular the ubiquitin-proteasome system (UPS). Beta-amyloid peptide (Aβ), which is involved in the processes of neurodegeneration in AD, is a substrate of this system, however its effect on UPS activity is still poorly explored. Here we found that Aβ peptides inhibited the proteolytic activity of the antiapoptotic Arg/N-end rule pathway that is a part of UPS. We identified arginyltransferase Ate1 as a specific component of the Arg/N-end rule pathway targeted by Aβs. Aβ bearing the familial English H6R mutation, known to cause early-onset AD, had an even greater inhibitory effect on protein degradation through the Arg/N-end rule pathway than intact Aβ. This effect was associated with a significant decrease in Ate1-1 and Ate1-3 catalytic activity. We also found that the loss of Ate1 in neuroblastoma Neuro-2a cells eliminated the apoptosis-inducing effects of Aβ peptides. Together, our results show that the apoptotic effect of Aβ peptides is linked to their impairment of Ate1 catalytic activity leading to suppression of the Arg/N-end rule pathway proteolytic activity and ultimately cell death.
- Published
- 2019
43. Pharmacokinetics and Molecular Modeling Indicate nAChRα4-Derived Peptide HAEE Goes through the Blood–Brain Barrier
- Author
-
Stanislav I Shram, Sergey A. Kozin, Alexander A. Makarov, Yurii A Zolotarev, Alexei A. Adzhubei, Anna P. Tolstova, Alexander K Dadayan, Oleg Talibov, Vladimir A. Mitkevich, and Nikolai F Myasoyedov
- Subjects
0301 basic medicine ,Male ,Models, Molecular ,Biodistribution ,LRP1 ,Peptide ,Pharmacology ,Receptors, Nicotinic ,Blood–brain barrier ,blood–brain barrier ,Biochemistry ,Microbiology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Pharmacokinetics ,Alzheimer Disease ,medicine ,Animals ,Rats, Wistar ,Molecular Biology ,chemistry.chemical_classification ,Amyloid beta-Peptides ,Tetrapeptide ,Chemistry ,beta-amyloid ,peptide drug ,Brain ,Biological Transport ,Peptide Fragments ,QR1-502 ,Rats ,Mice, Inbred C57BL ,Nicotinic acetylcholine receptor ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Transcytosis ,Blood-Brain Barrier ,α4β2 nicotinic acetylcholine receptor ,Alzheimer’s disease ,receptor-mediated transcytosis ,Rabbits ,Peptides ,030217 neurology & neurosurgery - Abstract
One of the treatment strategies for Alzheimer’s disease (AD) is based on the use of pharmacological agents capable of binding to beta-amyloid (Aβ) and blocking its aggregation in the brain. Previously, we found that intravenous administration of the synthetic tetrapeptide Acetyl-His-Ala-Glu-Glu-Amide (HAEE), which is an analogue of the 35–38 region of the α4 subunit of α4β2 nicotinic acetylcholine receptor and specifically binds to the 11–14 site of Aβ, reduced the development of cerebral amyloidogenesis in a mouse model of AD. In the current study on three types of laboratory animals, we determined the biodistribution and tissue localization patterns of HAEE peptide after single intravenous bolus administration. The pharmacokinetic parameters of HAEE were established using uniformly tritium-labeled HAEE. Pharmacokinetic data provided evidence that HAEE goes through the blood–brain barrier. Based on molecular modeling, a role of LRP1 in receptor-mediated transcytosis of HAEE was proposed. Altogether, the results obtained indicate that the anti-amyloid effect of HAEE, previously found in a mouse model of AD, most likely occurs due to its interaction with Aβ species directly in the brain.
- Published
- 2021
44. Pharmacokinetics and Molecular Modeling Indicate nAChRα4-Derived Peptide HAEE Goes through the Blood–Brain Barrier
- Author
-
Zolotarev, Yurii A., primary, Mitkevich, Vladimir A., additional, Shram, Stanislav I., additional, Adzhubei, Alexei A., additional, Tolstova, Anna P., additional, Talibov, Oleg B., additional, Dadayan, Alexander K., additional, Myasoyedov, Nikolai F., additional, Makarov, Alexander A., additional, and Kozin, Sergey A., additional
- Published
- 2021
- Full Text
- View/download PDF
45. Interaction Interface of Aβ 42 with Human Na,K-ATPase Studied by MD and ITC and Inhibitor Screening by MD.
- Author
-
Adzhubei, Alexei A., Tolstova, Anna P., Strelkova, Maria A., Mitkevich, Vladimir A., Petrushanko, Irina Yu., and Makarov, Alexander A.
- Subjects
ISOTHERMAL titration calorimetry ,ALZHEIMER'S disease ,MOLECULAR dynamics ,MEMORY disorders ,NEURODEGENERATION - Abstract
Alzheimer's disease (AD) is a neurodegenerative disease accompanied by progressive cognitive and memory dysfunction due to disruption of normal electrotonic properties of neurons and neuronal loss. The Na,K-ATPase interaction with beta amyloid (Aβ) plays an important role in AD pathogenesis. It has been shown that Na,K-ATPase activity in the AD brain was significantly lower than those in age-matched control brain. The interaction of Aβ
42 with Na,K-ATPase and subsequent oligomerization leads to inhibition of the enzyme activity. In this study interaction interfaces between three common Aβ42 isoforms, and different conformations of human Na,K-ATPase (α1β1) have been obtained using molecular modeling, including docking and molecular dynamics (MD). Interaction sites of Na,K-ATPase with Aβ42 are localized between extracellular parts of α- and β- subunits and are practically identical for Na,K-ATPase at different conformations. Thermodynamic parameters for the formation of Na,K-ATPase:Aβ42 complex at different conformations acquired by isothermal titration calorimetry (ITC) are similar, which is in line with the data of molecular modeling. Similarity of Na,K-ATPase interaction interfaces with Aβ in all conformations allowed us to cross-screen potential inhibitors for this interaction and find pharmaceutical compounds that could block it. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
46. Identification of α4β2 nAChR interaction site with Aβ 42 and development of tetrapeptide capable of breaking this interaction
- Author
-
Vladimir A. Mitkevich, Irina V. Shelukhina, Victor I. Tsetlin, Alexei A. Adzhubei, Sergey A. Kozin, Evgeny P. Barykin, Alexander A. Makarov, and Alexandra I. Garifulina
- Subjects
Tetrapeptide ,Epidemiology ,Chemistry ,Health Policy ,α4β2 nachr ,Interaction site ,Cell biology ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Identification (biology) ,Neurology (clinical) ,Geriatrics and Gerontology ,Receptor - Published
- 2020
- Full Text
- View/download PDF
47. The Post-Translational Modifications, Localization, and Mode of Attachment of Non-Covalently Bound Glucanosyltransglycosylases of Yeast Cell Wall as a Key to Understanding their Functioning
- Author
-
Irina B. Kudryashova, Yaroslav V. Tkachev, Alice V. Alessenko, Sergei A. Kuznetsov, Alexei A. Adzhubei, Valentina V. Rekstina, Fanis A Sabirzyanov, Natalia E Snalina, Tatyana A. Sabirzyanova, Rustam H. Ziganshin, Tatyana S. Kalebina, and Anastasia A Bykova
- Subjects
0301 basic medicine ,Antifungal Agents ,Glycosylation ,Saccharomyces cerevisiae Proteins ,Bgl2 ,Genes, Fungal ,Molecular Conformation ,Saccharomyces cerevisiae ,Catalysis ,Article ,lcsh:Chemistry ,Inorganic Chemistry ,Cell wall ,03 medical and health sciences ,Bacterial microcompartment ,Cell Wall ,Transferases ,post-translational modifications ,Molecule ,Amino Acid Sequence ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Glucans ,Spectroscopy ,Glucan ,chemistry.chemical_classification ,030102 biochemistry & molecular biology ,Glucan Endo-1,3-beta-D-Glucosidase ,Organic Chemistry ,General Medicine ,microcompartments ,Yeast ,Computer Science Applications ,Scw4 ,030104 developmental biology ,Enzyme ,lcsh:Biology (General) ,lcsh:QD1-999 ,chemistry ,Covalent bond ,Glucosyltransferases ,glucanosyltransglycosylases ,Biophysics ,Phosphorylation ,Protein Processing, Post-Translational ,Glucosidases - Abstract
Glucan linked to proteins is a natural mega-glycoconjugate (mGC) playing the central role as a structural component of a yeast cell wall (CW). Regulation of functioning of non-covalently bound glucanosyltransglycosylases (ncGTGs) that have to remodel mGC to provide CW extension is poorly understood. We demonstrate that the main ncGTGs Bgl2 and Scw4 have phosphorylated and glutathionylated residues and are represented in CW as different pools of molecules having various firmness of attachment. Identified pools contain Bgl2 molecules with unmodified peptides, but differ from each other in the presence and combination of modified ones, as well as in the presence or absence of other CW proteins. Correlation of Bgl2 distribution among pools and its N-glycosylation was not found. Glutathione affects Bgl2 conformation, probably resulting in the mode of its attachment and enzymatic activity. Bgl2 from the pool of unmodified and monophosphorylated molecules demonstrates the ability to fibrillate after isolation from CW. Revealing of Bgl2 microcompartments and their mosaic arrangement summarized with the results obtained give the evidence that the functioning of ncGTGs in CW can be controlled by reversible post-translational modifications and facilitated due to their compact localization. The hypothetical scheme of distribution of Bgl2 inside CW is represented.
- Published
- 2020
48. Erratum for Dubrovsky et al., 'Inhibition of HIV Replication by Apolipoprotein A-I Binding Protein Targeting the Lipid Rafts'
- Author
-
Michael Bukrinsky, Beda Brichacek, Nigora Mukhamedova, Richard B. Jones, Larisa Dubrovsky, Christophe Vanpouille, Leonid Margolis, Soo-Ho Choi, Yury I. Miller, Adam R. Ward, Alexei A. Adzhubei, Tatiana Pushkarsky, and Dmitri Sviridov
- Subjects
Apolipoprotein A-I-Binding Protein ,0303 health sciences ,030306 microbiology ,Chemistry ,Human immunodeficiency virus (HIV) ,medicine.disease_cause ,Virology ,Microbiology ,QR1-502 ,03 medical and health sciences ,Replication (statistics) ,medicine ,Erratum ,Lipid raft ,030304 developmental biology - Abstract
Apolipoprotein A-I binding protein (AIBP) is a protein involved in regulation of lipid rafts and cholesterol efflux. AIBP has been suggested to function as a protective factor under several sets of pathological conditions associated with increased abundance of lipid rafts, such as atherosclerosis and acute lung injury. Here, we show that exogenously added AIBP reduced the abundance of lipid rafts and inhibited HIV replication
- Published
- 2020
49. Inhibition of HIV Replication by Apolipoprotein A-I Binding Protein Targeting the Lipid Rafts
- Author
-
Adam R. Ward, Nigora Mukhamedova, Soo-Ho Choi, Michael Bukrinsky, Dmitri Sviridov, Larisa Dubrovsky, Alexei A. Adzhubei, Tatiana Pushkarsky, Beda Brichacek, Christophe Vanpouille, Richard B. Jones, Yury I. Miller, and Leonid Margolis
- Subjects
fusion ,exosomes ,Lung injury ,Microbiology ,Host-Microbe Biology ,AIBP ,03 medical and health sciences ,hla ,0302 clinical medicine ,Downregulation and upregulation ,Virology ,Gene silencing ,Lipid raft ,030304 developmental biology ,lipid rafts ,0303 health sciences ,Gene knockdown ,Nef ,Chemistry ,Binding protein ,virus diseases ,HIV ,Microvesicles ,QR1-502 ,3. Good health ,Cell biology ,lipids (amino acids, peptides, and proteins) ,Cell activation ,extracellular vesicles ,030217 neurology & neurosurgery ,Research Article - Abstract
Apolipoprotein A-I binding protein (AIBP) is a recently identified innate anti-inflammatory factor. Here, we show that AIBP inhibited HIV replication by targeting lipid rafts and reducing virus-cell fusion. Importantly, AIBP selectively reduced levels of rafts on cells stimulated by an inflammatory stimulus or treated with extracellular vesicles containing HIV-1 protein Nef without affecting rafts on nonactivated cells. Accordingly, fusion of monocyte-derived macrophages with HIV was sensitive to AIBP only in the presence of Nef. Silencing of endogenous AIBP significantly upregulated HIV-1 replication. Interestingly, HIV-1 replication in cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, was not inhibited by AIBP. These results suggest that AIBP is an innate anti-HIV factor that targets virus-cell fusion., Apolipoprotein A-I binding protein (AIBP) is a protein involved in regulation of lipid rafts and cholesterol efflux. AIBP has been suggested to function as a protective factor under several sets of pathological conditions associated with increased abundance of lipid rafts, such as atherosclerosis and acute lung injury. Here, we show that exogenously added AIBP reduced the abundance of lipid rafts and inhibited HIV replication in vitro as well as in HIV-infected humanized mice, whereas knockdown of endogenous AIBP increased HIV replication. Endogenous AIBP was much more abundant in activated T cells than in monocyte-derived macrophages (MDMs), and exogenous AIBP was much less effective in T cells than in MDMs. AIBP inhibited virus-cell fusion, specifically targeting cells with lipid rafts mobilized by cell activation or Nef-containing exosomes. MDM-HIV fusion was sensitive to AIBP only in the presence of Nef provided by the virus or exosomes. Peripheral blood mononuclear cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, bound less AIBP than cells from donors with other HLA genotypes and were not protected by AIBP from rapid HIV-1 replication. These results provide the first evidence for the role of Nef exosomes in regulating HIV-cell fusion by modifying lipid rafts and suggest that AIBP is an innate factor that restricts HIV replication by targeting lipid rafts.
- Published
- 2020
- Full Text
- View/download PDF
50. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project
- Author
-
Birney, Ewan, Stamatoyannopoulos, John A., Dutta, Anindya, Guigo, Roderic, Gingeras, Thomas R., Margulies, Elliott H., Weng, Zhiping, Snyder, Michael, Dermitzakis, Emmanouil T., Thurman, Robert E., Kuehn, Michael S., Taylor, Christopher M., Neph, Shane, Koch, Christoph M., Asthana, Saurabh, Malhotra, Ankit, Adzhubei, Ivan, Greenbaum, Jason A., Andrews, Robert M., Flicek, Paul, Boyle, Patrick J., Cao, Hua, Carter, Nigel P., Clelland, Gayle K., Davis, Sean, Day, Nathan, Dhami, Pawandeep, Dillon, Shane C., Dorschner, Michael O., Fiegler, Heike, Giresi, Paul G., Goldy, Jeff, Hawrylycz, Michael, Haydock, Andrew, Humbert, Richard, James, Keith D., Johnson, Brett E., Johnson, Ericka M., Frum, Tristan T., Rosenzweig, Elizabeth R., Karnani, Neerja, Lee, Kirsten, Lefebvre, Gregory C., Navas, Patrick A., Neri, Fidencio, Parker, Stephen C. J., Sabo, Peter J., Sandstrom, Richard, Shafer, Anthony, Vetrie, David, Weaver, Molly, Wilcox, Sarah, Yu1, Man, Collins, Francis S., Dekker, Job, Lieb, Jason D., Tullius, Thomas D., Crawford, Gregory E., Sunyaev, Shamil, Noble, William S., Dunham, Ian, Denoeud, France, Reymond, Alexandre, Kapranov, Philipp, Rozowsky, Joel, Zheng, Deyou, Castelo, Robert, Frankish, Adam, Harrow, Jennifer, Ghosh, Srinka, Sandelin, Albin, Hofacker, Ivo L., Baertsch, Robert, Keefe, Damian, Dike, Sujit, Cheng, Jill, Hirsch, Heather A., Sekinger, Edward A., Lagarde, Julien, Abril, Josep F., Shahab, Atif, Flamm, Christoph, Fried, Claudia, Hackermuller, Jorg, Hertel, Jana, Lindemeyer, Manja, Missal, Kristin, Tanzer, Andrea, Washietl, Stefan, Korbel, Jan, Emanuelsson, Olof, Pedersen, Jakob S., Holroyd, Nancy, Taylor, Ruth, Swarbreck, David, Matthews, Nicholas, Dickson, Mark C., Thomas, Daryl J., Weirauch, Matthew T., Gilbert, James, Drenkow, Jorg, Bell, Ian, Zhao, XiaoDong, Srinivasan, K.G., Sung, Wing-Kin, Ooi, Hong Sain, Chiu, Kuo Ping, Foissac, Sylvain, Alioto, Tyler, Brent, Michael, Pachter, Lior, Tress, Michael L., Valencia, Alfonso, Choo, Siew Woh, Choo, Chiou Yu, Ucla, Catherine, Manzano, Caroline, Wyss, Carine, Cheung, Evelyn, Clark, Taane G., Brown, James B., Ganesh, Madhavan, Patel, Sandeep, Tammana, Hari, Chrast, Jacqueline, Henrichsen, Charlotte N., Kai, Chikatoshi, Kawai, Jun, Nagalakshmi, Ugrappa, Wu, Jiaqian, Lian, Zheng, Lian, Jin, Newburger, Peter, Zhang, Xueqing, Bickel, Peter, Mattick, John S., Carninci, Piero, Hayashizaki, Yoshihide, Weissman, Sherman, Hubbard, Tim, Myers, Richard M., Rogers, Jane, Stadler, Peter F., Lowe, Todd M., Wei, Chia-Lin, Ruan, Yijun, Struhl, Kevin, Gerstein, Mark, Antonarakis, Stylianos E., Fu, Yutao, Green, Eric D., Karaoz, Ulaş, Siepel, Adam, Taylor, James, Liefer, Laura A., Wetterstrand, Kris A., Good, Peter J., Feingold, Elise A., Guyer, Mark S., Cooper, Gregory M., Asimenos, George, Dewey, Colin N., Hou, Minmei, Nikolaev, Sergey, Montoya-Burgos, Juan I., Loytynoja, Ari, Whelan, Simon, Pardi, Fabio, Massingham, Tim, Huang, Haiyan, Zhang, Nancy R., Holmes, Ian, Mullikin, James C., Ureta-Vidal, Abel, Paten, Benedict, Seringhaus, Michael, Church, Deanna, Rosenbloom, Kate, Kent, W. James, Stone, Eric A., Batzoglou, Serafim, Goldman, Nick, Hardison, Ross C., Haussler, David, Miller, Webb, Sidow, Arend, Trinklein, Nathan D., Zhang, Zhengdong D., Barrera, Leah, Stuart, Rhona, King, David C., Ameur, Adam, Enroth, Stefan, Bieda, Mark C., Kim, Jonghwan, Bhinge, Akshay A., Jiang, Nan, Liu, Jun, Yao, Fei, Vega, Vinsensius B., Lee, Charlie W.H., Ng, Patrick, Yang, Annie, Moqtaderi, Zarmik, Zhu, Zhou, Xu, Xiaoqin, Squazzo, Sharon, Oberley, Matthew J., Inman, David, Singer, Michael A., Richmond, Todd A., Munn, Kyle J., Rada-Iglesias, Alvaro, Wallerman, Ola, Komorowski, Jan, Fowler, Joanna C., Couttet, Phillippe, Bruce, Alexander W., Dovey, Oliver M., Ellis, Peter D., Langford, Cordelia F., Nix, David A., Euskirchen, Ghia, Hartman, Stephen, Urban, Alexander E., Kraus, Peter, Van Calcar, Sara, Heintzman, Nate, Hoon Kim, Tae, Wang, Kun, Qu, Chunxu, Hon, Gary, Luna, Rosa, Glass, Christopher K., Rosenfeld, M. Geoff, Aldred, Shelley Force, Cooper, Sara J., Halees, Anason, Lin, Jane M., Shulha, Hennady P., Zhang, Xiaoling, Xu, Mousheng, Haidar, Jaafar N. S., Yu, Yong, Birney*, Ewan, Iyer, Vishwanath R., Green, Roland D., Wadelius, Claes, Farnham, Peggy J., Ren, Bing, Harte, Rachel A., Hinrichs, Angie S., Trumbower, Heather, Clawson, Hiram, Hillman-Jackson, Jennifer, Zweig, Ann S., Smith, Kayla, Thakkapallayil, Archana, Barber, Galt, Kuhn, Robert M., Karolchik, Donna, Armengol, Lluis, Bird, Christine P., de Bakker, Paul I. W., Kern, Andrew D., Lopez-Bigas, Nuria, Martin, Joel D., Stranger, Barbara E., Woodroffe, Abigail, Davydov, Eugene, Dimas, Antigone, Eyras, Eduardo, Hallgrimsdottir, Ingileif B., Huppert, Julian, Zody, Michael C., Abecasis, Goncalo R., Estivill, Xavier, Bouffard, Gerard G., Guan, Xiaobin, Hansen, Nancy F., Idol, Jacquelyn R., Maduro, Valerie V.B., Maskeri, Baishali, McDowell, Jennifer C., Park, Morgan, Thomas, Pamela J., Young, Alice C., Blakesley, Robert W., Baylor College of Medicine, Human Genome Sequencing Center, Muzny, Donna M., Sodergren, Erica, Wheeler, David A., Worley, Kim C., Jiang, Huaiyang, Weinstock, George M., Gibbs, Richard A., Graves, Tina, Fulton, Robert, Mardis, Elaine R., Wilson, Richard K., Clamp, Michele, Cuff, James, Gnerre, Sante, Jaffe, David B., Chang, Jean L., Lindblad-Toh, Kerstin, Lander, Eric S., Koriabine, Maxim, Nefedov, Mikhail, Osoegawa, Kazutoyo, Yoshinaga, Yuko, Zhu, Baoli, and de Jong, Pieter J.
- Subjects
Environmental issues ,Science and technology ,Zoology and wildlife conservation - Abstract
Author(s): The ENCODE Project Consortium; Analysis Coordination; Ewan Birney (corresponding author) [1]; John A. Stamatoyannopoulos (corresponding author) [2]; Anindya Dutta (corresponding author) [3]; Roderic Guigó (corresponding author) [4, 5]; Thomas [...]
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.