Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Bioconjug Chem ; 35(3): 312-323, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38420925

ABSTRACT

Developing effective amyloidosis inhibitors poses a significant challenge due to the dynamic nature of the protein structures, the complex interplay of interfaces in protein-protein interactions, and the irreversible nature of amyloid assembly. The interactions of amyloidogenic polypeptides with other peptides play a pivotal role in modulating amyloidosis and fibril formation. This study presents a novel approach for designing and synthesizing amyloid interaction surfaces using segments derived from the amyloid-promoting sequence of amyloid ß-peptide [VF(Aß(18-19)/FF(Aß(19-20)/LVF(Aß(17-19)/LVFF(Aß(17-20)], where VF, FF, LVF and LVFF stands for valine phenylalanine dipeptide, phenylalanine phenylalanine dipeptide, leucine valine phenylalanine tripeptide and leucine valine phenylalanine phenylalanine tetrapeptide, respectively. These segments are conjugated with side-chain proline-based methacrylate polymers serving as potent lysozyme amyloidosis inhibitors and demonstrating reduced cytotoxicity of amyloid aggregations. Di-, tri-, and tetra-peptide conjugated chain transfer agents (CTAs) were synthesized and used for the reversible addition-fragmentation chain transfer polymerization of tert-butoxycarbonyl (Boc)-proline methacryloyloxyethyl ester (Boc-Pro-HEMA). Deprotection of Boc-groups from the side-chain proline pendants resulted in water-soluble polymers with defined peptide chain ends as peptide-polymer bioconjugates. Among them, the LVFF-conjugated polymer acted as a potent inhibitor with significantly suppressed lysozyme amyloidosis, a finding supported by comprehensive spectroscopic, microscopic, and computational analyses. These results unveil the synergistic effect between the segment-derived amyloid ß-peptide and side-chain proline-based polymers, offering new prospects for targeting lysozyme amyloidosis.


Subject(s)
Amyloid beta-Peptides , Amyloidosis , Humans , Amyloid beta-Peptides/chemistry , Proline , Leucine , Polymers/chemistry , Muramidase , Amyloidosis/drug therapy , Amyloidosis/metabolism , Amyloid , Dipeptides/pharmacology , Phenylalanine , Valine
2.
J Biol Chem ; 300(1): 105551, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38072045

ABSTRACT

Bromodomain-PHD finger protein 1 (BRPF1) belongs to the BRPF family of bromodomain-containing proteins. Bromodomains are exclusive reader modules that recognize and bind acetylated histones and non-histone transcription factors to regulate gene expression. The biological functions of acetylated histone recognition by BRPF1 bromodomain are well characterized; however, the function of BRPF1 regulation via non-histone acetylation is still unexplored. Therefore, identifying the non-histone interactome of BRPF1 is pivotal in deciphering its role in diverse cellular processes, including its misregulation in diseases like cancer. Herein, we identified the non-histone interacting partners of BRPF1 utilizing a protein engineering-based approach. We site-specifically introduced the unnatural photo-cross-linkable amino acid 4-azido-L-phenylalanine into the bromodomain of BRPF1 without altering its ability to recognize acetylated histone proteins. Upon photoirradiation, the engineered BRPF1 generates a reactive nitrene species, cross-linking interacting partners with spatio-temporal precision. We demonstrated the robust cross-linking efficiency of the engineered variant with reported histone ligands of BRPF1 and further used the variant reader to cross-link its interactome. We also characterized novel interacting partners by proteomics, suggesting roles for BRPF1 in diverse cellular processes. BRPF1 interaction with interleukin enhancer-binding factor 3, one of these novel interacting partners, was further validated by isothermal titration calorimetry and co-IP. Lastly, we used publicly available ChIP-seq and RNA-seq datasets to understand the colocalization of BRPF1 and interleukin enhancer-binding factor 3 in regulating gene expression in the context of hepatocellular carcinoma. Together, these results will be crucial for full understanding of the roles of BRPF1 in transcriptional regulation and in the design of small-molecule inhibitors for cancer treatment.


Subject(s)
Adaptor Proteins, Signal Transducing , Azides , Bromodomain Containing Proteins , DNA-Binding Proteins , Acetylation , Adaptor Proteins, Signal Transducing/metabolism , Azides/metabolism , Histones/metabolism , Interleukins/metabolism , Protein Binding , Humans , Bromodomain Containing Proteins/metabolism , DNA-Binding Proteins/metabolism
3.
J Mol Graph Model ; 126: 108642, 2024 01.
Article in English | MEDLINE | ID: mdl-37797430

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most aggressive and life-threatening cancers. Although multiple treatment options are available, the prognosis of HCC patients is poor due to metastasis and drug resistance. Hence, discovering novel targets is essential for better therapeutic development for HCC. In this study, we used the cancer genome atlas (TCGA) dataset to analyze the expression of bromodomain-containing proteins in HCC, as bromodomains are emerging attractive therapeutic targets. Our analysis identified BRPF1 as the most highly upregulated gene in HCC among the 43 bromodomain-containing genes. Upregulation of BRPF1 was significantly associated with poorer patient survival. Therefore, targeting BRPF1 may be an approach for HCC treatment. Previously, several potential inhibitors of BRPF1 bromodomain have been discovered. However, due to the limited clinical success of the current inhibitors, we aim to search for new inhibitors with high affinity and specificity for the BRPF1 bromodomain. In this study, we utilized high-throughput virtual screening methods to screen synthetic and natural compound databases against the BRPF1 bromodomain. In addition, we used machine learning-based QSAR modeling to predict the IC50 values of the selected BRPF1 bromodomain inhibitors. Extensive MD simulations were used to calculate the binding free energies of BRPF1 bromodomain and inhibitor complexes. Using this approach, we identified four lead scaffolds with a similar or better binding affinity towards the BRPF1 bromodomain than the previously reported inhibitors. Overall, this study discovered some promising compounds that have the potential to act as potent BRPF1 bromodomain inhibitors.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Nuclear Proteins/chemistry , DNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Liver Neoplasms/drug therapy
5.
Biochem J ; 480(9): 629-647, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37075063

ABSTRACT

TRIM24 is a multi-functional chromatin reader, and it binds to the estrogen receptor to activate estrogen-dependent target genes associated with tumor development. TRIM24 is known to ubiquitinate p53 via an N-terminal RING domain and binds a specific combinatorial histone signature of H3K4me0/H3K23ac via its C-terminal plant homeodomain (PHD) and bromodomain (Bromo). Aberrant expression of TRIM24 positively correlates with H3K23ac levels, and high levels of both TRIM24 and H3K23ac predict poor survival of breast cancer patients. Little has been explored about the acetylated histone H4 (H4ac) signatures of TRIM24 and their biological functions. Herein, we report novel H4ac binding partners of TRIM24 and their localization in the genome. Isothermal titration calorimetry binding assay on the histone peptides revealed that the TRIM24 PHD-Bromo preferably binds to H4K5ac, H4K8ac, and H4K5acK8ac compared with other acetylated histone H4 ligands. Co-immunoprecipitation on the endogenous histones suggests that the recognition of H4ac by Bromo does not interfere with the recognition of H3K4me0 mark by the PHD domain of TRIM24. Consistent with this, TRIM24 PHD-Bromo exhibits minimal discrimination among H4ac binding partners at endogenous histone and nucleosome levels. Moreover, ChIP-seq analysis revealed that the H4K5ac and H4K8ac histone signatures strongly co-localize near the transcription start sites of different hub genes or TRIM24-targeted genes in breast cancer. In addition, the KEGG pathway analysis demonstrates that the TRIM24 and its H4ac targets are associated with several important biological pathways. Our findings describe that the H4ac recognition by TRIM24 PHD-Bromo enables access to the chromatin for specific transcriptional regulation.


Subject(s)
Breast Neoplasms , Histones , Humans , Female , Histones/metabolism , Chromatin , DNA-Binding Proteins/metabolism , Protein Domains , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Acetylation , Protein Binding , Carrier Proteins/metabolism
6.
Biomater Sci ; 11(10): 3574-3588, 2023 May 16.
Article in English | MEDLINE | ID: mdl-37000481

ABSTRACT

To address the obstacles in insulin protein homeostasis leading to the formation of neurotoxic amyloid plaques associated with different diseases, herein we have synthesized block copolymers using the reversible addition-fragmentation chain transfer (RAFT) polymerization method, composed of tert-butoxycarbonyl (Boc) protected leucine and acetyl (Ac) protected glucose pendant moieties, respectively. Selective or dual deprotection of Boc- and Ac-groups from leucine and/or glucose moieties resulted in amphiphilic polymers, which self-assembled into nanoaggregates in aqueous medium, confirmed by critical aggregation concentration (CAC) determination, dynamic light scattering (DLS) and transmission electron microscopy (TEM). These glycopolymeric nanoassemblies were used to study the inhibition rates of insulin fibrillation and were found to impede the fibrillation of the insulin protein. Using several biophysical techniques, we observed that hydrophobic, electrostatic, and hydrogen bonding interactions were responsible for binding the insulin monomer/oligomer with various glycopolymeric aggregates, inhibiting insulin fibrillation. Tyrosine (Tyr) and Nile red (NR) fluorescence measurements manifested the hydrophobic interactions, whereas temperature-dependent fluorescence and isothermal titration calorimetry (ITC) measurements revealed respectively the hydrogen bonding and electrostatic interactions involved in the inhibition process of insulin amyloid formation. Molecular dynamics simulations further confirmed the involvement of different interactions among polymer-protein residues in averting the fibrillation process.


Subject(s)
Insulin , Polymers , Insulin/chemistry , Leucine , Polymers/chemistry , Molecular Dynamics Simulation
7.
Biochemistry ; 62(2): 270-280, 2023 01 17.
Article in English | MEDLINE | ID: mdl-35786907

ABSTRACT

Combinatorial readout of histone post-translational modifications by tandem reader modules mediates crosstalk among different histone modifications. To identify the domain-specific interactome of the tandem reader, we engineered the dual bromodomain of TATA-binding protein-associated factor-1 (TAF1) to carry a photoactivatable unnatural amino acid, 4-azido-l-phenylalanine (AzF), via amber suppressor mutagenesis. Using computational approaches, we modeled the targeted residues of TAF1 with AzF to predict the cross-linking distance between the reactive arylazide and its interacting partner. We developed three photoactivatable TAF1 tandem-bromodomain analogues, viz., Y1403AzF in bromodomain 1 (BD1), W1526AzF in bromodomain 2 (BD2), and Y1403AzF/W1526AzF in both BD1 and BD2. Circular dichroism and a thermal shift assay were used to confirm the structural integrity of the engineered readers. Using the TAF1 tandem-bromodomain analogues, we characterized their histone ligand binding properties by isothermal titration calorimetry and photo-cross-linking experiments. We found that the dual bromodomain of TAF1 independently binds and cross-links to different acetylated histone ligands. We further used the engineered BD1 and BD2 analogues of the TAF1 tandem readers to identify their domain-specific interacting partners at the cellular level. Both BD1 and BD2 independently cross-link to a unique interactome, and importantly, the dual cross-linker carrying TAF1 analogue could capture both BD1- and BD2-specific interactomes. Our work concludes that BD1 and BD2 of the TAF1 tandem reader independently recognize their interacting partners to regulate downstream cellular functions.


Subject(s)
Azides , Histones , Histones/metabolism , Photochemistry , Transcription Factors/chemistry , Protein Binding
8.
J Biomol Struct Dyn ; 41(20): 10569-10581, 2023 12.
Article in English | MEDLINE | ID: mdl-36524430

ABSTRACT

Bromodomains are evolutionarily conserved structural motifs that recognize acetylated lysine residues on histone tails. They play a crucial role in shaping chromatin architecture and regulating gene expression in various biological processes. Mutations in bromodomains containing proteins lead to multiple human diseases, which makes them attractive target for therapeutic intervention. Extensive studies have been done on BRD4 as a target for several cancers, such as Acute Myeloid Leukemia (AML) and Burkitt Lymphoma. Several potential inhibitors have been identified against the BRD4 bromodomain. However, most of these inhibitors have drawbacks such as non-specificity and toxicity, decreasing their appeal and necessitating the search for novel non-toxic inhibitors. This study aims to address this need by virtually screening natural compounds from the NPASS database against the Kac binding site of BRD4-BD1 using high throughput molecular docking followed by similarity clustering, pharmacokinetic screening, MD simulation and MM-PBSA binding free energy calculations. Using this approach, we identified five natural product inhibitors having a similar or better binding affinity to the BRD4 bromodomain compared to JQ1 (previously reported inhibitor of BRD4). Further systematic analysis of these inhibitors resulted in the top three hits: NPC268484 (Palodesangren-B), NPC295021 (Candidine) and NPC313112 (Buxifoliadine-D). Collectively, our in silico results identified some promising natural products that have the potential to act as potent BRD4-BD1 inhibitors and can be considered for further validation through future in vitro and in vivo studies.Communicated by Ramaswamy H. Sarma.


Subject(s)
Cell Cycle Proteins , Nuclear Proteins , Humans , Nuclear Proteins/chemistry , Molecular Docking Simulation , Cell Cycle Proteins/metabolism , Transcription Factors/chemistry , Computer Simulation
9.
Biochemistry ; 61(17): 1774-1789, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35976792

ABSTRACT

HBO1 [HAT bound to the origin recognition complex (ORC)], a member of the MYST family of histone acetyltransferases (HATs), was initially identified as a binding partner of ORC that acetylates free histone H3, H4, and nucleosomal H3. It functions as a quaternary complex with the BRPF (BRPF1/2/3) scaffolding protein and two accessory proteins, ING4/5 and Eaf6. Interaction of BRPF2 with HBO1 has been shown to be important for regulating H3K14 acetylation during embryonic development. However, how BRPF2 directs the HBO1 HAT complex to chromatin to regulate its HAT activity toward nucleosomal substrates remains unclear. Our findings reveal novel interacting partners of the BRPF2 bromodomain that recognizes different acetyllysine residues on the N-terminus of histone H4, H3, and H2A and preferentially binds to H4K5ac, H4K8ac, and H4K5acK12ac modifications. In addition, mutational analysis of the BRPF2 bromodomain coupled with isothermal titration calorimetry binding and pull-down assays on the histone substrates identified critical residues responsible for acetyllysine binding. Moreover, the BRPF2 bromodomain could enrich H4K5ac mark-bearing mononucleosomes compared to other acetylated H4 marks. Consistent with this, ChIP-seq analysis revealed that BRPF2 strongly co-localizes with HBO1 at histone H4K5ac and H4K8ac marks near the transcription start sites in the genome. Our study provides novel insights into how the histone binding function of the BRPF2 bromodomain directs the recruitment of the HBO1 HAT complex to chromatin to regulate gene expression.


Subject(s)
Histone Code , Histones , Acetylation , Chromatin , Histone Acetyltransferases/metabolism , Histones/metabolism , Nucleosomes , Protein Processing, Post-Translational
10.
Chem Commun (Camb) ; 57(77): 9866-9869, 2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34490864

ABSTRACT

The site-specific installation of light-activable crosslinker unnatural amino acids offers a powerful approach to trap transient protein-protein interactions both in vitro and in vivo. Herein, we engineer a bromodomain to introduce 4-benzoyl-L-phenylalanine (BzF) using amber suppressor mutagenesis without compromising its ability to recognize the acetylated histone proteins. We demonstrate the high crosslinking efficiency of the engineered reader towards the interacting partners and its suitability for profiling the transient bromodomain interactome.


Subject(s)
Amino Acids/chemistry , Cross-Linking Reagents/chemistry , Lysine/chemistry , Protein Engineering , Lysine/analogs & derivatives , Photochemical Processes
11.
Chem Asian J ; 16(21): 3404-3412, 2021 Nov 02.
Article in English | MEDLINE | ID: mdl-34448544

ABSTRACT

Bromodomains are evolutionarily conserved reader modules that recognize acetylated lysine residues on the histone tails to facilitate gene transcription. The bromodomain and PHD finger containing protein 3 (BRPF3) is a scaffolding protein that forms a tetrameric complex with HBO1 histone acetyltransferase (HAT) and two other subunits, which is known to regulate the HAT activity and substrate specificity. However, its molecular mechanism, histone ligands, and biological functions remain unknown. Herein, we identify mono- (H4K5ac) and di- (H4K5acK12ac) acetylated histone peptides as novel interacting partners of the BRPF3 bromodomain. Consistent with this, pull-down assays on purified histones from human cells confirm the interaction of BRPF3 bromodomain with acetylated histone H4. Further, MD simulation studies highlight the binding mode of acetyllysine (Kac) and the stability of bromodomain-histone peptide complexes. Collectively, our findings provide a key insight into how histone targets of the BRPF3 bromodomain direct the recruitment of HBO1 complex to chromatin for downstream transcriptional regulation.


Subject(s)
Histone Acetyltransferases/metabolism , Histones/metabolism , Acetylation , Histone Acetyltransferases/chemistry , Histones/analysis , Humans , Molecular Dynamics Simulation
SELECTION OF CITATIONS
SEARCH DETAIL
...