Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Commun (Camb) ; 59(56): 8696-8699, 2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37347155

ABSTRACT

In the search for foldamer inhibitors of the histone chaperone ASF1, we explored the possibility of substituting four α-residues (≈one helix turn) by 3-urea segments and scanned the sequence of a short α-helical peptide known to bind ASF1. By analysing the impact of the different foldamer replacements within the peptide chain, we uncovered new binding modes of the peptide-urea chimeras to ASF1.


Subject(s)
Histone Chaperones , Histones , Histone Chaperones/metabolism , Histones/chemistry , Molecular Chaperones/chemistry , Cell Cycle Proteins/metabolism , Peptides/pharmacology , Peptides/metabolism
2.
Sci Adv ; 7(12)2021 03.
Article in English | MEDLINE | ID: mdl-33741589

ABSTRACT

Sequence-specific oligomers with predictable folding patterns, i.e., foldamers, provide new opportunities to mimic α-helical peptides and design inhibitors of protein-protein interactions. One major hurdle of this strategy is to retain the correct orientation of key side chains involved in protein surface recognition. Here, we show that the structural plasticity of a foldamer backbone may notably contribute to the required spatial adjustment for optimal interaction with the protein surface. By using oligoureas as α helix mimics, we designed a foldamer/peptide hybrid inhibitor of histone chaperone ASF1, a key regulator of chromatin dynamics. The crystal structure of its complex with ASF1 reveals a notable plasticity of the urea backbone, which adapts to the ASF1 surface to maintain the same binding interface. One additional benefit of generating ASF1 ligands with nonpeptide oligourea segments is the resistance to proteolysis in human plasma, which was highly improved compared to the cognate α-helical peptide.


Subject(s)
Histone Chaperones , Peptides , Humans , Peptides/chemistry , Protein Conformation, alpha-Helical , Urea/chemistry
3.
Liver Int ; 41(1): 20-32, 2021 01.
Article in English | MEDLINE | ID: mdl-33190346

ABSTRACT

The recent outbreak of coronavirus disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has resulted in a world-wide pandemic. Disseminated lung injury with the development of acute respiratory distress syndrome (ARDS) is the main cause of mortality in COVID-19. Although liver failure does not seem to occur in the absence of pre-existing liver disease, hepatic involvement in COVID-19 may correlate with overall disease severity and serve as a prognostic factor for the development of ARDS. The spectrum of liver injury in COVID-19 may range from direct infection by SARS-CoV-2, indirect involvement by systemic inflammation, hypoxic changes, iatrogenic causes such as drugs and ventilation to exacerbation of underlying liver disease. This concise review discusses the potential pathophysiological mechanisms for SARS-CoV-2 hepatic tropism as well as acute and possibly long-term liver injury in COVID-19.


Subject(s)
COVID-19/complications , Liver Diseases/etiology , Liver/virology , SARS-CoV-2 , Viral Tropism , Angiotensin-Converting Enzyme 2/physiology , Cholestasis/etiology , Humans , Non-alcoholic Fatty Liver Disease/etiology
4.
Cell Chem Biol ; 26(11): 1573-1585.e10, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31543461

ABSTRACT

Anti-silencing function 1 (ASF1) is a conserved H3-H4 histone chaperone involved in histone dynamics during replication, transcription, and DNA repair. Overexpressed in proliferating tissues including many tumors, ASF1 has emerged as a promising therapeutic target. Here, we combine structural, computational, and biochemical approaches to design peptides that inhibit the ASF1-histone interaction. Starting from the structure of the human ASF1-histone complex, we developed a rational design strategy combining epitope tethering and optimization of interface contacts to identify a potent peptide inhibitor with a dissociation constant of 3 nM. When introduced into cultured cells, the inhibitors impair cell proliferation, perturb cell-cycle progression, and reduce cell migration and invasion in a manner commensurate with their affinity for ASF1. Finally, we find that direct injection of the most potent ASF1 peptide inhibitor in mouse allografts reduces tumor growth. Our results open new avenues to use ASF1 inhibitors as promising leads for cancer therapy.


Subject(s)
Cell Cycle Proteins/metabolism , Drug Design , Molecular Chaperones/metabolism , Peptides/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epitopes/chemistry , Epitopes/metabolism , Female , Histones/chemistry , Histones/metabolism , Humans , Kinetics , Mice , Mice, Inbred BALB C , Molecular Chaperones/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , Peptides/metabolism , Peptides/pharmacology , Peptides/therapeutic use , Thermodynamics , Transplantation, Homologous
5.
Chembiochem ; 20(7): 891-895, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30512234

ABSTRACT

Inhibiting the histone H3-ASF1 (anti-silencing function 1) protein-protein interaction (PPI) represents a potential approach for treating numerous cancers. As an α-helix-mediated PPI, constraining the key histone H3 helix (residues 118-135) is a strategy through which chemical probes might be elaborated to test this hypothesis. In this work, variant H3118-135 peptides bearing pentenylglycine residues at the i and i+4 positions were constrained by olefin metathesis. Biophysical analyses revealed that promotion of a bioactive helical conformation depends on the position at which the constraint is introduced, but that the potency of binding towards ASF1 is unaffected by the constraint and instead that enthalpy-entropy compensation occurs.


Subject(s)
Alkenes/chemistry , Cell Cycle Proteins/metabolism , Histones/metabolism , Molecular Chaperones/metabolism , Peptide Fragments/metabolism , Amino Acid Sequence , Histones/chemistry , Humans , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Thermodynamics
6.
Nucleic Acids Res ; 43(3): 1905-17, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25618846

ABSTRACT

MCM2 is a subunit of the replicative helicase machinery shown to interact with histones H3 and H4 during the replication process through its N-terminal domain. During replication, this interaction has been proposed to assist disassembly and assembly of nucleosomes on DNA. However, how this interaction participates in crosstalk with histone chaperones at the replication fork remains to be elucidated. Here, we solved the crystal structure of the ternary complex between the histone-binding domain of Mcm2 and the histones H3-H4 at 2.9 Å resolution. Histones H3 and H4 assemble as a tetramer in the crystal structure, but MCM2 interacts only with a single molecule of H3-H4. The latter interaction exploits binding surfaces that contact either DNA or H2B when H3-H4 dimers are incorporated in the nucleosome core particle. Upon binding of the ternary complex with the histone chaperone ASF1, the histone tetramer dissociates and both MCM2 and ASF1 interact simultaneously with the histones forming a 1:1:1:1 heteromeric complex. Thermodynamic analysis of the quaternary complex together with structural modeling support that ASF1 and MCM2 could form a chaperoning module for histones H3 and H4 protecting them from promiscuous interactions. This suggests an additional function for MCM2 outside its helicase function as a proper histone chaperone connected to the replication pathway.


Subject(s)
Cell Cycle Proteins/metabolism , Histones/metabolism , Minichromosome Maintenance Complex Component 2/metabolism , Molecular Chaperones/metabolism , Animals , Calorimetry , Chromatography, Gel , Drosophila melanogaster , Humans , Magnetic Resonance Spectroscopy , Minichromosome Maintenance Complex Component 2/chemistry , Protein Binding , Protein Conformation , Thermodynamics , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL