Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Proc Natl Acad Sci U S A ; 111(48): 17110-5, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25406323

ABSTRACT

Proteins endocytosed from serum are degraded in the lysosomes. However, serum albumin (SA) and IgG, through its Fc part, bind to the neonatal Fc receptor (FcRn) at low pH in the endosome after endocytosis, and are transported back to the cellular surface, where they are released into the bloodstream, resulting in an extended serum circulation time. Association with Fc or SA has been used to prolong the in vivo half-life of biopharmaceuticals, using the interaction with FcRn to improve treatment regimens. This has been achieved either directly, by fusion or conjugation to Fc or SA, or indirectly, using SA-binding proteins. The present work takes this principle one step further, presenting small affinity proteins that bind directly to FcRn, mediating extension of the serum half-life of fused biomolecules. Phage display technology was used to select affibody molecules that can bind to FcRn in the pH-dependent manner required for rescue by FcRn. The biophysical and binding properties were characterized in vitro, and the affibody molecules were found to bind to FcRn more strongly at low pH than at neutral pH. Attachment of the affibody molecules to a recombinant protein, already engineered for increased half-life, resulted in a nearly threefold longer half-life in mice. These tags should have general use as fusion partners to biopharmaceuticals to extend their half-lives in vivo.


Subject(s)
Carrier Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Receptors, Fc/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Animals , Binding, Competitive , Carrier Proteins/genetics , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Half-Life , HeLa Cells , Histocompatibility Antigens Class I/genetics , Humans , Hydrogen-Ion Concentration , Male , Mice, Inbred Strains , Peptide Library , Protein Binding , Receptors, Fc/genetics , Recombinant Fusion Proteins/blood
2.
J Biol Chem ; 286(7): 5234-41, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21138843

ABSTRACT

The therapeutic and diagnostic efficiency of engineered small proteins, peptides, and chemical drug candidates is hampered by short in vivo serum half-life. Thus, strategies to tailor their biodistribution and serum persistence are highly needed. An attractive approach is to take advantage of the exceptionally long circulation half-life of serum albumin or IgG, which is attributed to a pH-dependent interaction with the neonatal Fc receptor (FcRn) rescuing these proteins from intracellular degradation. Here, we present molecular evidence that a minimal albumin binding domain (ABD) derived from streptococcal protein G can be used for efficient half-life extension by indirect targeting of FcRn. We show that ABD, and ABD recombinantly fused to an Affibody molecule, in complex with albumin does not interfere with the strictly pH-dependent FcRn-albumin binding kinetics. The same result was obtained in the presence of IgG. An in vivo study performed in rat confirmed that the clinically relevant human epidermal growth factor 2 (HER2)-targeting Affibody molecule fused to ABD has a similar half-life and biodistribution profile as serum albumin. The proof-of-concept described may be broadly applicable to extend the in vivo half-life of short lived biological or chemical drugs ultimately resulting in enhanced therapeutic or diagnostic efficiency, a more favorable dosing regimen, and improved patient compliance.


Subject(s)
Bacterial Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Receptors, Fc/metabolism , Recombinant Fusion Proteins/metabolism , Serum Albumin/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/pharmacology , Half-Life , Histocompatibility Antigens Class I/genetics , Humans , Immunoglobulin G/genetics , Immunoglobulin G/pharmacology , Protein Binding , Rats , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Fc/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Schistosoma japonicum , Serum Albumin/genetics , Serum Albumin/pharmacology
3.
Chembiochem ; 13(7): 1024-31, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22532524

ABSTRACT

Affinity molecules labeled with different reporter groups, such as fluorophores or radionuclides, are valuable research tools used in a variety of applications. One class of engineered affinity proteins is Affibody molecules, which are small (6.5 kDa) proteins that can be produced by solid phase peptide synthesis (SPPS), thereby allowing site-specific incorporation of reporter groups during synthesis. The Affibody molecules are triple-helix proteins composed of a variable part, which gives the protein its binding specificity, and a constant part, which is identical for all Affibody molecules. In the present study, native chemical ligation (NCL) has been applied for combinatorial assembly of Affibody molecules from peptide fragments produced by Fmoc SPPS. The concept is demonstrated for the synthesis of three different Affibody molecules. The cysteine residue introduced at the site of ligation can be used for directed immobilization and does not interfere with the function of the investigated proteins. This strategy combines a high-yield production method with facilitated preparation of proteins with different C-terminal modifications.


Subject(s)
Antibodies/chemistry , Biomimetic Materials/chemistry , Peptide Fragments/chemistry , Amino Acid Sequence , Antibodies/metabolism , Biomimetic Materials/chemical synthesis , Biomimetic Materials/metabolism , Combinatorial Chemistry Techniques , Humans , Immobilized Proteins/chemical synthesis , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Ligands , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Protein Engineering
4.
Pharmaceutics ; 14(11)2022 Nov 19.
Article in English | MEDLINE | ID: mdl-36432709

ABSTRACT

Targeted molecular radiation therapy is a promising emerging treatment modality in oncology, and peptide synthesis may shorten the time to reach the clinical stage. In this study, we have explored Chemo-Enzymatic Peptide Synthesis, or CEPS, as a new means of producing a therapeutic HER2 targeted Affibody® molecule, comprising a C-terminal albumin binding domain (ABD) for half-life extension and a total length of 108 amino acids. In addition, a DOTA moiety could be incorporated at N-terminus directly during the synthesis step and subsequently utilized for site-specific radiolabeling with the therapeutic radionuclide 177Lu. Retained thermodynamic stability as well as retained binding to both HER2 and albumin was verified. Furthermore, HER2 binding specificity of the radiolabeled Affibody molecule was confirmed by an in vitro saturation assay showing a significantly higher cell-bound activity of SKOV-3 (high HER2 expression) compared with BxPC3 (low HER2 expression), both in the presence and absence of HSA. In vivo evaluation in mice bearing HER2 expressing xenografts also showed specific tumor targeting as well as extended time in circulation and reduced kidney uptake compared with a HER2 targeted Affibody molecule without the ABD moiety. To conclude, we have demonstrated that CEPS can be used for production of Affibody-fusion molecules with retained in vitro and in vivo functionality.

5.
Mol Imaging Biol ; 23(2): 241-249, 2021 04.
Article in English | MEDLINE | ID: mdl-33098025

ABSTRACT

PURPOSE: In vivo imaging of programmed death ligand 1 (PD-L1) during immunotherapy could potentially monitor changing PD-L1 expression and PD-L1 expression heterogeneity within and across tumors. Some protein constructs can be used for same-day positron emission tomography (PET) imaging. Previously, we evaluated the PD-L1-targeting Affibody molecule [18F]AlF-NOTA-ZPD-L1_1 as a PET tracer in a mouse tumor model of human PD-L1 expression. In this study, we evaluated the affinity-matured Affibody molecule ZPD-L1_4, to determine if improved affinity for PD-L1 resulted in increased in vivo targeting of PD-L1. PROCEDURES: ZPD-L1_4 was conjugated with NOTA and radiolabeled with either [18F]AlF or 68Ga. [18F]AlF-NOTA-ZPD-L1_4 and [68Ga]NOTA-ZPD-L1_4 were evaluated in immunocompromised mice with LOX (PD-L1+) and SUDHL6 (PD-L1-) tumors with PET and ex vivo biodistribution measurements. In addition, whole-body PET studies were performed in rhesus monkeys to predict human biodistribution in a model with tracer binding to endogenous PD-L1, and to calculate absorbed radiation doses. RESULTS: Ex vivo biodistribution measurements showed that both tracers had > 25 fold higher accumulation in LOX tumors than SUDHL6 ([18F]AlF-NOTA-ZPD-L1_4: LOX: 8.7 ± 0.7 %ID/g (N = 4) SUDHL6: 0.2 ± 0.01 %ID/g (N = 6), [68Ga]NOTA-ZPD-L1_4: LOX: 15.8 ± 1.0 %ID/g (N = 6) SUDHL6: 0.6 ± 0.1 %ID/g (N = 6)), considerably higher than ZPD-L1_1. In rhesus monkeys, both PET tracers showed fast clearance through kidneys and low background signal in the liver ([18F]AlF-NOTA-ZPD-L1_4: 1.26 ± 0.13 SUV, [68Ga]NOTA-ZPD-L1_4: 1.11 ± 0.06 SUV). PD-L1-expressing lymph nodes were visible in PET images, indicating in vivo PD-L1 targeting. Dosimetry estimates suggest that both PET tracers can be used for repeated clinical studies, although high kidney accumulation may limit allowable radioactive doses. CONCLUSIONS: [18F]AlF-NOTA-ZPD-L1_4 and [68Ga]NOTA-ZPD-L1_4 are promising candidates for same-day clinical PD-L1 PET imaging, warranting clinical evaluation. The ability to use either [18F] or [68Ga] may expand access to clinical sites.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , B7-H1 Antigen/metabolism , Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Radiometry/methods , Radiopharmaceuticals/pharmacokinetics , Animals , Antibodies, Monoclonal/administration & dosage , B7-H1 Antigen/immunology , Cell Line, Tumor , Fluorine Radioisotopes , Gallium Radioisotopes , Humans , Immunotherapy/methods , Macaca mulatta , Mice , Molecular Imaging/methods , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Radiopharmaceuticals/administration & dosage , Tissue Distribution , Xenograft Model Antitumor Assays
6.
J Nucl Med ; 58(11): 1852-1857, 2017 11.
Article in English | MEDLINE | ID: mdl-28588151

ABSTRACT

Programmed death ligand 1 (PD-L1) is an immune regulatory ligand that binds to the T-cell immune check point programmed death 1. Tumor expression of PD-L1 is correlated with immune suppression and poor prognosis. It is also correlated with therapeutic efficacy of programmed death 1 and PD-L1 inhibitors. In vivo imaging may enable real-time follow-up of changing PD-L1 expression and heterogeneity evaluation of PD-L1 expression across tumors in the same subject. We have radiolabeled the PD-L1-binding Affibody molecule NOTA-ZPD-L1_1 with 18F and evaluated its in vitro and in vivo binding affinity, targeting, and specificity. Methods: The affinity of the PD-L1-binding Affibody ligand ZPD-L1_1 was evaluated by surface plasmon resonance. Labeling was accomplished by maleimide coupling of NOTA to a unique cysteine residue and chelation of 18F-AlF. In vivo studies were performed in PD-L1-positive, PD-L1-negative, and mixed tumor-bearing severe combined immunodeficiency mice. Tracer was injected via the tail vein, and dynamic PET scans were acquired for 90 min, followed by γ-counting biodistribution. Immunohistochemical staining with an antibody specific for anti-PD-L1 (22C3) was used to evaluate the tumor distribution of PD-L1. Immunohistochemistry results were then compared with ex vivo autoradiographic images obtained from adjacent tissue sections. Results: NOTA-ZPD-L1_1 was labeled, with a radiochemical yield of 15.1% ± 5.6%, radiochemical purity of 96.7% ± 2.0%, and specific activity of 14.6 ± 6.5 GBq/µmol. Surface plasmon resonance showed a NOTA-conjugated ligand binding affinity of 1 nM. PET imaging demonstrated rapid uptake of tracer in the PD-L1-positive tumor, whereas the PD-L1-negative control tumor showed little tracer retention. Tracer clearance from most organs and blood was quick, with biodistribution showing prominent kidney retention, low liver uptake, and a significant difference between PD-L1-positive (percentage injected dose per gram [%ID/g] = 2.56 ± 0.33) and -negative (%ID/g = 0.32 ± 0.05) tumors (P = 0.0006). Ex vivo autoradiography showed excellent spatial correlation with immunohistochemistry in mixed tumors. Conclusion: Our results show that Affibody ligands can be effective at targeting tumor PD-L1 in vivo, with good specificity and rapid clearance. Future studies will explore methods to reduce kidney activity retention and further increase tumor uptake.


Subject(s)
B7-H1 Antigen/metabolism , Fluorine Radioisotopes , Positron-Emission Tomography/methods , Radiopharmaceuticals , Affinity Labels , Animals , Antibodies, Monoclonal , Autoradiography , Female , Fluorine Radioisotopes/pharmacokinetics , Humans , Immunohistochemistry , Isotope Labeling/methods , Male , Mice, SCID , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Organometallic Compounds , Radiopharmaceuticals/pharmacokinetics , Surface Plasmon Resonance , Tissue Distribution
7.
J Mol Biol ; 350(5): 964-73, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-15978617

ABSTRACT

EMSY is a recently discovered gene encoding a BRCA2-associated protein and is amplified in some sporadic breast and ovarian cancers. The EMSY sequence contains no known domain except for a conserved approximately 100 residue segment at the N terminus. This so-called ENT domain is unique in the human genome, although multiple copies are found in Arabidopsis proteins containing members of the Royal family of chromatin remodelling domains. Here, we report the crystal structure of the ENT domain of EMSY, consisting of a unique arrangement of five alpha-helices that fold into a helical bundle arrangement. The fold shares regions of structural homology with the DNA-binding domain of homeodomain proteins. The ENT domain forms a homodimer via the anti-parallel packing of the extended N-terminal alpha-helix of each molecule. It is stabilized mainly by hydrophobic residues at the dimer interface and has a dissociation constant in the low micromolar range. The dimerisation of EMSY mediated by the ENT domain could provide flexibility for it to bind two or more different substrates simultaneously.


Subject(s)
Crystallography, X-Ray , Repressor Proteins/chemistry , Binding Sites , Dimerization , Humans , Hydrophobic and Hydrophilic Interactions , Neoplasm Proteins , Nuclear Proteins , Protein Structure, Tertiary , Structural Homology, Protein
8.
Biomed Res Int ; 2015: 605427, 2015.
Article in English | MEDLINE | ID: mdl-26075248

ABSTRACT

In drug development, the "onus" of the low R&D efficiency has been put traditionally onto the drug discovery process (i.e., finding the right target or "binding" functionality). Here, we show that manufacturing is not only a central component of product success, but also that, by integrating manufacturing and discovery activities in a "holistic" interpretation of QbD methodologies, we could expect to increase the efficiency of the drug discovery process as a whole. In this new context, early risk assessment, using developability methodologies and computational methods in particular, can assist in reducing risks during development in a cost-effective way. We define specific areas of risk and how they can impact product quality in a broad sense, including essential aspects such as product efficacy and patient safety. Emerging industry practices around developability are introduced, including some specific examples of applications to biotherapeutics. Furthermore, we suggest some potential workflows to illustrate how developability strategies can be introduced in practical terms during early drug development in order to mitigate risks, reduce drug attrition and ultimately increase the robustness of the biopharmaceutical supply chain. Finally, we also discuss how the implementation of such methodologies could accelerate the access of new therapeutic treatments to patients in the clinic.


Subject(s)
Biopharmaceutics/methods , Drug Design , Drug Industry/methods , Humans
9.
Protein Sci ; 13(3): 617-25, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14978302

ABSTRACT

53BP1 interacts with the DNA-binding core domain of the tumor suppressor p53 and enhances p53-mediated transcriptional activation. The p53-binding region of 53BP1 maps to the C-terminal BRCT domains, which are homologous to those found in the breast cancer protein BRCA1 and in other proteins involved in DNA repair. Here we compare the thermodynamic behavior of the BRCT domains of 53BP1 and BRCA1 and examine their ability to interact with the p53 core domain. The free energies of unfolding are of similar magnitude, although slightly higher for 53BP1-BRCT, and both populate an aggregation-prone partly folded intermediate. Interaction studies performed in vitro by analytical size-exclusion chromatography, analytical ultracentrifugation, and isothermal titration calorimetry reveal that 53BP1-BRCT interacts with p53 with a K(d) in the low micromolar range. Despite their homology with 53BP1-BRCT domains, the BRCT domains of BRCA1 did not bind p53 with any detectable affinity. In summary, although other studies have indicated that the BRCT domains of both BRCA1 and 53BP1 interact with p53 core domain, the quantitative biophysical measurements performed here indicate that only 53BP1 can bind. Although both proteins may be involved in the same DNA repair pathways, our study indicates that a direct role in p53 function is unique to 53BP1.


Subject(s)
BRCA1 Protein/chemistry , Intracellular Signaling Peptides and Proteins , Amino Acid Sequence , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Binding Sites/genetics , Calorimetry , Carrier Proteins , Chromatography, Gel , Cysteine Endopeptidases/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphoproteins , Protein Binding , Protein Denaturation , Protein Folding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thermodynamics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor p53-Binding Protein 1 , Ultracentrifugation
10.
J Nucl Med ; 54(6): 961-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23528382

ABSTRACT

UNLABELLED: Because of their better penetration, smaller targeting proteins may be superior to antibodies for radioimmunotherapy of solid tumors. Therefore, Affibody molecules (6.5 kDa) have a potential for being suitable as targeted moiety for radiolabeled therapeutic proteins. Previous studies have demonstrated that a fusion of an Affibody molecule with an albumin-binding domain (ABD) provides a strong noncovalent binding to albumin in vivo. This strong noncovalent binding can be used for reduction of the renal uptake of the Affibody molecule while maintaining a size smaller than that of an antibody, which is important when using residualizing radionuclide labels conjugated to Affibody molecules. The goal of this study was to design and evaluate a new targeting Affibody-ABD fusion protein with improved biodistribution properties for radionuclide therapy. METHODS: A novel Affibody-based construct, ZHER2:2891-ABD035-DOTA (ABY-027), was created by fusion of the reengineered HER2-binding Affibody molecule ZHER2:2891 to the N terminus of the high-affinity ABD035, and a maleimido-derivative of DOTA was conjugated at the C terminus of the construct. Binding and processing of (177)Lu-ABY-027 by HER2-expressing cells were evaluated in vitro. Targeting of HER2-expressing SKOV-3 xenografts was evaluated in BALB/C nu/nu mice and compared with targeting of previously reported ABD-(ZHER2:342)2. RESULTS: The binding affinity (dissociation constant) of ABY-027 to HER2 (74 pM) was the same as for the parental ZHER2:2891 (76 pM). ABY-027 was stably labeled with (177)Lu and (111)In with preserved specific binding to HER2-expressing cells in vitro. In vivo receptor saturation experiments demonstrated that targeting of SKOV-3 xenografts in BALB/C nu/nu mice was HER2-specific. (177)Lu-ABY-027 demonstrated substantially (2- to 3-fold) lower renal and hepatic uptake than previously assessed HER2-specific Affibody-based albumin-binding agents. Tumor uptake of radiolabeled ABY-027 at 48 h after injection was 2-fold higher than that for previously reported ABD-(ZHER2:342)2. CONCLUSION: An optimized molecular design of an ABD fusion protein resulted in an Affibody molecule construct with better properties for therapy. Fully preserved in vivo targeting of the fusion protein was shown in xenografted mice. Site-specific coupling of DOTA provides a uniform conjugate and creates the potential for labeling with a broad range of therapeutic radionuclides. The biodistribution of (177)Lu-ABY-027 in a murine model suggests it is more suitable for therapy than alternative approaches.


Subject(s)
Albumins/metabolism , Lutetium/therapeutic use , Radioisotopes/therapeutic use , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Animals , Binding Sites , Cell Line, Tumor , Female , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Isotope Labeling , Mice , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/radiotherapy , Protein Structure, Tertiary , Protein Transport , Radiometry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Substrate Specificity , Tissue Distribution
11.
Protein Sci ; 19(12): 2319-29, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20886513

ABSTRACT

The aggregation of amyloid-ß (Aß) peptides is believed to be a major factor in the onset and progression of Alzheimer's disease. Molecules binding with high affinity and selectivity to Aß-peptides are important tools for investigating the aggregation process. An Aß-binding Affibody molecule, ZAß3 , has earlier been selected by phage display and shown to bind Aß(1-40) with nanomolar affinity and to inhibit Aß-peptide aggregation. In this study, we create truncated functional versions of the ZAß3 Affibody molecule better suited for chemical synthesis production. Engineered Affibody molecules of different length were produced by solid phase peptide synthesis and allowed to form covalently linked homodimers by S-S-bridges. The N-terminally truncated Affibody molecules ZAß3 (12-58), ZAß3 (15-58), and ZAß3 (18-58) were produced in considerably higher synthetic yield than the corresponding full-length molecule ZAß3 (1-58). Circular dichroism spectroscopy and surface plasmon resonance-based biosensor analysis showed that the shortest Affibody molecule, ZAß3 (18-58), exhibited complete loss of binding to the Aß(1-40)-peptide, while the ZAß3 (12-58) and ZAß3 (15-58) Affibody molecules both displayed approximately one order of magnitude higher binding affinity to the Aß(1-40)-peptide compared to the full-length Affibody molecule. Nuclear magnetic resonance spectroscopy showed that the structure of Aß(1-40) in complex with the truncated Affibody dimers is very similar to the previously published solution structure of the Aß(1-40)-peptide in complex with the full-length ZAß3 Affibody molecule. This indicates that the N-terminally truncated Affibody molecules ZAß3 (12-58) and ZAß3 (15-58) are highly promising for further engineering and future use as binding agents to monomeric Aß(1-40).


Subject(s)
Amyloid beta-Peptides/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Amyloid beta-Peptides/chemistry , Circular Dichroism , Magnetic Resonance Spectroscopy , Peptides/chemistry , Protein Binding , Protein Engineering
12.
EMBO Rep ; 6(7): 675-80, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15947784

ABSTRACT

EMSY is a large nuclear protein that binds to the transactivation domain of BRCA2. EMSY contains an approximately 100-residue segment at the amino terminus called the ENT (EMSY N-terminal) domain. Plant proteins containing ENT domains also contain members of the royal family of chromatin-remodelling domains. It has been proposed that EMSY may have a role in chromatin-related processes. This is supported by the observation that a number of chromatin-regulator proteins, including HP1beta and BS69, bind directly to EMSY by means of a conserved motif adjacent to the ENT domain. Here, we report the crystal structure of residues 1-108 of EMSY at 2.0 A resolution. The structure contains both the ENT domain and the HP1beta/BS69-binding motif. This binding motif forms an extended peptide-like conformation that adopts distinct orientations in each subunit of the dimer. Biophysical and nuclear magnetic resonance analyses show that the main complex formed by EMSY and the chromoshadow domain of HP1 (HP1-CSD) consists of one EMSY dimer sandwiched between two HP1-CSD dimers. The HP1beta-binding motif is necessary and sufficient for EMSY to bind to the chromoshadow domain of HP1beta.


Subject(s)
Carrier Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Repressor Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites/genetics , Carrier Proteins/chemistry , Cell Cycle Proteins , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/chemistry , Co-Repressor Proteins , Crystallography , DNA-Binding Proteins , Dimerization , Humans , Mice , Molecular Sequence Data , Neoplasm Proteins , Nuclear Proteins , Protein Structure, Tertiary , Repressor Proteins/genetics , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL