Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Death Discov ; 9(1): 34, 2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36707507

ABSTRACT

m6A is an important RNA methylation in progression of various human cancers. As the m6A reader protein, YTHDF1 is reported to accelerate m6A-modified mRNAs translation in cytoplasm. It is highly expressed in various human cancers and contributes to the progression and metastasis of cancers. YTHDF1 was closely associated with poor prognosis and also used as a molecular marker for clinical diagnosis or therapy in human cancers. It has been reported to promote chemoresistance to Adriamycin, Cisplatin and Olaparib by increasing mRNA stability of its target molecule. Moreover, it contributes to CSC-like characteristic of tumor cells and inducing the antitumor immune microenvironment. Here, we reviewed the clinical diagnostic and prognostic values of YTHDF1, as well as the molecular mechanisms of YTHDF1 in progression and metastasis of human cancers.

2.
Commun Biol ; 4(1): 526, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33953337

ABSTRACT

The heterodimer of ATP-binding cassette transporter ABCG5 and ABCG8 mediates the excretion of sterols from liver and intestine, playing a critical role in cholesterol homeostasis. Here, we present the cryo-EM structure of ABCG5/G8 in complex with the Fab fragments from two monoclonal antibodies at 3.3Å resolution. The high-resolution structure reveals a unique dimer interface between the nucleotide-binding domains (NBD) of opposing transporters, consisting of an ordered network of salt bridges between the conserved NPXDFXXD motif and serving as a pivot point that may be important for the transport cycle. While mAb 11F4 increases the ATPase activity potentially by stabilization of the NBD dimer formation, mAb 2E10 inhibits ATP hydrolysis, likely by restricting the relative movement between the RecA and helical domain of ABCG8 NBD. Our study not only provides insights into the structural elements important for the transport cycle but also reveals novel epitopes for potential therapeutic interventions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 5/chemistry , ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 8/chemistry , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Cryoelectron Microscopy/methods , Lipoproteins/chemistry , Lipoproteins/metabolism , Adenosine Triphosphate/metabolism , Humans , Models, Molecular , Protein Conformation
3.
Front Immunol ; 12: 660198, 2021.
Article in English | MEDLINE | ID: mdl-33968063

ABSTRACT

The worldwide pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is unprecedented and the impact on public health and the global economy continues to be devastating. Although early therapies such as prophylactic antibodies and vaccines show great promise, there are concerns about the long-term efficacy and universal applicability of these therapies as the virus continues to mutate. Thus, protein-based immunogens that can quickly respond to viral changes remain of continued interest. The Spike protein, the main immunogen of this virus, displays a highly dynamic trimeric structure that presents a challenge for therapeutic development. Here, guided by the structure of the Spike trimer, we rationally design new Spike constructs that show a uniquely high stability profile while simultaneously remaining locked into the immunogen-desirable prefusion state. Furthermore, our approach emphasizes the relationship between the highly conserved S2 region and structurally dynamic Receptor Binding Domains (RBD) to enable vaccine development as well as the generation of antibodies able to resist viral mutation.


Subject(s)
Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Angiotensin-Converting Enzyme 2/metabolism , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Binding Sites/genetics , Binding Sites/immunology , COVID-19/immunology , COVID-19/pathology , Cell Line , HEK293 Cells , Humans , Protein Domains/genetics , Protein Domains/immunology , Protein Stability , SARS-CoV-2/genetics
4.
Sci Adv ; 6(25): eabb1989, 2020 06.
Article in English | MEDLINE | ID: mdl-32596471

ABSTRACT

The intestinal absorption of cholesterol is mediated by a multipass membrane protein, Niemann-Pick C1-Like 1 (NPC1L1), the molecular target of a cholesterol lowering therapy ezetimibe. While ezetimibe gained Food and Drug Administration approval in 2002, its mechanism of action has remained unclear. Here, we present two cryo-electron microscopy structures of NPC1L1, one in its apo form and the other complexed with ezetimibe. The apo form represents an open state in which the N-terminal domain (NTD) interacts loosely with the rest of NPC1L1, leaving the NTD central cavity accessible for cholesterol loading. The ezetimibe-bound form signifies a closed state in which the NTD rotates ~60°, creating a continuous tunnel enabling cholesterol movement into the plasma membrane. Ezetimibe blocks cholesterol transport by occluding the tunnel instead of competing with cholesterol binding. These findings provide insight into the molecular mechanisms of NPC1L1-mediated cholesterol transport and ezetimibe inhibition, paving the way for more effective therapeutic development.

5.
Elife ; 92020 03 09.
Article in English | MEDLINE | ID: mdl-32149605

ABSTRACT

Transient receptor potential canonical (TRPC) proteins form nonselective cation channels that play physiological roles in a wide variety of cells. Despite growing evidence supporting the therapeutic potential of TRPC6 inhibition in treating pathological cardiac and renal conditions, mechanistic understanding of TRPC6 function and modulation remains obscure. Here we report cryo-EM structures of TRPC6 in both antagonist-bound and agonist-bound states. The structures reveal two novel recognition sites for the small-molecule modulators corroborated by mutagenesis data. The antagonist binds to a cytoplasm-facing pocket formed by S1-S4 and the TRP helix, whereas the agonist wedges at the subunit interface between S6 and the pore helix. Conformational changes upon ligand binding illuminate a mechanistic rationale for understanding TRPC6 modulation. Furthermore, structural and mutagenesis analyses suggest several disease-related mutations enhance channel activity by disrupting interfacial interactions. Our results provide principles of drug action that may facilitate future design of small molecules to ameliorate TRPC6-mediated diseases.


Subject(s)
Calcium Channel Blockers/metabolism , Diglycerides/metabolism , TRPC6 Cation Channel/chemistry , Binding Sites , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Cryoelectron Microscopy , Diglycerides/chemistry , Diglycerides/pharmacology , Glomerulosclerosis, Focal Segmental/genetics , Humans , Ligands , Models, Molecular , Mutation , Protein Conformation , Protein Domains , TRPC6 Cation Channel/agonists , TRPC6 Cation Channel/antagonists & inhibitors , TRPC6 Cation Channel/metabolism
6.
PLoS One ; 14(10): e0222702, 2019.
Article in English | MEDLINE | ID: mdl-31600235

ABSTRACT

Slope one is a popular recommendation algorithm due to its simplicity and high efficiency for sparse data. However, it often suffers from under-fitting since the global information of all relevant users/items are considered. In this paper, we propose a new scheme called enhanced slope one recommendation through local information embedding. First, we employ clustering algorithms to obtain the user clusters as well as item clusters to represent local information. Second, we predict ratings using the local information of users and items in the same cluster. The local information can detect strong localized associations shared within clusters. Third, we design different fusion approaches based on the local information embedding. In this way, both under-fitting and over-fitting problems are alleviated. Experiment results on the real datasets show that our approaches defeats slope one in terms of both mean absolute error and root mean square error.


Subject(s)
Cluster Analysis , Data Interpretation, Statistical , Mathematics , Algorithms , Habits , Humans , Linear Models , Research Design
7.
PLoS One ; 14(5): e0217716, 2019.
Article in English | MEDLINE | ID: mdl-31136640

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0216095.].

8.
PLoS One ; 14(5): e0216095, 2019.
Article in English | MEDLINE | ID: mdl-31042744

ABSTRACT

Trastuzumab and pertuzumab are monoclonal antibodies that bind to distinct subdomains of the extracellular domain of human epidermal growth factor receptor 2 (HER2). Adding these monoclonal antibodies to the treatment regimen of HER2-positive breast cancer has changed the paradigm for treatment in that form of cancer. Synergistic activity has been observed with the combination of these two antibodies leading to hypotheses regarding the mechanism(s) and to the development of bispecific antibodies to maximize the clinical effect further. Although the individual crystal structures of HER2-trastuzumab and HER2-pertuzumab revealed the distinct binding sites and provided the structural basis for their anti-tumor activities, detailed structural information on the HER2-trastuzumab-pertuzumab complex has been elusive. Here we present the cryo-EM structure of HER2-trastuzumab-pertuzumab at 4.36 Å resolution. Comparison with the binary complexes reveals no cooperative interaction between trastuzumab and pertuzumab, and provides key insights into the design of novel, high-avidity bispecific molecules with potentially greater clinical efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized/ultrastructure , Receptor, ErbB-2/ultrastructure , Trastuzumab/ultrastructure , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/ultrastructure , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Cryoelectron Microscopy/methods , Female , Humans , Taxoids/therapeutic use , Trastuzumab/pharmacology
9.
J Biol Chem ; 293(38): 14678-14688, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30068552

ABSTRACT

Fibroblast growth factor 21 (FGF21), an endocrine hormone in the FGF family, plays a critical role in regulating metabolic homeostasis and has emerged as a therapeutic target for metabolic diseases, including Type 2 diabetes mellitus. FGF21 functions through a receptor complex that consists of an FGF receptor (FGFR) and a co-receptor ß-Klotho. Here, we identify and biochemically and structurally characterize 39F7, a high-affinity agonistic monoclonal antibody (mAb) against ß-Klotho that mimics FGF21 function. The co-crystal structure of ß-Klotho KL1 domain in complex with 39F7 Fab revealed that the recognition of 39F7 is centered on Trp-295 of ß-Klotho in a FGF21 noncompetitive manner. KL1 adopts a (ß/α)8 TIM barrel fold which resembles that of ß-glycosylceramidase, but lacks molecular features for enzymatic activity, suggesting that KL1 functions as a scaffold protein instead. In vitro characterization demonstrated that, although 39F7 does not compete with FGF21, it is specific for ß-Klotho/FGFR1c activation. Furthermore, the agonistic activity of 39F7 required the full IgG molecule to be bivalent, suggesting that 39F7 functions by promoting receptor/co-receptor dimerization. Supported by negative stain EM analysis of full-length ß-Klotho, we propose a molecular model wherein the agonistic antibody 39F7 acts in a ß-Klotho- and FGFR1c-dependent manner, mimicking FGF21 activity. More importantly, 39F7 offers promising therapeutic potential in the axis of FGF21 signaling as an antibody therapy alternative to FGF21 analogs for treatment of metabolic diseases.


Subject(s)
Antibodies, Monoclonal/immunology , Fibroblast Growth Factors/metabolism , Membrane Proteins/immunology , Animals , Antibody Affinity , CHO Cells , Cricetulus , Crystallography, X-Ray , Humans , Klotho Proteins , Membrane Proteins/agonists , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Microscopy, Electron , Protein Binding , Protein Conformation , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction
10.
Chin J Nat Med ; 15(6): 442-450, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28629534

ABSTRACT

The aims of the present study were to determine the effects of heparin-derived oligosaccharides (HDOs) on vascular intimal hyperplasia (IH) in balloon-injured carotid artery and to elucidate the underlying mechanisms of action. An animal model was established by rubbing the endothelia within the common carotid artery (CCA) in male rabbits. The rabbits were fed a high-cholesterol diet. Arterial IH was determined by histopathological changes to the CCA. Serum lipids were detected using an automated biochemical analysis. Expressions of mRNAs for vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), vascular cell adhesion molecule-1 (VCAM-1), monocyte chemoattractant protein-1 (MCP-1), scavenger receptor class B type I (SR-BI), and ATP-binding cassette transporter A1 (ABCA-1) were analyzed using reverse transcription polymerase chain reaction assays. Expressions of VEGF, VCAM-1, MCP-1, SR-BI and ABCA-1 proteins were analyzed by Western blotting. Enzyme-linked immunosorbent assays were used to quantify expression levels of VEGF and bFGF. Our results showed that administration of HDO significantly inhibited CCA histopathology and restenosis induced by balloon injury. The treatment with HDOs significantly decreased the mRNA and protein expression levels of VEGF, bFGF, VCAM-1, MCP-1, and SR-BI in the arterial wall; however, ABCA-1 expression level was elevated. HDO treatment led to a reduction in serum lipids (total cholesterol, triglycerides, high-density and low-density lipoproteins). Our results from the rabbit model indicated that HDOs could ameliorate IH and underlying mechanism might involve VEGF, bFGF, VCAM-1, MCP-1, SR-BI, and ABCA-1.


Subject(s)
Carotid Artery Injuries/drug therapy , Heparin/therapeutic use , Oligosaccharides/therapeutic use , Tunica Intima/pathology , ATP Binding Cassette Transporter 1/analysis , Animals , Carotid Artery Injuries/pathology , Chemokine CCL2/analysis , Hyperplasia , Male , Rabbits , Vascular Cell Adhesion Molecule-1/analysis , Vascular Endothelial Growth Factor A/analysis
11.
Cell ; 148(3): 530-42, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22304919

ABSTRACT

Budding of COPI-coated vesicles from Golgi membranes requires an Arf family G protein and the coatomer complex recruited from cytosol. Arf is also required with coatomer-related clathrin adaptor complexes to bud vesicles from the trans-Golgi network and endosomal compartments. To understand the structural basis for Arf-dependent recruitment of a vesicular coat to the membrane, we determined the structure of Arf1 bound to the γζ-COP subcomplex of coatomer. Structure-guided biochemical analysis reveals that a second Arf1-GTP molecule binds to ßδ-COP at a site common to the γ- and ß-COP subunits. The Arf1-binding sites on coatomer are spatially related to PtdIns4,5P(2)-binding sites on the endocytic AP2 complex, providing evidence that the orientation of membrane binding is general for this class of vesicular coat proteins. A bivalent GTP-dependent binding mode has implications for the dynamics of coatomer interaction with the Golgi and for the selection of cargo molecules.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Animals , COP-Coated Vesicles/chemistry , COP-Coated Vesicles/metabolism , Golgi Apparatus/metabolism , Guanosine Triphosphate/metabolism , Intracellular Membranes/metabolism , Models, Molecular , Sequence Alignment
12.
Structure ; 19(8): 1084-96, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21827945

ABSTRACT

Activation of procaspase-9 on the apoptosome is a pivotal step in the intrinsic cell death pathway. We now provide further evidence that caspase recruitment domains of pc-9 and Apaf-1 form a CARD-CARD disk that is flexibly tethered to the apoptosome. In addition, a 3D reconstruction of the pc-9 apoptosome was calculated without symmetry restraints. In this structure, p20 and p10 catalytic domains of a single pc-9 interact with nucleotide binding domains of adjacent Apaf-1 subunits. Together, disk assembly and pc-9 binding create an asymmetric proteolysis machine. We also show that CARD-p20 and p20-p10 linkers play important roles in pc-9 activation. Based on the data, we propose a proximity-induced association model for pc-9 activation on the apoptosome. We also show that pc-9 and caspase-3 have overlapping binding sites on the central hub. These binding sites may play a role in pc-3 activation and could allow the formation of hybrid apoptosomes with pc-9 and caspase-3 proteolytic activities.


Subject(s)
Apoptosomes/chemistry , Caspase 3/chemistry , Caspase 9/chemistry , Amino Acid Sequence , Apoptosis , Apoptotic Protease-Activating Factor 1/chemistry , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/genetics , Enzyme Activation , Humans , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Thrombin/chemistry
13.
Structure ; 19(1): 128-40, 2011 Jan 12.
Article in English | MEDLINE | ID: mdl-21220123

ABSTRACT

The Drosophila Apaf-1 related killer forms an apoptosome in the intrinsic cell death pathway. In this study we show that Dark forms a single ring when initiator procaspases are bound. This Dark-Dronc complex cleaves DrICE efficiently; hence, a single ring represents the Drosophila apoptosome. We then determined the 3D structure of a double ring at ∼6.9 Å resolution and created a model of the apoptosome. Subunit interactions in the Dark complex are similar to those in Apaf-1 and CED-4 apoptosomes, but there are significant differences. In particular, Dark has "lost" a loop in the nucleotide-binding pocket, which opens a path for possible dATP exchange in the apoptosome. In addition, caspase recruitment domains (CARDs) form a crown on the central hub of the Dark apoptosome. This CARD geometry suggests that conformational changes will be required to form active Dark-Dronc complexes. When taken together, these data provide insights into apoptosome structure, function, and evolution.


Subject(s)
Apoptosomes/chemistry , Caspases/chemistry , Drosophila Proteins/chemistry , Drosophila melanogaster/chemistry , Animals , Cryoelectron Microscopy , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Structural Homology, Protein
14.
Structure ; 18(5): 571-83, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20462491

ABSTRACT

Apaf-1 coassembles with cytochrome c to form the apoptosome, which then binds and activates procaspase-9 (pc-9). We removed pc-9 catalytic domains from the holoapoptosome by site-directed thrombinolysis. A structure of the resulting apoptosome-pc-9 CARD complex was then determined at approximately 9.5 A resolution. In our model, the central hub is constructed like other AAA+ protein rings but also contains novel features. At higher radius, the regulatory region of each Apaf-1 is comprised of tandem seven and eight blade beta-propellers with cytochrome c docked between them. Remarkably, Apaf-1 CARDs are disordered in the ground state. During activation, each Apaf-1 CARD interacts with a pc-9 CARD and these heterodimers form a flexibly tethered "disk" that sits above the central hub. When taken together, the data reveal conformational changes during Apaf-1 assembly that allow pc-9 activation. The model also provides a plausible explanation for the effects of NOD mutations that have been mapped onto the central hub.


Subject(s)
Apoptosomes/metabolism , Caspase 9/metabolism , Proteins/chemistry , Proteins/metabolism , Cytochrome c Group , Cytochromes c/chemistry , Cytochromes c/metabolism , Cytochromes c/pharmacology , Humans , Proteins/physiology
15.
J Mol Biol ; 355(3): 577-89, 2006 Jan 20.
Article in English | MEDLINE | ID: mdl-16310803

ABSTRACT

The Drosophila Apaf-1 related killer (Dark) forms an apoptosome that activates Dronc, an apical procaspase in the intrinsic cell death pathway. To study this process, we assembled a large Dark complex in the presence of dATP. Remarkably, we found that cytochrome c was not required for assembly and when added, cytochrome c did not bind to the Dark complex. We then determined a 3D structure of the Dark complex at 18.8A resolution using electron cryo-microscopy and single particle methods. In the structure, eight Dark subunits form a wheel-like particle and two of these rings associate face-to-face. In contrast, Apaf-1 forms a single ring that is comprised of seven subunits and each Apaf-1 binds a molecule of cytochrome c. We then used relevant crystal structures to model the Dark complex. This analysis shows that a single Dark ring and the Apaf-1 apoptosome share many key features. When taken together, the data suggest that a single ring in the Dark complex may represent the Drosophila apoptosome. Thus, our analysis provides a domain model of this complex and gives insights into its function.


Subject(s)
Apoptosis , Drosophila Proteins/chemistry , Drosophila/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Proteins/chemistry , Animals , Apoptotic Protease-Activating Factor 1 , Cryoelectron Microscopy , Cytochromes c/chemistry , Drosophila Proteins/ultrastructure , Humans , Protein Subunits/chemistry , Proteins/ultrastructure
16.
Structure ; 13(11): 1725-35, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16271896

ABSTRACT

Apaf-1 and cytochrome c coassemble in the presence of dATP to form the apoptosome. We have determined a structure of the apoptosome at 12.8 A resolution by using electron cryomicroscopy and single-particle methods. We then docked appropriate crystal structures into the map to create an accurate domain model. Thus, we found that seven caspase recruitment domains (CARDs) form a central ring within the apoptosome. At a larger radius, seven copies of the nucleotide binding and oligomerization domain (NOD) associate laterally to form the hub, which encircles the CARD ring. Finally, an arm-like helical domain (HD2) links each NOD to a pair of beta propellers, which bind a single cytochrome c. This model provides insights into the roles of dATP and cytochrome c in assembly. Our structure also reveals how a CARD ring and the central hub combine to create a platform for procaspase-9 activation.


Subject(s)
Apoptosis/physiology , Apoptotic Protease-Activating Factor 1/chemistry , Cytochromes c/chemistry , Deoxyadenine Nucleotides/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/metabolism , Cell Death/physiology , Crystallography, X-Ray , Cytochromes c/metabolism , Humans , Protein Structure, Tertiary , Sequence Analysis, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...