Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Biophys J ; 123(2): 235-247, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38102828

RESUMO

The use of bispecific antibodies as T cell engagers can bypass the normal T cell receptor-major histocompatibility class interaction, redirect the cytotoxic activity of T cells, and lead to highly efficient tumor cell killing. However, this immunotherapy also causes significant on-target off-tumor toxicologic effects, especially when it is used to treat solid tumors. To avoid these adverse events, it is necessary to understand the fundamental mechanisms involved in the physical process of T cell engagement. We developed a multiscale computational framework to reach this goal. The framework combines simulations on the intercellular and multicellular levels. On the intercellular level, we simulated the spatial-temporal dynamics of three-body interactions among bispecific antibodies, CD3 and tumor-associated antigens (TAAs). The derived number of intercellular bonds formed between CD3 and TAAs was further transferred to the multicellular simulations as the input parameter of adhesive density between cells. Through the simulations under various molecular and cellular conditions, we were able to gain new insights into how to adopt the most appropriate strategy to maximize the drug efficacy and avoid the off-target effect. For instance, we discovered that the low antibody-binding affinity resulted in the formation of large clusters at the cell-cell interface, which could be important to control the downstream signaling pathways. We also tested different molecular architectures of the bispecific antibody and suggested the existence of an optimal length in regulating the T cell engagement. Overall, the current multiscale simulations serve as a proof-of-concept study to help in the future design of new biological therapeutics.


Assuntos
Anticorpos Biespecíficos , Neoplasias , Humanos , Linfócitos T , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/uso terapêutico , Complexo CD3/farmacologia , Neoplasias/tratamento farmacológico , Imunoterapia/métodos
2.
Proteins ; 92(4): 567-580, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38050713

RESUMO

Cells detect changes in their external environments or communicate with each other through proteins on their surfaces. These cell surface proteins form a complicated network of interactions in order to fulfill their functions. The interactions between cell surface proteins are highly dynamic and, thus, challenging to detect using traditional experimental techniques. Here, we tackle this challenge using a computational framework. The primary focus of the framework is to develop new tools to identify interactions between domains in the immunoglobulin (Ig) fold, which is the most abundant domain family in cell surface proteins. These interactions could be formed between ligands and receptors from different cells or between proteins on the same cell surface. In practice, we collected all structural data on Ig domain interactions and transformed them into an interface fragment pair library. A high-dimensional profile can then be constructed from the library for a given pair of query protein sequences. Multiple machine learning models were used to read this profile so that the probability of interaction between the query proteins could be predicted. We tested our models on an experimentally derived dataset that contains 564 cell surface proteins in humans. The cross-validation results show that we can achieve higher than 70% accuracy in identifying the PPIs within this dataset. We then applied this method to a group of 46 cell surface proteins in Caenorhabditis elegans. We screened every possible interaction between these proteins. Many interactions recognized by our machine learning classifiers have been experimentally confirmed in the literature. In conclusion, our computational platform serves as a useful tool to help identify potential new interactions between cell surface proteins in addition to current state-of-the-art experimental techniques. The tool is freely accessible for use by the scientific community. Moreover, the general framework of the machine learning classification can also be extended to study the interactions of proteins in other domain superfamilies.


Assuntos
Aprendizado de Máquina , Proteínas de Membrana , Humanos , Sequência de Aminoácidos , Imunoglobulinas , Ligantes
3.
J Proteome Res ; 21(2): 349-359, 2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-34978816

RESUMO

The interactions between ectodomains of cell surface proteins are vital players in many important cellular processes, such as regulating immune responses, coordinating cell differentiation, and shaping neural plasticity. However, while the construction of a large-scale protein interactome has been greatly facilitated by the development of high-throughput experimental techniques, little progress has been made to support the discovery of extracellular interactome for cell surface proteins. Harnessed by the recent advances in computational modeling of protein-protein interactions, here we present a structure-based online database for the extracellular interactome of cell surface proteins in humans, called EXCESP. The database contains both experimentally determined and computationally predicted interactions among all type-I transmembrane proteins in humans. All structural models for these interactions and their binding affinities were further computationally modeled. Moreover, information such as expression levels of each protein in different cell types and its relation to various signaling pathways from other online resources has also been integrated into the database. In summary, the database serves as a valuable addition to the existing online resources for the study of cell surface proteins. It can contribute to the understanding of the functions of cell surface proteins in the era of systems biology.


Assuntos
Proteínas de Membrana , Biologia de Sistemas , Biologia Computacional/métodos , Humanos , Proteínas de Membrana/genética , Mapeamento de Interação de Proteínas/métodos , Transdução de Sinais
4.
PLoS Comput Biol ; 17(3): e1008825, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33684103

RESUMO

The activation and differentiation of T-cells are mainly directly by their co-regulatory receptors. T lymphocyte-associated protein-4 (CTLA-4) and programed cell death-1 (PD-1) are two of the most important co-regulatory receptors. Binding of PD-1 and CTLA-4 with their corresponding ligands programed cell death-ligand 1 (PD-L1) and B7 on the antigen presenting cells (APC) activates two central co-inhibitory signaling pathways to suppress T cell functions. Interestingly, recent experiments have identified a new cis-interaction between PD-L1 and B7, suggesting that a crosstalk exists between two co-inhibitory receptors and the two pairs of ligand-receptor complexes can undergo dynamic oligomerization. Inspired by these experimental evidences, we developed a coarse-grained model to characterize the assembling of an immune complex consisting of CLTA-4, B7, PD-L1 and PD-1. These four proteins and their interactions form a small network motif. The temporal dynamics and spatial pattern formation of this network was simulated by a diffusion-reaction algorithm. Our simulation method incorporates the membrane confinement of cell surface proteins and geometric arrangement of different binding interfaces between these proteins. A wide range of binding constants was tested for the interactions involved in the network. Interestingly, we show that the CTLA-4/B7 ligand-receptor complexes can first form linear oligomers, while these oligomers further align together into two-dimensional clusters. Similar phenomenon has also been observed in other systems of cell surface proteins. Our test results further indicate that both co-inhibitory signaling pathways activated by B7 and PD-L1 can be down-regulated by the new cis-interaction between these two ligands, consistent with previous experimental evidences. Finally, the simulations also suggest that the dynamic and the spatial properties of the immune complex assembly are highly determined by the energetics of molecular interactions in the network. Our study, therefore, brings new insights to the co-regulatory mechanisms of T cell activation.


Assuntos
Complexo Antígeno-Anticorpo , Células Apresentadoras de Antígenos , Linfócitos T , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/metabolismo , Células Apresentadoras de Antígenos/química , Células Apresentadoras de Antígenos/metabolismo , Antígenos B7/química , Antígenos B7/metabolismo , Antígeno B7-H1/química , Antígeno B7-H1/metabolismo , Antígeno CTLA-4/química , Antígeno CTLA-4/metabolismo , Biologia Computacional , Humanos , Simulação de Dinâmica Molecular , Receptor de Morte Celular Programada 1/química , Receptor de Morte Celular Programada 1/metabolismo , Ligação Proteica , Linfócitos T/química , Linfócitos T/metabolismo
5.
Proteins ; 89(7): 884-895, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33620752

RESUMO

Multi-domain proteins are not only formed through natural evolution but can also be generated by recombinant DNA technology. Because many fusion proteins can enhance the selectivity of cell targeting, these artificially produced molecules, called multi-specific biologics, are promising drug candidates, especially for immunotherapy. Moreover, the rational design of domain linkers in fusion proteins is becoming an essential step toward a quantitative understanding of the dynamics in these biopharmaceutics. We developed a computational framework to characterize the impacts of peptide linkers on the dynamics of multi-specific biologics. Specifically, we first constructed a benchmark containing six types of linkers that represent various lengths and degrees of flexibility and used them to connect two natural proteins as a test system. We then projected the microsecond dynamics of these proteins generated from Anton onto a coarse-grained conformational space. We further analyzed the similarity of dynamics among different proteins in this low-dimensional space by a neural-network-based classification model. Finally, we applied hierarchical clustering to place linkers into different subgroups based on the classification results. The clustering results suggest that the length of linkers, which is used to spatially separate different functional modules, plays the most important role in regulating the dynamics of this fusion protein. Given the same number of amino acids, linker flexibility functions as a regulator of protein dynamics. In summary, we illustrated that a new computational strategy can be used to study the dynamics of multi-domain fusion proteins by a combination of long timescale molecular dynamics simulation, coarse-grained feature extraction, and artificial intelligence.


Assuntos
Aminoácidos/química , Inteligência Artificial , Mutagênese Insercional/métodos , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/química , Aminoácidos/metabolismo , Benchmarking , Biologia Computacional/métodos , Expressão Gênica , Humanos , Simulação de Dinâmica Molecular , Maleabilidade , Domínios Proteicos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
6.
J Membr Biol ; 254(4): 397-407, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34189599

RESUMO

During the final step of the bacteriophage infection cycle, the cytoplasmic membrane of host cells is disrupted by small membrane proteins called holins. The function of holins in cell lysis is carried out by forming a highly ordered structure called lethal lesion, in which the accumulation of holins in the cytoplasmic membrane leads to the sudden opening of a hole in the middle of this oligomer. Previous studies showed that dimerization of holins is a necessary step to induce their higher order assembly. However, the molecular mechanism underlying the holin-mediated lesion formation is not well understood. In order to elucidate the functions of holin, we first computationally constructed a structural model for our testing system: the holin S105 from bacteriophage lambda. All atom molecular dynamic simulations were further applied to refine its structure and study its dynamics as well as interaction in lipid bilayer. Additional simulations on association between two holins provide supportive evidence to the argument that the C-terminal region of holin plays a critical role in regulating the dimerization. In detail, we found that the adhesion of specific nonpolar residues in transmembrane domain 3 (TMD3) in a polar environment serves as the driven force of dimerization. Our study therefore brings insights to the design of binding interfaces between holins, which can be potentially used to modulate the dynamics of lesion formation.


Assuntos
Bacteriófago lambda , Proteínas Virais , Sequência de Aminoácidos , Bacteriófago lambda/química , Bacteriófago lambda/metabolismo , Dimerização , Sequências Hélice-Volta-Hélice , Proteínas Virais/química , Proteínas Virais/metabolismo
7.
Arch Biochem Biophys ; 710: 109001, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34352244

RESUMO

The enzyme cGAS functions as a sensor that recognizes the cytosolic DNA from foreign pathogen. The activation of the protein triggers the transcription of inflammatory genes, leading into the establishment of an antipathogen state. An interesting new discovery is that the detection of DNA by cGAS induced the formation of liquid-like droplets. However how cells regulate the formation of these droplets is still not fully understood. In order to unravel the molecular mechanism beneath the DNA-mediated phase separation of cGAS, we developed a polymer-based coarse-grained model which takes into accounts the basic structural organization in DNA and cGAS, as well as the binding properties between these biomolecules. This model was further integrated into a hybrid simulation algorithm. With this computational method, a multi-step kinetic process of aggregation between cGAS and DNA was observed. Moreover, we systematically tested the model under different concentrations and binding parameters. Our simulation results show that phase separation requires both cGAS dimerization and protein-DNA interactions, whereas polymers can be kinetically trapped in small aggregates under strong binding affinities. Additionally, we demonstrated that supramolecular assembly can be facilitated by increasing the number of functional modules in protein or DNA polymers, suggesting that multivalency and intrinsic disordered regions play positive roles in regulating phase separation. This is consistent to previous experimental evidences. Taken together, this is, to the best of our knowledge, the first computational model to study condensation of cGAS-DNA complexes. While the method can reach the timescale beyond the capability of atomic-level MD simulations, it still includes information about spatial arrangement of functional modules in biopolymers that is missing in the mean-field theory. Our work thereby adds a useful dimension to a suite of existing experimental and computational techniques to study the dynamics of phase separation in biological systems.


Assuntos
DNA/química , DNA/metabolismo , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Algoritmos , Simulação por Computador , Humanos , Cinética , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Modelos Biológicos , Modelos Moleculares , Agregados Proteicos , Transdução de Sinais
8.
J Chem Phys ; 154(5): 055101, 2021 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-33557556

RESUMO

Binding of cell surface receptors with their extracellular ligands initiates various intracellular signaling pathways. However, our understanding of the cellular functions of these receptors is very limited due to the fact that in vivo binding between ligands and receptors has only been successfully measured in a very small number of cases. In living cells, receptors are anchored on surfaces of the plasma membrane, which undergoes thermal undulations. Moreover, it has been observed in various systems that receptors can be organized into oligomers prior to ligand binding. It is not well understood how these cellular factors play roles in regulating the dynamics of ligand-receptor interactions. Here, we tackled these problems by using a coarse-grained kinetic Monte Carlo simulation method. Using this method, we demonstrated that the membrane undulations cause a negative effect on ligand-receptor interactions. We further found that the preassembly of membrane receptors on the cell surface can not only accelerate the kinetics of ligand binding but also reduce the noises during the process. In general, our study highlights the importance of membrane environments in regulating the function of membrane receptors in cells. The simulation method can be potentially applied to specific receptor systems involved in cell signaling.


Assuntos
Simulação por Computador , Receptores de Superfície Celular/metabolismo , Membrana Celular/metabolismo , Ligantes , Método de Monte Carlo
9.
Biophys J ; 119(10): 2116-2126, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33113350

RESUMO

Scaffold proteins are central players in regulating the spatial-temporal organization of many important signaling pathways in cells. They offer physical platforms to downstream signaling proteins so that their transient interactions in a crowded and heterogeneous environment of cytosol can be greatly facilitated. However, most scaffold proteins tend to simultaneously bind more than one signaling molecule, which leads to the spatial assembly of multimeric protein complexes. The kinetics of these protein oligomerizations are difficult to quantify by traditional experimental approaches. To understand the functions of scaffold proteins in cell signaling, we developed a, to our knowledge, new hybrid simulation algorithm in which both spatial organization and binding kinetics of proteins were implemented. We applied this new technique to a simple network system that contains three molecules. One molecule in the network is a scaffold protein, whereas the other two are its binding targets in the downstream signaling pathway. Each of the three molecules in the system contains two binding motifs that can interact with each other and are connected by a flexible linker. By applying the new simulation method to the model, we show that the scaffold proteins will promote not only thermodynamics but also kinetics of cell signaling given the premise that the interaction between the two signaling molecules is transient. Moreover, by changing the flexibility of the linker between two binding motifs, our results suggest that the conformational fluctuations in a scaffold protein play a positive role in recruiting downstream signaling molecules. In summary, this study showcases the capability of computational simulation in understanding the general principles of scaffold protein functions.


Assuntos
Proteínas , Transdução de Sinais , Simulação por Computador , Cinética , Ligação Proteica , Proteínas/metabolismo , Termodinâmica
10.
Proteins ; 88(5): 698-709, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31710744

RESUMO

The interactions between tumor necrosis factors (TNFs) and their corresponding receptors (TNFRs) play a pivotal role in inflammatory responses. Upon ligand binding, TNFR receptors were found to form oligomers on cell surfaces. However, the underlying mechanism of oligomerization is not fully understood. In order to tackle this problem, molecular dynamics (MD) simulations have been applied to the complex between TNF receptor-1 (TNFR1) and its ligand TNF-α as a specific test system. The simulations on both all-atom (AA) and coarse-grained (CG) levels achieved the similar results that the extracellular domains of TNFR1 can undergo large fluctuations on plasma membrane, while the dynamics of TNFα-TNFR1 complex is much more constrained. Using the CG model with the Martini force field, we are able to simulate the systems that contain multiple TNFα-TNFR1 complexes with the timescale of microseconds. We found that complexes can aggregate into oligomers on the plasma membrane through the lateral interactions between receptors at the end of the CG simulations. We suggest that this spatial organization is essential to the efficiency of signal transduction for ligands that belong to the TNF superfamily. We further show that the aggregation of two complexes is initiated by the association between the N-terminal domains of TNFR1 receptors. Interestingly, the cis-interfaces between N-terminal regions of two TNF receptors have been observed in the previous X-ray crystallographic experiment. Therefore, we provide supportive evidence that cis-interface is of functional importance in triggering the receptor oligomerization. Taken together, our study brings insights to understand the molecular mechanism of TNF signaling.


Assuntos
Membrana Celular/química , Simulação de Dinâmica Molecular , Receptores Tipo I de Fatores de Necrose Tumoral/química , Fator de Necrose Tumoral alfa/química , Sítios de Ligação , Membrana Celular/metabolismo , Humanos , Cinética , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Termodinâmica , Fator de Necrose Tumoral alfa/metabolismo
11.
Int J Mol Sci ; 21(5)2020 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-32150842

RESUMO

Ligands in the tumor necrosis factor (TNF) superfamily are one major class of cytokines that bind to their corresponding receptors in the tumor necrosis factor receptor (TNFR) superfamily and initiate multiple intracellular signaling pathways during inflammation, tissue homeostasis, and cell differentiation. Mutations in the genes that encode TNF ligands or TNFR receptors result in a large variety of diseases. The development of therapeutic treatment for these diseases can be greatly benefitted from the knowledge on binding properties of these ligand-receptor interactions. In order to complement the limitations in the current experimental methods that measure the binding constants of TNF/TNFR interactions, we developed a new simulation strategy to computationally estimate the association and dissociation between a ligand and its receptor. We systematically tested this strategy to a comprehensive dataset that contained structures of diverse complexes between TNF ligands and their corresponding receptors in the TNFR superfamily. We demonstrated that the binding stabilities inferred from our simulation results were compatible with existing experimental data. We further compared the binding kinetics of different TNF/TNFR systems, and explored their potential functional implication. We suggest that the transient binding between ligands and cell surface receptors leads into a dynamic nature of cross-membrane signal transduction, whereas the slow but strong binding of these ligands to the soluble decoy receptors is naturally designed to fulfill their functions as inhibitors of signal activation. Therefore, our computational approach serves as a useful addition to current experimental techniques for the quantitatively comparison of interactions across different members in the TNF and TNFR superfamily. It also provides a mechanistic understanding to the functions of TNF-associated cell signaling pathways.


Assuntos
Simulação por Computador , Conformação Proteica , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Humanos , Cinética , Ligantes , Ligação Proteica , Receptores do Fator de Necrose Tumoral/química , Transdução de Sinais , Fator de Necrose Tumoral alfa/química
12.
Phys Chem Chem Phys ; 21(5): 2463-2471, 2019 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-30652698

RESUMO

Proteins carry out their diverse functions in cells by forming interactions with each other. The dynamics of these interactions are quantified by the measurement of association and dissociation rate constants. Relative to the efforts made to model the association of biomolecules, little has been studied to understand the principles of protein complex dissociation. Using the interaction between colicin E9 endonucleases and immunity proteins as a test system, here we develop a coarse-grained simulation method to explore the dissociation mechanisms of protein complexes. The interactions between proteins in the complex are described by the knowledge-based potential that was constructed by the statistics from available protein complexes in the structural database. Our study provides the supportive evidences to the dual recognition mechanism for the specificity of binding between E9 DNase and immunity proteins, in which the conserved residues of helix III of Im2 and Im9 proteins act as the anchor for binding, while the sequence variations in helix II make positive or negative contributions to specificity. Beyond that, we further suggest that this binding specificity is rooted in the process of complex dissociation instead of association. While we increased the flexibility of protein complexes, we further found that they are less prone to dissociation, suggesting that conformational fluctuations of protein complexes play important functional roles in regulating their binding and dissociation. Our studies therefore bring new insights to the molecule mechanisms of protein-protein interactions, while the method can serve as a new addition to a suite of existing computational tools for the simulations of protein complexes.


Assuntos
Proteínas de Transporte/metabolismo , Colicinas/metabolismo , Endodesoxirribonucleases/metabolismo , Proteínas de Escherichia coli/metabolismo , Sítios de Ligação , Proteínas de Transporte/química , Colicinas/química , Endodesoxirribonucleases/química , Escherichia coli/química , Proteínas de Escherichia coli/química , Interações Hidrofóbicas e Hidrofílicas , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica , Eletricidade Estática
13.
Phys Rev Lett ; 119(10): 108102, 2017 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-28949191

RESUMO

To provide insights into the stabilizing mechanisms of trimethylamine-N-oxide (TMAO) on protein structures, we perform all-atom molecular dynamics simulations of peptides and the Trp-cage miniprotein. The effects of TMAO on the backbone and charged residues of peptides are found to stabilize compact conformations, whereas effects of TMAO on nonpolar residues lead to peptide swelling. This suggests competing mechanisms of TMAO on proteins, which accounts for hydrophobic swelling, backbone collapse, and stabilization of charge-charge interactions. These mechanisms are observed in Trp cage.


Assuntos
Metilaminas/química , Simulação de Dinâmica Molecular , Peptídeos/química , Proteínas/química , Interações Hidrofóbicas e Hidrofílicas , Conformação Molecular
14.
Proteins ; 83(11): 1963-72, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26264694

RESUMO

Here, we provide insights into the thermodynamic properties of A ß16-21 dissociation from an amyloid fibril using all-atom molecular dynamics simulations in explicit water. An umbrella sampling protocol is used to compute potentials of mean force (PMF) as a function of the distance ξ between centers-of-mass of the A ß16-21 peptide and the preformed fibril at nine temperatures. Changes in the enthalpy and the entropic energy are determined from the temperature dependence of these PMF(s) and the average volume of the simulation box is computed as a function of ξ. We find that the PMF at 310 K is dominated by enthalpy while the entropic energy does not change significantly during dissociation. The volume of the system decreases during dissociation. Moreover, the magnitude of this volume change also decreases with increasing temperature. By defining dock and lock states using the solvent accessible surface area (SASA), we find that the behavior of the electrostatic energy is different in these two states. It increases (unfavorable) and decreases (favorable) during dissociation in lock and dock states, respectively, while the energy due to Lennard-Jones interactions increases continuously in these states. Our simulations also highlight the importance of hydrophobic interactions in accounting for the stability of A ß16-21.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Eletricidade Estática , Termodinâmica
15.
Comput Biol Chem ; 110: 108080, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38643609

RESUMO

The physical interactions between proteins are largely determined by the structural properties at their binding interfaces. It was found that the binding interfaces in distinctive protein complexes are highly similar. The structural properties underlying different binding interfaces could be further captured by artificial intelligence. In order to test this hypothesis, we broke protein-protein binding interfaces into pairs of interacting fragments. We employed a generative model to encode these interface fragment pairs in a low-dimensional latent space. After training, new conformations of interface fragment pairs were generated. We found that, by only using a small number of interface fragment pairs that were generated by artificial intelligence, we were able to guide the assembly of protein complexes into their native conformations. These results demonstrate that the conformational space of fragment pairs at protein-protein binding interfaces is highly degenerate. Features in this degenerate space can be well characterized by artificial intelligence. In summary, our machine learning method will be potentially useful to search for and predict the conformations of unknown protein-protein interactions.


Assuntos
Inteligência Artificial , Ligação Proteica , Proteínas , Proteínas/química , Proteínas/metabolismo , Conformação Proteica , Modelos Moleculares , Sítios de Ligação
16.
Biosystems ; 243: 105264, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38964652

RESUMO

Computational analysis of paratope-epitope interactions between antibodies and their corresponding antigens can facilitate our understanding of the molecular mechanism underlying humoral immunity and boost the design of new therapeutics for many diseases. The recent breakthrough in artificial intelligence has made it possible to predict protein-protein interactions and model their structures. Unfortunately, detecting antigen-binding sites associated with a specific antibody is still a challenging problem. To tackle this challenge, we implemented a deep learning model to characterize interaction patterns between antibodies and their corresponding antigens. With high accuracy, our model can distinguish between antibody-antigen complexes and other types of protein-protein complexes. More intriguingly, we can identify antigens from other common protein binding regions with an accuracy of higher than 70% even if we only have the epitope information. This indicates that antigens have distinct features on their surface that antibodies can recognize. Additionally, our model was unable to predict the partnerships between antibodies and their particular antigens. This result suggests that one antigen may be targeted by more than one antibody and that antibodies may bind to previously unidentified proteins. Taken together, our results support the precision of antibody-antigen interactions while also suggesting positive future progress in the prediction of specific pairing.

17.
J Cell Commun Signal ; 17(3): 657-671, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36167956

RESUMO

TNFα is a highly pleiotropic cytokine inducing inflammatory signaling pathways. It is initially presented on plasma membrane of cells (mTNFα), and also exists in a soluble variant (sTNFα) after cleavage. The ligand is shared by two structurally similar receptors, TNFR1 and TNFR2. Interestingly, while sTNFα preferentially stimulates TNFR1, TNFR2 signaling can only be activated by mTNFα. How can two similar receptors respond to the same ligand in such a different way? We employed computational simulations in multiple scales to address this question. We found that both mTNFα and sTNFα can trigger the clustering of TNFR1. The size of clusters induced by sTNFα is constantly larger than the clusters induced by mTNFα. The systems of TNFR2, on the other hand, show very different behaviors. Only when the interactions between TNFR2 are very weak, mTNFα can trigger the receptors to form very large clusters. Given the same weak binding affinity, only small oligomers were obtained in the system of sTNFα. Considering that TNF-mediated signaling is modulated by the ligand-induced clustering of receptors on cell surface, our study provided the mechanistic foundation to the phenomenon that different isoforms of the ligand can lead to highly distinctive signaling patterns for its receptors.

18.
Protein Sci ; 32(2): e4552, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36541820

RESUMO

The development of artificial protein cages has recently gained massive attention due to their promising application prospect as novel delivery vehicles for therapeutics. These nanoparticles are formed through a process called self-assembly, in which individual subunits spontaneously arrange into highly ordered patterns via non-covalent but specific interactions. Therefore, the first step toward the design of novel engineered protein cages is to understand the general mechanisms of their self-assembling dynamics. Here we have developed a new computational method to tackle this problem. Our method is based on a coarse-grained model and a diffusion-reaction simulation algorithm. Using a tetrahedral cage as test model, we showed that self-assembly of protein cage requires of a seeding process in which specific configurations of kinetic intermediate states are identified. We further found that there is a critical concentration to trigger self-assembly of protein cages. This critical concentration allows that cages can only be successfully assembled under a persistently high concentration. Additionally, phase diagram of self-assembly has been constructed by systematically testing the model across a wide range of binding parameters. Finally, our simulations demonstrated the importance of protein's structural flexibility in regulating the dynamics of cage assembly. In summary, this study throws lights on the general principles underlying self-assembly of large cage-like protein complexes and thus provides insights to design new nanomaterials.


Assuntos
Nanopartículas , Nanoestruturas , Proteínas/química , Simulação por Computador , Cinética
19.
Comput Biol Chem ; 103: 107823, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36682326

RESUMO

Proteins in the tumor necrosis factor (TNF) superfamily (TNFSF) regulate diverse cellular processes by interacting with their receptors in the TNF receptor (TNFR) superfamily (TNFRSF). Ligands and receptors in these two superfamilies form a complicated network of interactions, in which the same ligand can bind to different receptors and the same receptor can be shared by different ligands. In order to study these interactions on a systematic level, a TNFSF-TNFRSF interactome was constructed in this study by searching the database which consists of both experimentally measured and computationally predicted protein-protein interactions (PPIs). The interactome contains a total number of 194 interactions between 18 TNFSF ligands and 29 TNFRSF receptors in human. We modeled the structure for each ligand-receptor interaction in the network. Their binding affinities were further computationally estimated based on modeled structures. Our computational outputs, which are all publicly accessible, serve as a valuable addition to the currently limited experimental resources to study TNF-mediated cell signaling.


Assuntos
Receptores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa , Humanos , Ligantes , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/metabolismo
20.
bioRxiv ; 2023 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-37333150

RESUMO

The use of bispecific antibodies as T cell engagers can bypass the normal TCR-MHC interaction, redirect the cytotoxic activity of T-cells, and lead to highly efficient tumor cell killing. However, this immunotherapy also causes significant on-target off-tumor toxicologic effects, especially when they were used to treat solid tumors. In order to avoid these adverse events, it is necessary to understand the fundamental mechanisms during the physical process of T cell engagement. We developed a multiscale computational framework to reach this goal. The framework combines simulations on the intercellular and multicellular levels. On the intercellular level, we simulated the spatial-temporal dynamics of three-body interactions among bispecific antibodies, CD3 and TAA. The derived number of intercellular bonds formed between CD3 and TAA were further transferred into the multicellular simulations as the input parameter of adhesive density between cells. Through the simulations under various molecular and cellular conditions, we were able to gain new insights of how to adopt the most appropriate strategy to maximize the drug efficacy and avoid the off-target effect. For instance, we discovered that the low antibody binding affinity resulted in the formation of large clusters at the cell-cell interface, which could be important to control the downstream signaling pathways. We also tested different molecular architectures of the bispecific antibody and suggested the existence of an optimal length in regulating the T cell engagement. Overall, the current multiscale simulations serve as a prove-of-concept study to help the future design of new biological therapeutics. SIGNIFICANCE: T-cell engagers are a class of anti-cancer drugs that can directly kill tumor cells by bringing T cells next to them. However, current treatments using T-cell engagers can cause serious side-effects. In order to reduce these effects, it is necessary to understand how T cells and tumor cells interact together through the connection of T-cell engagers. Unfortunately, this process is not well studied due to the limitations in current experimental techniques. We developed computational models on two different scales to simulate the physical process of T cell engagement. Our simulation results provide new insights into the general properties of T cell engagers. The new simulation methods can therefore serve as a useful tool to design novel antibodies for cancer immunotherapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA