Structural Studies of Hot Target Proteins: Techniques, Insights, and Challenges
Hot target proteins, defined as proteins with significant relevance to disease treatment, biomolecular engineering, and drug discovery, have emerged as a focal point of structural biology. Understanding their structural and functional dynamics is critical for advancing therapeutic interventions and biotechnological applications. Recent advances in methodologies, including X-ray crystallography, nuclear magnetic resonance (NMR), cryo-electron microscopy (Cryo-EM), and computational modeling, have accelerated insights into the complex architectures of these proteins.
Creative Biostructure specializes in structural biology and membrane protein technology. Our services include X-ray crystallography, Cryo-EM, NMR spectroscopy, and more, all designed to support your target protein structure research needs.
Spotlight on Recent Advances
Hot Target Proteins: Structural Studies Driving the Future of Drug Design
Hot target proteins occupy a unique position in biomedical research due to their role in cellular processes and their potential for therapeutic targeting. These proteins often include kinases, ion channels, G protein-coupled receptors (GPCRs), and immune checkpoint proteins. Understanding their structure is critical to unraveling mechanisms of action and guiding rational drug design. Structural studies have evolved from static snapshots of molecular configurations to dynamic representations that integrate conformational flexibility and intermolecular interactions. This transition underscores the importance of high-resolution techniques and computational approaches in revealing the nuances of protein functionality.
Methodological Approaches to Studying Hot Target Proteins
X-Ray Crystallography: High-Resolution Structural Insights into Hot Target Proteins
X-ray crystallography has long been a cornerstone of structural biology, providing atomic-level detail that allows researchers to resolve the three-dimensional structures of proteins and protein-ligand complexes. However, this technique faces challenges with membrane proteins and highly dynamic proteins due to difficulties in crystallization and capturing conformational flexibility. Techniques such as serial femtosecond (SFX) crystallography have improved the ability to study dynamic processes and overcome the challenges posed by difficult-to-crystallize proteins, such as membrane proteins. Notable successes include resolving the structures of ion channels and enzymes that are central to disease pathways.
Figure 1. Utilizing SFX for studying SARS-CoV-2. SFX Results for the MPro and NendoU proteins from SARS-CoV-2. Top: Mpro structure at RT determined using SFX (PDB entry 7CWB), with a zoomed in view of the substrate docking site. Bottom: RT-SFX structure of the hexameric NendoU protein, colored by B-factors. The different binding pocket flexibility is highlighted for the two trimers comprising the hexamer. (Botha and Fromme, 2023)
NMR: Dynamic and Atomic-Level Characterization of Target Protein Structures
NMR spectroscopy offers a complementary approach that is excellent for elucidating the dynamics of proteins in solution. This technique is particularly useful for small to medium-sized proteins, allowing the study of folding pathways, ligand binding, and transient interactions. Advances in high-field magnets and isotopic labeling have extended NMR's applicability to larger protein complexes, providing insights into their dynamics and interactions.
Figure 2. NMR is a "gold standard" method in drug design and discovery. (Emwas et al., 2020)
Cryo-EM: Visualizing Complex Protein Structures in Near-Native States
Cryo-EM has revolutionized structural studies of macromolecular complexes, allowing for high-resolution visualization without the need for crystallization. Recent advances in direct electron detectors and image-processing algorithms have enabled the study of dynamic protein complexes, such as viral proteins and GPCRs, in near-native states.
Figure 3. Cryo-electron microscopy is used for GPCR research and drug discovery in endocrinology and metabolism. The basis of GPCR ligand recognition and signal transduction. a, Different ligand binding pockets in G protein-coupled receptors (GPCRs). From left to right: GPR84 bound to lipid 3-OH-C12 (Protein Databank (PDB): 8J18); GHSR bound to peptide ligand ghrelin (PDB: 7NA7); glucagon-like peptide 1 receptor (GLP1R) bound to peptide ligand Ex4-D-Ala (PDB: 7S1M); and chemokine receptor CCR6 bound to protein ligand CCL20 (PDB: 6WWZ). The ligands are shown in surface view (showing the external shape of the molecules), and the secondary structure of the receptors is shown. b, Structural comparison between the inactive melanocortin 4 receptor (MC4R) (PDB: 6W25) and Gs-coupled active MC4R (PDB: 7F55), and also between the inactive D4R (PDB: 5WIU) and Gi-coupled active D4R (PDB: 8IRU). The agonists for MC4R and D4R are shown in surface view together with the conserved toggle switches (W2586.48 and W4076.48, respectively) showing the covalent bonds between amino acid atoms. (Duan et al., 2024)
Computational Modeling: In Silico Approaches for Predicting and Refining Protein Structures
In silico approaches, including molecular dynamics (MD) simulations, homology modeling, and artificial intelligence-driven tools such as AlphaFold, have emerged as powerful complements to experimental techniques. These methods facilitate prediction of protein structures, exploration of conformational landscapes, and identification of binding sites for drug design.
Figure 4. AlphaFold produces highly accurate structures. a, The performance of AlphaFold on the CASP14 dataset (n = 87 protein domains) relative to the top-15 entries (out of 146 entries), group numbers correspond to the numbers assigned to entrants by CASP. b, Our prediction of CASP14 target T1049 (PDB 6Y4F, blue) compared with the true (experimental) structure (green). Four residues in the C terminus of the crystal structure are B-factor outliers and are not depicted. c, CASP14 target T1056 (PDB 6YJ1). An example of a well-predicted zinc-binding site (AlphaFold has accurate side chains even though it does not explicitly predict the zinc ion). d, CASP target T1044 (PDB 6VR4)—a 2,180-residue single chain—was predicted with correct domain packing (the prediction was made after CASP using AlphaFold without intervention). e, Model architecture. Arrows show the information flow among the various components described in this paper. Array shapes are shown in parentheses with s, number of sequences; r, number of residues; c, number of channels. (Jumper et al., 2021)
Key Insights from Structural Studies
Kinases: Gatekeepers of Cellular Signaling
Kinases regulate cellular processes through phosphorylation and are implicated in numerous diseases, including cancer and autoimmune disorders. In cancer, kinase mutations or overexpression can lead to dysregulated signaling pathways, promoting cell proliferation and survival. Structural studies have elucidated the mechanisms of kinase activation and inhibition, leading to the development of targeted therapies such as imatinib for chronic myeloid leukemia.
Figure 5. Structural features of the kinase catalytic domain and inhibitor binding modes. a | Structural overview of the kinase catalytic domain, based on the structure of protein kinase A (PKA) in ribbon representation. The main secondary structure elements are labelled in parts a and b. The ATP cofactor is shown in ball and stick representation. Mobile structural elements that regulate kinase activity including the glycine-rich loop (G-loop; blue), the activation segment (turquoise) and αC helix (red) are highlighted. Two Mg2+ ions are shown as solid spheres. b | Aligned catalytic (blue) and regulatory (yellow) spine residues in the PKA active state. The gatekeeper residue (M120, grey) bridges the two spines. The adenine ring of ATP bridges N-terminal and C-terminal lobe catalytic spine residues. (Attwood et al., 2021)
G-Protein-Coupled Receptors (GPCRs): Dynamic Signal Transducers
GPCRs are a large family of membrane proteins involved in signal transduction, playing critical roles in various physiological processes. Structural elucidation of GPCR-ligand complexes has been transformative for drug discovery, providing insight into ligand specificity and receptor activation. GPCRs are targeted by approximately 34% of FDA-approved drugs, highlighting their importance in treating diseases such as hypertension, asthma, and psychiatric disorders. Breakthroughs in cryo-EM have enabled the visualization of GPCRs bound to diverse ligands and signaling proteins, guiding the design of biased agonists for targeted therapies.
Ion Channels: Gateways to Cellular Communication
Ion channels facilitate the transport of ions across membranes and play critical roles in neural signaling and homeostasis. Structural studies have elucidated the gating mechanisms of voltage- and ligand-gated ion channels, informing the development of drugs for epilepsy, pain, and cardiac arrhythmias. In particular, cryo-EM has provided high-resolution structures of ion channels in various conformational states.
Figure 6. 3D reconstruction of rabbit TRPV2 and overall topology of the channel. (a) Cryo-EM reconstruction, showing the four-fold-symmetric TRPV2 homotetramer. Each promoter is colored differently. (b) The atomic model of TRPV2 built from the EM density, with the domain architecture delineated by different colors. (Zubcevic et al., 2016)
Immune Checkpoint Proteins: Modulators of Immune Responses
Immune checkpoint proteins, such as PD-1 and CTLA-4, regulate immune responses and have become hot targets in cancer immunotherapy. The development of immune checkpoint inhibitors, such as pembrolizumab (anti-PD-1) and ipilimumab (anti-CTLA-4), has revolutionized cancer treatment by restoring anti-tumor immunity and improving patient outcomes.
Figure 7. Structure of the Immune Checkpoint Receptor PD-1 and Its Ligands PD-L1/PD-L2. (Zak et al., 2017)
Hot-Spot Analysis for Drug Discovery Targeting Protein-Protein Interactions
Protein-protein interactions (PPIs) are central to many biological processes and represent a rich landscape for therapeutic intervention. However, targeting PPIs is often challenging due to their large and flat interfaces, which lack well-defined binding pockets typically found in enzyme or receptor active sites. Hot-spot analysis has emerged as a powerful strategy to address the challenge of targeting protein-protein interactions (PPIs), enabling the identification of key regions within PPI interfaces that are critical for binding and functional interactions. This approach has shown promise in developing inhibitors for traditionally "undruggable" targets in cancer, infectious diseases, and autoimmune disorders.
Hot spots are small, energetically significant regions within the protein interface that contribute disproportionately to the binding free energy. These regions often consist of residues with specific physicochemical properties, such as hydrophobicity or hydrogen bonding potential, that anchor the interaction and stabilize the PPI. Identifying these hot spots provides an opportunity to design small molecules, peptides or biologics that can selectively disrupt or modulate the interaction, offering potential therapeutic benefits.
Advanced techniques such as alanine scanning mutagenesis, molecular dynamics simulations, and computational docking are widely used to map and validate hot spots. Combining these methods with experimental approaches such as X-ray crystallography, nuclear magnetic resonance (NMR), or cryo-electron microscopy (cryo-EM) allows detailed characterization of the interaction interface.
By focusing on hot-spot regions, researchers can develop highly specific and potent inhibitors of PPIs, transforming traditionally "undruggable" targets into viable drug discovery opportunities in fields such as oncology, immunology, and infectious diseases.
Figure 8. Hot-spot residues and pathological mutations in the interaction between Type I TGF-β receptor with FKBP12. The x-ray structure of the complex between Type I TGF-β receptor (surface) and FKBP12 (ribbon) is shown (PDB 1B6C). (A) Residues involving mutations annotated in Humsavar (pathological: T200I, S241L, D266Y; unclassified: N267H) are shown in blue or magenta. Predicted hot-spot residues by docking (using pyDock NIP values) are shown in red or magenta. Interestingly, N267 residue (from N267H mutation, annotated as unclassified) is predicted as hot-spot (magenta). (B) Detail of protein-protein interface. Residues involving mutations T200I, D266Y, and N267H are shown before (left) and after modeling mutation (right). (Rosell and Fernández-Recio, 2018)
An Overview of Hot Targets in Global Innovative Drugs
The global landscape of innovative drug discovery is shaped by a number of hot protein targets, each of which plays a pivotal role in fighting disease and advancing therapeutic solutions. Below are some of the most notable targets and their importance:
- Epidermal Growth Factor Receptor (EGFR): A well-established target in oncology, EGFR mutations are implicated in several cancers, including lung and colorectal cancer. Structural studies have elucidated binding interactions with tyrosine kinase inhibitors (TKIs), aiding in the development of next-generation therapeutics.
- Human Epidermal Growth Factor Receptor 2 (HER2): Overexpressed in certain breast and gastric cancers, HER2 is a key target for monoclonal antibodies and antibody-drug conjugates. Understanding its dimerization and activation mechanisms has been crucial for therapeutic advances.
- Insulin: An essential protein for glucose metabolism, insulin remains central to the treatment of diabetes. Structural studies have provided insight into insulin-receptor binding and the design of long-acting insulin analogues.
- CD3: This component of the T cell receptor complex is critical for modulating the immune response. Structural studies have supported the development of bispecific antibodies that target CD3 to direct T cells against tumor cells.
- Amyloid Beta (Aβ): A hallmark of Alzheimer's disease, Aβ aggregation is a target for therapeutic intervention. Structural studies have elucidated aggregation pathways and interactions with potential inhibitors.
- Calcium Channels: These ion channels regulate calcium influx and are essential for processes such as muscle contraction and neurotransmission. Structural studies have enabled the design of blockers for the treatment of cardiovascular and neurological diseases.
- Programmed Death-Ligand 1 (PD-L1): An immune checkpoint protein, PD-L1 interacts with PD-1 to suppress immune responses. Structural studies have facilitated the development of checkpoint inhibitors that restore anti-tumor immunity.
- CD19: Expressed on B cells, CD19 is a target for chimeric antigen receptor (CAR) T-cell therapies. Structural insights into CD19-antibody interactions have improved therapeutic efficacy and reduced off-target effects.
- COVID-19 Spike Glycoprotein: The spike protein of SARS-CoV-2 mediates viral entry and is the primary target for vaccines and neutralizing antibodies. Structural studies of the spike protein have guided the design of vaccines, such as mRNA vaccines, by identifying key epitopes that induce potent neutralizing antibodies, providing protection against COVID-19.
- Androgen Receptor (AR): A key player in prostate cancer, AR is a target for anti-androgens. Structural studies have revealed ligand binding mechanisms and resistance mutations, leading to the design of more effective therapies.
Implications for Drug Discovery and Biotechnology
- Structure-Based Drug Design: High-resolution structures guide the rational design of small molecules and biologics, reducing the time and cost of drug development.
- Biomolecular Engineering: Structural insights facilitate the engineering of proteins with enhanced stability, specificity, and catalytic efficiency for industrial applications.
- Therapeutic Targeting: Understanding protein interactions and conformational changes enables the development of allosteric modulators and multitarget drugs.
Challenges in Structural Studies of Hot Target Proteins
- Crystallization Difficulties: Many hot target proteins, particularly membrane proteins, are challenging to crystallize due to their hydrophobic nature and conformational flexibility.
- Dynamic Behavior: Capturing the dynamic conformational changes of proteins requires sophisticated techniques and data integration from multiple sources.
- Protein Production: The production of sufficient quantities of functional proteins, especially those requiring post-translational modifications, remains a bottleneck.
- Data Interpretation: High-resolution structures often require careful interpretation to relate static snapshots to dynamic biological processes.
Structural studies of hot protein targets have transformed our understanding of fundamental biological processes and catalyzed innovations in drug discovery and biotechnology. By integrating experimental techniques with computational approaches, researchers can address the challenges posed by dynamic and complex proteins. Future advances in technology and interdisciplinary collaboration promise to unlock new opportunities for therapeutic intervention and biomolecular engineering, reinforcing the central role of structural biology in addressing global health and industrial challenges.
Creative Biostructure offers a range of structural assay services including X-ray crystallography, cryo-electron microscopy (Cryo-EM), NMR spectroscopy services, as well as dedicated structure-based drug design (SBDD) services. Contact us today to learn more about how we can support your research project.
References
- Attwood MM, Fabbro D, Sokolov AV, Knapp S, Schiöth HB. Trends in kinase drug discovery: targets, indications and inhibitor design. Nat Rev Drug Discov. 2021;20(11):839-861.
- Botha S, Fromme P. Review of serial femtosecond crystallography including the COVID-19 pandemic impact and future outlook. Structure. 2023;31(11):1306-1319.
- Duan J, He XH, Li SJ, Xu HE. Cryo-electron microscopy for GPCR research and drug discovery in endocrinology and metabolism. Nat Rev Endocrinol. 2024;20(6):349-365.
- Emwas AH, Szczepski K, Poulson BG, et al. NMR as a "gold standard" method in drug design and discovery. Molecules. 2020;25(20):4597.
- Farooq Z, Howell LA, McCormick PJ. Probing GPCR dimerization using peptides. Front Endocrinol. 2022;13.
- Jumper J, Evans R, Pritzel A, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596(7873):583-589.
- Rosell M, Fernández-Recio J. Hot-spot analysis for drug discovery targeting protein-protein interactions. Expert Opinion on Drug Discovery. 2018;13(4):327-338.
- Zak KM, Grudnik P, Magiera K, Dömling A, Dubin G, Holak TA. Structural biology of the immune checkpoint receptor PD-1 and its ligands PD-l1/PD-l2. Structure. 2017;25(8):1163-1174.
- Zubcevic L, Herzik MA, Chung BC, Liu Z, Lander GC, Lee SY. Cryo-electron microscopy structure of the TRPV2 ion channel. Nat Struct Mol Biol. 2016;23(2):180-186.