Fact-checked by Grok 2 weeks ago

Structural biology

Structural biology is a multidisciplinary field at the intersection of , , and physics that focuses on determining the three-dimensional atomic structures of biological macromolecules, such as proteins, nucleic acids, and their complexes, to elucidate their functions, interactions, and roles in cellular processes. This discipline emerged in the mid-20th century, building on foundational advances in from the early 1900s, and has revolutionized our understanding of the molecular machinery of life by revealing how structural features enable biological activity at the atomic level. Key techniques in structural biology include , which provides high-resolution structures of crystallized macromolecules and accounts for the majority of entries in the (PDB); (NMR) spectroscopy, ideal for studying dynamic structures in solution; and cryo-electron microscopy (cryo-EM), which excels at visualizing large, flexible complexes in near-native states. Recent integrative approaches combine these methods with computational modeling, such as for structure prediction, to address complex systems like protein networks in cellular contexts, overcoming limitations of individual techniques. The field's historical development traces back to 1912, when William and Lawrence Bragg pioneered X-ray crystallography to resolve atomic arrangements in simple crystals, laying the groundwork for applying these methods to biomolecules. By the 1950s, landmark achievements like the determination of the DNA double helix structure by Watson, Crick, Franklin, and Wilkins marked the birth of structural biology as a distinct discipline, shifting focus from sequence to structure in understanding heredity and protein function. Over the subsequent decades, the integration of NMR in the 1970s–1980s for solution-state analysis and the cryo-EM revolution in the 2010s, achieving resolutions below 3 Å, have expanded the scope to encompass dynamic and transient states previously inaccessible. As of November 2025, with over 240,000 structures deposited in the PDB, structural biology informs drug discovery, biotechnology, and synthetic biology by providing atomic insights into disease mechanisms and enzyme catalysis. Beyond core techniques, emerging hybrid methods like cross-linking mass spectrometry (XL-MS) and small-angle X-ray scattering (SAXS) enable the study of macromolecular assemblies in solution and , capturing interactions that govern cellular organization. Cryo-electron (cryo-ET), a cutting-edge extension of cryo-EM, visualizes entire cellular environments at nanometer resolution, bridging the gap between isolated structures and . These advances underscore structural biology's pivotal role in addressing , such as modeling entire proteomes or designing therapeutics for complex diseases like cancer and neurodegeneration.

Overview

Definition and Scope

Structural biology is a multidisciplinary field at the intersection of , , and physics that focuses on elucidating the three-dimensional structures and of biological macromolecules, such as proteins, nucleic acids, , and their complexes, through the application of physical and computational methods. This field seeks to reveal the atomic-level architecture that governs molecular behavior, providing foundational insights into how these components assemble and interact within living systems. The scope of structural biology extends across a wide range of spatial scales, from atomic resolutions on the order of angstroms (typically Å) to the organization of large macromolecular assemblies. It encompasses the hierarchical levels of molecular organization, progressing from the primary sequence of in proteins or in nucleic acids, through secondary structures like alpha-helices and beta-sheets, to folds and assemblies of multi-subunit complexes. This comprehensive approach highlights the intricate relationships between sequence, structure, and overall molecular function. Central to the field are key biological macromolecules, including proteins—such as enzymes that catalyze reactions and receptors that mediate signaling—and nucleic acids like DNA and RNA, which store and transmit genetic information. The study emphasizes their interactions, including how lipids contribute to membrane-embedded complexes and how these elements form functional assemblies essential for cellular processes. Unlike related disciplines, structural biology prioritizes the development and application of structure-determination techniques over functional assays, as seen in enzymology, or molecular synthesis, as emphasized in aspects of biochemistry. This distinction underscores its role in providing precise spatial models that complement but do not directly assay biochemical activities or synthetic pathways.

Importance in Biology and Medicine

Structural biology plays a pivotal role in elucidating how the three-dimensional architecture of biomolecules governs their biological , providing foundational insights into cellular processes. The principle that determines is exemplified in enzymes, where the precise shape of active sites enables substrate binding and catalysis, as seen in the catalytic mechanisms of serine proteases. Similarly, in receptors, binding pockets dictate ligand specificity and , influencing pathways from responses to immune activation. These revelations have deepened understanding of molecular interactions essential for life, such as protein-nucleic acid complexes in . In , structural biology has revolutionized rational by enabling the visualization of target-ligand interactions at atomic , accelerating the development of therapeutics. A example is the design of HIV-1 protease inhibitors, where crystal structures of the enzyme-inhibitor complexes guided the creation of drugs like , transforming AIDS treatment from symptomatic management to viral suppression. This approach has extended to other diseases, including cancer and viral infections, by identifying exploitable structural vulnerabilities in proteins. Beyond direct , structural insights inform diagnostics and , such as tailoring inhibitors to patient-specific mutations. The field's broader impacts span , disease pathology, and emerging technologies. In evolution, conserved protein folds across species—such as the in diverse enzymes—highlight structural stability as a driver of functional adaptation over billions of years, underscoring common ancestry. In diseases like Alzheimer's, structural studies of misfolded proteins, including amyloid-beta aggregates and tau tangles, reveal mechanisms of and aggregation propagation, guiding therapeutic strategies to prevent misfolding. Structural biology also underpins , where engineered protein structures enable novel biocatalysts and for sustainable manufacturing. Societally, these advances have fueled growth; notable recognitions include the Nobel Prize in Chemistry for ribosome structures, which illuminated protein and targeting, the 2017 Nobel Prize in Chemistry for the development of cryo-electron , and the 2024 Nobel Prize in Chemistry for computational and structure prediction.

History

Early Foundations (Pre-1950s)

The foundations of structural biology emerged from early observations of biological materials and the development of conceptual frameworks linking molecular architecture to function, predating the sophisticated biophysical techniques of the mid-20th century. In 1665, published , in which he described his microscopic examinations of cork slices, revealing box-like compartments that he termed "cells" due to their resemblance to monastic chambers; this marked the first documented visualization of cellular structures, laying groundwork for understanding at a . Hooke's work with compound microscopes highlighted the potential of optical tools to reveal subvisible features of living matter, influencing subsequent generations of biologists. Conceptual advances in the late further tied structure to biological activity, particularly through studies of . In 1877, German physiologist Wilhelm Kühne coined the term "" (from en zymē, meaning "in ") to describe proteinaceous catalysts like and , explicitly connecting their chemical function to potential structural properties without bacterial involvement. This idea shifted focus from vitalistic views to mechanistic ones, positing that enzymes' specificity arose from inherent molecular forms, a notion that would underpin later structural investigations. The early 20th century introduced physical methods to probe molecular structures, with X-ray diffraction emerging as a pivotal tool. In 1912, demonstrated that crystals could diffract X-rays, revealing their wave nature and enabling the study of atomic arrangements in ordered materials; this discovery, confirmed through experiments with zinc blende and rock salt, earned Laue the 1914 and opened avenues for analyzing biological crystals. Building on this, British physicist William Astbury applied X-ray diffraction to protein fibers in the 1930s, producing patterns from stretched and unstretched wool and hair that distinguished "alpha" (compact) and "beta" (extended) forms, suggesting recurring structural motifs in fibrous proteins like . Biochemical progress complemented these physical insights, emphasizing sequence as a structural . During the 1940s, developed methods to determine sequences in proteins, beginning with insulin; by isolating and identifying end groups like phenylisothiocyanate derivatives, he established that proteins possessed defined linear orders of residues, challenging views of them as amorphous colloids. This work, culminating in insulin's elucidation, provided essential data for modeling three-dimensional folds. These pre-1950s efforts collectively established the intellectual and methodological bedrock for resolving atomic-level structures in the ensuing decades.

Key Developments (1950s–2000)

In 1951, chemist , drawing on bond angle constraints and data, proposed the alpha-helix—a right-handed coil stabilized by hydrogen bonds—and the beta-sheet—pleated strands linked by interchain bonds—as stable polypeptide configurations, predicting their prevalence in proteins. Influential researchers like advanced applications to nucleic acids in the early 1950s; her 1951-1952 studies at produced high-resolution fiber patterns of DNA, revealing helical densities that informed subsequent models. The 1950s marked a pivotal era in structural biology with the elucidation of the DNA double helix structure by and , building on X-ray diffraction data from and , which revealed the molecule's antiparallel strands and base-pairing mechanism essential for genetic replication. This breakthrough, confirmed through model-building and fiber diffraction patterns, transformed understanding of heredity and laid the groundwork for . Shortly thereafter, in 1959, John Kendrew's team achieved the first three-dimensional structure of a protein, sperm whale , at low resolution (approximately 6 ) using , later refined to 2 in 1960, unveiling the polypeptide chain's irregular folding around the group and challenging prior assumptions of highly ordered protein architectures. In 1960, extended these advances by determining the structure of at 5.5 Å resolution, identifying its arrangement of four subunits and the positioning of groups, which provided initial insights into oxygen-binding . These milestones, recognized with the 1962 awarded to Kendrew and Perutz, established atomic-level visualization of biomolecules as feasible and spurred global efforts in determination. Parallel to these developments, (NMR) spectroscopy emerged as a complementary technique for studying proteins in solution during the 1960s, with the first high-resolution spectra of proteins like reported, enabling analysis of local environments without . By the , NMR studies had begun to reveal details of structures, such as tertiary base-pairing interactions in transfer RNA (tRNA) through , complementing crystallographic data and highlighting dynamic aspects of biomolecular folding. Key milestones in the 1980s and 1990s included partial structural determinations of ribosome components, with and Heinz-Günther Wittmann obtaining the first crystals of ribosomal subunits in 1980, followed by medium-resolution models in the 1990s that illuminated RNA-protein interactions central to protein synthesis. These efforts culminated in high-impact recognitions, such as the 2009 for ribosome structures, though foundational work spanned the late 20th century. Institutional advancements supported this progress, notably the establishment of the (PDB) in 1971 at , which centralized deposition and dissemination of atomic coordinates, starting with seven structures and growing to facilitate collaborative research. Concurrently, the advent of facilities in the 1970s, such as the Daresbury Laboratory's NINA ring and later storage rings like , provided intense X-ray beams that dramatically improved data quality and resolution for macromolecular crystallography.

Modern Advances (2000–Present)

The advent of cryo-electron microscopy (cryo-EM) in the 2010s marked a transformative era in structural biology, enabling high-resolution imaging of biomolecules in near-native states without the need for . Key innovations, including direct electron detectors and advanced image processing algorithms, propelled resolutions from an average of 15 Å in 2010 to approximately 6 Å by 2020, with many structures now routinely achieving sub-3 Å detail. This "resolution revolution" was recognized by the , awarded to , , and Richard Henderson for developing cryo-EM techniques that preserve hydrated samples in vitreous ice and reconstruct three-dimensional structures from noisy two-dimensional projections. Artificial intelligence (AI) has further revolutionized structure prediction, achieving accuracies rivaling experimental methods for previously intractable proteins. DeepMind's , first showcased at the CASP13 competition in 2018 and refined in AlphaFold 2 by 2020–2021, leverages on multiple alignments and geometric constraints to predict protein folds with near-atomic precision, often below 2 Å for many targets. This breakthrough addressed over 200 million protein without experimental structures, enabling rapid insights into disease-related proteins and drug targets. Complementing this, the Baker laboratory's RoseTTAFold, introduced in 2021, employs a three-track integrating , distance, and coordinate data, delivering comparable accuracy while being computationally accessible for broader use. These AI-driven breakthroughs were recognized by the 2024 , awarded to David Baker for computational protein design and to and John Jumper for . These tools have democratized structural biology, predicting structures for unsolved proteins and accelerating discoveries in areas like mechanisms and assemblies. High-throughput techniques and integrative approaches have expanded the scope of structural determination, particularly for dynamic or heterogeneous systems. Serial femtosecond crystallography (SFX), utilizing free-electron lasers (XFELs), emerged in the early 2010s to analyze microcrystals in a "diffraction-before-destruction" manner, yielding time-resolved snapshots of protein dynamics at without radiation damage. Hybrid methods combining cryo-EM, (NMR) spectroscopy, and computational modeling have become standard for resolving large complexes, such as ribosomes or membrane transporters, by integrating low-resolution envelopes with atomic models to capture conformational ensembles. These synergies have enabled studies of transient states, like intermediates, that elude single-technique approaches. The (PDB) has grown exponentially, surpassing 244,000 entries by 2025, reflecting the influx of cryo-EM and AI-generated models alongside traditional structures. Complementing this, the Electron Microscopy Data Bank (EMDB) has archived over 50,000 density maps by late 2025, facilitating validation and refinement of models from cryo-EM data. These databases underscore the field's shift toward scalable, integrative structural insights, supporting global research in and beyond.

Fundamental Concepts

Molecular Structures and Levels of Organization

Structural biology examines the of biological macromolecules, primarily proteins and nucleic acids, which determines their three-dimensional architecture. The primary structure of a protein consists of the linear of linked by bonds, serving as the foundational for higher-order folding. This dictates the potential for subsequent structural levels, as established by Christian Anfinsen's experiments showing that regains native activity upon renaturation from denatured states, implying primary structure sufficiency for folding. Secondary structure arises from local hydrogen bonding patterns along the polypeptide backbone, forming elements such as α-helices and β-sheets. In α-helices, the backbone carbonyl oxygen of residue i hydrogen-bonds to the amide hydrogen of residue i+4, creating a right-handed coil stabilized by 3.6 residues per turn. β-sheets involve hydrogen bonds between adjacent strands, either parallel or antiparallel, resulting in pleated structures that contribute to the overall scaffold. These motifs are constrained by the , which maps allowable φ (phi) and ψ (psi) dihedral angles based on steric hindrance; for instance, most residues favor regions corresponding to α-helices (φ ≈ -60°, ψ ≈ -45°) or β-sheets (φ ≈ -120°, ψ ≈ 120°), as derived from early analyses of known protein structures. Tertiary structure represents the global three-dimensional folding of a single polypeptide chain, driven by interactions between side chains, including hydrophobic effects that bury nonpolar residues in the core while exposing polar ones on the surface. Disulfide bonds between residues further stabilize this fold, particularly in extracellular proteins. structure involves the assembly of multiple polypeptide subunits into functional complexes, such as hemoglobin's tetrameric arrangement of two α and two β chains, held by non-covalent interactions. The folding process is conceptualized as a funnel-shaped energy landscape, where the native state lies at the minimum , guiding the chain through intermediate conformations with decreasing . Proteins exhibit diverse architectures, including globular forms like enzymes (e.g., lysozyme), which fold into compact, soluble shapes with hydrophobic cores, and fibrous forms like collagen, which form elongated triple helices from repeating Gly-X-Y sequences for structural support. Nucleic acids display analogous hierarchy: the primary structure is the nucleotide sequence, while secondary structure in DNA features the right-handed B-form double helix with Watson-Crick base pairing (A-T and G-C) and a pitch of 10.5 base pairs per turn in physiological conditions. RNA typically adopts the more compact A-form helix, with 11 base pairs per turn and a deeper major groove, facilitating interactions in ribosomal structures. DNA topology extends this organization through supercoiling, where underwinding or overwinding introduces torsional stress, quantified by linking number (Lk = Tw + Wr, with twist Tw and writhe Wr), influencing compaction and accessibility in chromatin. These static structures provide the scaffold for dynamic conformational changes explored elsewhere.

Dynamics and Conformational Changes

Biological macromolecules, particularly proteins, exist not as rigid structures but as dynamic ensembles of conformers—distinct spatial arrangements of atoms that interconvert through rotations around single bonds or larger rearrangements. These conformational changes are crucial for biological function, enabling processes such as ligand binding, catalysis, and . Static structures determined by techniques like provide snapshots of equilibrium states, serving as starting points for understanding motion. A key mechanism governing dynamics is allostery, where binding of a at one site modulates activity at a distant site through conformational shifts. The seminal Monod-Wyman-Changeux model describes this as a concerted transition between tense (T) and relaxed (R) states in oligomeric proteins, exemplified by hemoglobin's cooperative oxygen binding, where initial O₂ attachment stabilizes the high-affinity R state, facilitating subsequent bindings. In enzymes, the induced fit hypothesis posits that substrate binding induces reshaping for optimal , as proposed by Koshland, contrasting with rigid lock-and-key models and explaining specificity in reactions like hexokinase-glucose . Conformational dynamics span vast timescales, from picoseconds for bond vibrations and side-chain rotations to milliseconds for domain rearrangements like kinase lid opening, reflecting the entropy-enthalpy balance that drives folding and function. Protein folding pathways follow , which states that the native structure is thermodynamically determined by the sequence under physiological conditions, as demonstrated by ribonuclease refolding experiments showing spontaneous attainment of the lowest free-energy state. (IDPs), lacking fixed structures, populate diverse conformers and gain order upon binding partners, enabling regulatory roles in signaling, as seen in p53's flexible . These dynamics are probed using , which captures transient states by initiating changes with light pulses and monitoring spectral shifts. Techniques distinguish ensemble averages, like bulk tracking millisecond folding intermediates, from single-molecule views, such as revealing heterogeneous pathways in IDPs, providing insights into functional variability.

Experimental Techniques

X-ray Crystallography

determines the three-dimensional atomic structure of biological macromolecules, such as proteins and nucleic acids, by analyzing the patterns produced when X-rays interact with a of the . In a , molecules are arranged in a highly ordered, repeating , allowing X-rays to coherently from the electrons surrounding atoms. This scattering generates an pattern on a detector, which, when processed, yields information about interatomic distances and angles. The technique has been instrumental in elucidating over 199,000 protein structures deposited in the as of 2025, providing foundational insights into molecular function. The core principle of diffraction in is governed by , which specifies the conditions for constructive interference of the scattered from successive : n\lambda = 2d \sin[\theta](/page/Theta) Here, n is a positive integer representing the order of , \lambda is the of the incident (typically 0.7–1.5 for protein studies), d is the perpendicular distance between adjacent , and \theta is the angle between the incident beam and the . This ensures that only specific angles produce measurable intensity peaks, enabling the reconstruction of the distribution within the . The workflow for structure determination begins with protein crystallization, the most labor-intensive step, where purified macromolecules are screened against thousands of conditions involving buffers, salts, and precipitants to form ordered suitable for . These are often grown using vapor or hanging-drop methods, with sizes ranging from micrometers to millimeters. Once obtained, data are collected by exposing the crystal to an beam while rotating it on a ; modern experiments are predominantly performed at sources, which deliver brilliant, tunable beams that minimize and enable data acquisition from tiny crystals in seconds. The resulting images are indexed, integrated, and scaled to produce amplitudes. To obtain the full structure factors, phasing is required to determine the phase angles of the diffracted waves, as detectors measure only intensities (amplitudes squared). Common methods include multiple isomorphous replacement (), where heavy atoms like mercury or are introduced into isomorphous crystals to create phase differences, and multi-wavelength anomalous diffraction (MAD), which exploits wavelength-dependent anomalous scattering from atoms like (incorporated via substitution) at multiple energies near their . These techniques generate an electron density map via Fourier transformation, into which an initial atomic model is built using software like . Model refinement iteratively adjusts coordinates, lengths, and angles to optimize the fit between the model and the experimental , typically using programs such as REFMAC or phenix.refine. A key metric is the R-factor, which quantifies between observed (F_{obs}) and calculated (F_{calc}) structure factors: R = \frac{\sum ||F_{obs}| - |F_{calc}||}{\sum |F_{obs}|} For high-quality protein structures, the working R-factor is usually below 20%, with free R-factors (calculated on a subset of data withheld from refinement) around 22–25% to assess . Validation tools like MolProbity ensure geometric realism. X-ray crystallography excels at delivering atomic-resolution structures, often at ~1 Å, which resolve individual atoms, hydrogen bonds, and solvent molecules, facilitating detailed mechanistic interpretations. However, it requires well-diffracting crystals, which can be elusive for flexible or proteins, and captures only static conformations, potentially missing dynamic states relevant to . For non-crystallizable , it is sometimes complemented by cryo-electron microscopy. Pioneering applications include the 1959 determination of the structure at 2 resolution by , the first atomic model of a protein, which revealed its globular fold and heme-binding pocket, earning the 1962 . A contemporary milestone is the 2020 crystal structure of the receptor-binding domain bound to ACE2 at 2.45 resolution, which illuminated viral entry mechanisms and accelerated and therapeutic development.

Nuclear Magnetic Resonance (NMR) Spectroscopy

Nuclear Magnetic Resonance (NMR) spectroscopy is a technique that determines the three-dimensional structures and dynamics of biological macromolecules in solution by exploiting the magnetic properties of atomic nuclei. The fundamental principle relies on the of nuclear spins, such as those of hydrogen-1 (^1H), (^13C), and nitrogen-15 (^15N), in a strong external , which aligns the spins and allows detection of radiofrequency pulses at the Larmor frequency. Chemical shifts in the NMR spectrum arise from the local electronic environment surrounding each nucleus, providing information on the chemical identity and connectivity of atoms within the . Additionally, the (NOE) measures through-space dipolar interactions between nearby nuclei, yielding distance restraints typically limited to under 5 , which are crucial for reconstructing atomic-level structures. The workflow for NMR-based structure determination begins with sample preparation, where proteins are often uniformly or selectively labeled with stable isotopes like ^13C and ^15N to overcome the low natural abundance and enhance through multidimensional experiments. Spectra are acquired in multiple dimensions—such as COSY for scalar couplings, / triple-resonance experiments (e.g., HNCA) for sequential assignments, and NOESY for distance information—to resolve overlapping signals and assign resonances to specific atoms. Structure calculation then integrates these restraints, including NOE-derived distances and J-coupling-derived torsion angles, using computational methods like restrained or to generate an ensemble of conformers that satisfy the experimental data. A key strength of NMR spectroscopy lies in its ability to capture conformational dynamics and flexibility in , particularly for regions that are disordered or undergo motions on picosecond to millisecond timescales, which are often averaged out in crystalline states. It is particularly suited for studying smaller proteins up to approximately 50 kDa and complexes in near-native environments like micelles. However, limitations include spectral crowding in larger systems leading to overlap, the need for high sample concentrations (typically millimolar), and challenges in resolving long-range restraints beyond 5 . Seminal applications include the 1990s studies on , a 76-residue protein, where ^15N relaxation measurements revealed backbone dynamics, showing restricted motions in the structured core and higher flexibility in loops, linking dynamics to functional roles in ubiquitination. More recently, NMR has enabled structures of membrane proteins solubilized in micelles, such as the of , which forms a right-handed α-helical dimer with a -40° crossing angle, providing insights into homodimerization mechanisms essential for stability. These examples highlight NMR's role in elucidating solution-state behaviors inaccessible to other methods.

Cryo-Electron Microscopy (Cryo-EM)

Cryo-electron microscopy (cryo-EM) is a powerful imaging technique that visualizes biological macromolecules in their near-native, hydrated states by leveraging the scattering of an electron beam through frozen samples in a electron microscope. The core principle relies on the interaction of high-energy electrons with the sample, where elastic and events generate contrast in the resulting two-dimensional projections, enabling the capture of structural details without the need for or fixation. To preserve the native conformation and shell of biomolecules, samples are subjected to —a rapid freezing process that forms amorphous (vitreous) ice, preventing formation that could distort structures. This method, pioneered in the , allows imaging at cryogenic temperatures (around -180°C) to minimize beam-induced damage and maintain sample integrity. Two primary approaches underpin cryo-EM structural determination: single-particle analysis () and . In , thousands to millions of two-dimensional images of individual, randomly oriented particles are collected and computationally aligned to reconstruct a three-dimensional density map through iterative refinement and averaging, which enhances and reveals high-resolution features. , on the other hand, involves acquiring a series of tilted projections (typically from -60° to +60°) of a single specimen, followed by back-projection algorithms to generate a three-dimensional tomogram, particularly useful for studying macromolecular assemblies within their cellular context or thicker samples. Both methods exploit the phase contrast from defocus in the to visualize unstained, frozen-hydrated specimens, with being more common for purified complexes and tomography for applications. As of 2025, the contains over 245,000 total structures, with accounting for approximately 199,000, NMR around 14,600, and over 30,000, reflecting the rapid growth of EM methods. The workflow of cryo-EM begins with sample preparation, where purified biomolecules are applied to a perforated carbon , blotted to form a thin aqueous film, and rapidly frozen by plunging into liquid cooled by , achieving in milliseconds. Imaging occurs in a cryo-TEM equipped with direct electron detectors, which record high-frame-rate movies to correct for beam-induced motion and enable low-dose exposure (typically 20-50 electrons/Ų) to mitigate . Data processing involves motion correction, contrast transfer function (CTF) estimation, particle picking, two-dimensional classification to remove junk particles, and three-dimensional reconstruction using software such as RELION, which employs for alignment and refinement. Resolution is assessed via the (FSC) criterion, where a 0.143 cutoff indicates the point at which two independently refined maps correlate; by 2025, resolutions better than 4 Å are routine for many targets, with atomic-level details (<3 Å) achieved in numerous cases, exemplified by a 1.22 Å structure of apoferritin. A key strength of cryo-EM lies in its ability to study large macromolecular complexes, such as viruses and ribosomes, without requiring crystallization, making it ideal for heterogeneous or flexible assemblies that are recalcitrant to other techniques. For instance, it excels at resolving structures of icosahedral viruses up to megadalton sizes and multidomain enzymes in near-native states. However, limitations include radiation damage from the electron beam, which can alter sensitive bonds and cause bubbling in the ice, as well as challenges from sample heterogeneity, preferred orientations, and air-water interface adsorption, which may preclude high resolution for small (<100 kDa) or dynamic proteins. Cryo-EM structures can be integrated with X-ray crystallography data to build hybrid models for improved accuracy in validation or refinement. Significant milestones in cryo-EM include early low-resolution reconstructions of viruses in the 1990s, such as the poliovirus-receptor-membrane complex at approximately 32 Å, which demonstrated the technique's potential for studying viral entry mechanisms. The field advanced dramatically in the 2010s with near-atomic resolution structures of ; for example, in 2013, a 3.3 Å reconstruction of the bacterial 70S was obtained from over 30,000 particles, revealing detailed RNA-protein interactions and paving the way for broader adoption in . These achievements, driven by direct detectors and improved algorithms, transformed cryo-EM into a mainstream method for resolving complex biomolecular architectures.

Computational Approaches

Structure Prediction and Modeling

Structure prediction in structural biology involves computational techniques to determine the three-dimensional (3D) conformation of proteins and other biomolecules from their amino acid sequences or partial structural data, without relying on experimental determination. These methods are essential for understanding protein function, as the 3D structure dictates biological activity. Traditional approaches include , which builds models based on known structures of related proteins, and , which relies on physical principles to fold sequences from scratch. More recently, has revolutionized the field by achieving near-experimental accuracy for many targets. Homology modeling, also known as comparative modeling, exploits evolutionary conservation by aligning a target sequence to a template with known structure, typically identified through sequence similarity searches. The model is then constructed by copying coordinates from the template and refining variable regions via energy optimization. A widely used tool for this is , which employs satisfaction of spatial restraints derived from the alignment and stereochemical principles to generate atomic models. This method is most effective when sequence identity exceeds 30%, yielding models with root-mean-square deviation (RMSD) values below 2 Å from native structures, indicating high reliability for functional studies. Ab initio prediction, in contrast, does not require templates and uses physics-based algorithms to explore conformational space. Rosetta, a prominent suite, employs Monte Carlo sampling combined with a knowledge-based potential to assemble fragments and minimize energy, guided by principles like hydrophobic core formation and secondary structure propensities. Energy minimization in these methods often utilizes force fields such as , which compute the potential energy E as: E = \sum_{\text{bonds}} k_b (r - r_0)^2 + \sum_{\text{angles}} k_\theta (\theta - \theta_0)^2 + \sum_{\text{dihedrals}} k_\phi [1 + \cos(n\phi - \delta)] + \sum_{\text{non-bonded}} \left( \frac{A_{ij}}{r_{ij}^{12}} - \frac{B_{ij}}{r_{ij}^6} + \frac{q_i q_j}{\epsilon r_{ij}} \right) where terms account for bond lengths, angles, dihedrals, and van der Waals/electrostatic interactions, respectively, to identify low-energy conformations. This approach has succeeded for small proteins (<100 residues) but struggles with larger ones due to computational cost. Threading, or fold recognition, bridges homology and ab initio methods by evaluating how well a sequence "threads" onto known folds, even with low sequence similarity, using scoring functions that assess compatibility between sequence and structure. This technique identifies distant homologs by optimizing alignments that maximize burial of hydrophobic residues and minimize steric clashes. Protein threading has been pivotal in assigning folds to orphan sequences, enhancing genome-wide structural annotations. Deep learning methods, exemplified by AlphaFold2, integrate sequence, evolutionary multiple sequence alignments, and structural templates into a neural network with attention mechanisms to predict residue-residue distances and angles directly. Trained on Protein Data Bank structures, AlphaFold2 achieved median backbone RMSDs of 0.96 Å in the Critical Assessment of Structure Prediction (CASP14), surpassing all prior methods and enabling accurate predictions for proteins without close homologs. The CASP competitions, held biennially since 1994, have benchmarked these advances, with AlphaFold2's success marking a paradigm shift from physics- to data-driven prediction. A subsequent advancement, AlphaFold3, released in 2024, extends these capabilities to predict the structures of biomolecular complexes, including interactions between proteins, DNA, RNA, ligands, and ions, using a diffusion-based architecture for joint structure prediction. Practical impacts include the , released in 2021, which provides predicted structures for over 200 million proteins across eukaryotes, bacteria, and archaea, covering nearly all known sequences and accelerating research in uncharted proteomes. De novo design, where structures are engineered without natural templates, was demonstrated by the in 2003—a 93-residue α/β fold created using and structure prediction, validated by at 1.2 Å RMSD to the design model. These tools complement experimental techniques like for structure validation.

Simulation and Dynamics Analysis

Simulation and dynamics analysis in structural biology employs computational methods to model the temporal evolution of molecular structures, capturing motions, interactions, and conformational transitions that static structures cannot reveal. These approaches integrate physical principles to simulate atomic trajectories, providing insights into biological processes at timescales ranging from picoseconds to microseconds. Starting from predicted or experimentally derived structures, simulations extend these models by incorporating dynamic behavior, enabling the study of flexibility and stability in biomolecules such as and . Molecular dynamics (MD) simulations form the cornerstone of these analyses, solving Newton's second law of motion, \mathbf{F} = m \mathbf{a}, where force \mathbf{F} on each atom is derived from a potential energy function, mass m is the atomic mass, and acceleration \mathbf{a} determines velocity and position updates. These equations are numerically integrated using algorithms like Verlet or leap-frog, typically with timesteps of 1-2 femtoseconds to maintain energy conservation. Force fields, such as AMBER, parameterize the potential energy as a sum of bonded (bonds, angles, dihedrals) and non-bonded (van der Waals, electrostatic) terms, enabling accurate representation of intramolecular and intermolecular interactions in biomolecular systems. Popular software like GROMACS implements these simulations efficiently, supporting parallel computing for large-scale studies of protein folding and enzyme mechanisms. Monte Carlo (MC) sampling complements MD by generating configurations through random moves accepted or rejected via the Metropolis criterion, which is particularly useful for exploring conformational spaces at equilibrium without explicit time evolution. Enhanced sampling techniques, such as replica-exchange MD (REMD), address ergodicity limitations by running parallel simulations at different temperatures and periodically exchanging configurations to overcome energy barriers, thus improving convergence in free energy calculations. These methods illuminate key applications in structural biology, including the elucidation of ligand binding pathways, where MD trajectories reveal transient intermediates and binding affinities along free energy landscapes governed by \Delta G = -RT \ln K, with \Delta G as the free energy difference, R the gas constant, T temperature, and K the equilibrium constant. In protein-ligand docking, tools like employ genetic algorithms combined with empirical scoring functions to predict binding poses, often refined by subsequent MD to assess dynamic stability and induced fit effects. To bridge timescales inaccessible to all-atom MD (limited to nanoseconds without enhancements), coarse-grained (CG) models reduce resolution by grouping atoms into beads, accelerating simulations to microseconds or longer while preserving essential dynamics, as in studies of membrane protein assembly or large-scale conformational changes. Recent advances have dramatically extended simulation capabilities, with GPU-accelerated MD enabling routine access to microsecond timescales on consumer hardware, as demonstrated by optimized implementations in AMBER and GROMACS that leverage parallel processing for systems exceeding 100,000 atoms. Machine learning potentials, such as the ANI (Accurate Neural network for Interaction) model, further revolutionize dynamics analysis by approximating quantum mechanical energies and forces with near-density functional theory accuracy at force-field speeds, facilitating longer simulations of reactive events in enzymes or drug-target complexes without traditional force field approximations. These developments, building on foundational work from the 1970s, continue to integrate with experimental data for hybrid modeling, enhancing predictive power in structural biology.

Applications

Drug Discovery and Design

Structure-based drug discovery leverages atomic-level insights into target proteins to identify and refine small molecules that bind with high specificity and affinity. A core workflow begins with structure-based virtual screening, where libraries of compounds are docked computationally into the target's binding site to predict interactions, often scored by estimated binding free energies such as \Delta G values that quantify the thermodynamic favorability of complex formation. Promising hits are then optimized through iterative cycles of synthesis, biochemical testing, and structural validation, frequently using X-ray crystallography to visualize bound poses and guide modifications for improved potency and selectivity. This process accelerates lead identification by prioritizing molecules that fit the target's geometry, reducing the vast chemical space to tractable candidates. Seminal examples illustrate the impact of this approach. Imatinib (Gleevec), approved in 2001 for chronic myeloid leukemia, was designed using the crystal structure of the BCR-ABL kinase domain, enabling the development of an ATP-competitive inhibitor that precisely occupies the inactive conformation's binding pocket. Similarly, in response to the 2020 COVID-19 pandemic, structure-based design of inhibitors targeting the SARS-CoV-2 main protease (Mpro) yielded potent antivirals like (in Paxlovid), where crystallographic data of the protease's informed covalent warhead placement for rapid blockade. Key concepts in this field include fragment-based drug design, which screens low-molecular-weight fragments for weak binding detected via structural methods, then links or grows them into high-affinity leads while maintaining drug-like properties. Allosteric modulators represent another advance, binding sites distal from the orthosteric pocket to induce conformational changes that enhance or inhibit activity, as seen in structure-guided development of positive allosteric modulators for G-protein-coupled receptors. Structural biology also integrates with absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiling by modeling how ligand-protein interactions influence pharmacokinetics, such as predicting metabolic liabilities from exposed functional groups in bound poses. However, challenges persist, particularly target flexibility, where proteins adopt multiple conformations that complicate accurate and require modeling to capture dynamic landscapes.

Understanding Biological Processes

Structural biology provides critical insights into the mechanisms underlying cellular and organismal functions by revealing the atomic-level arrangements that drive dynamic biological processes. High-resolution structures obtained through techniques like , NMR spectroscopy, and cryo-EM enable researchers to visualize how proteins interact, undergo conformational changes, and facilitate key reactions in living systems. These visualizations not only explain the "how" of processes such as signaling, replication, and but also highlight the precise molecular interactions that ensure specificity and efficiency. In , G protein-coupled receptors (GPCRs) exemplify how structural insights illuminate activation cycles. GPCRs, the largest family of cell surface receptors, transduce extracellular signals into intracellular responses through conformational changes upon ligand binding. Cryo-EM and structures have shown that binding induces an outward tilt of transmembrane helix 6 (TM6), creating a binding pocket for heterotrimeric G proteins and enabling GDP-to-GTP exchange on the Gα subunit. This cycle, involving pre-coupled receptor-G protein assemblies, underscores the allosteric nature of GPCR signaling, where nucleotide-free intermediates stabilize effector interactions. DNA replication mechanisms are similarly decoded through polymerase structures, revealing fidelity checkpoints at the atomic scale. Replicative DNA polymerases, such as those in family A (e.g., Pol I) and family B (e.g., eukaryotic Pol δ), feature a hand-like with fingers, palm, and thumb domains that close upon correct dNTP binding, aligning the substrate for nucleotidyl transfer. Structural studies demonstrate an induced-fit mechanism where mismatched fail to trigger this closure, preventing errors during duplication and contributing to a polymerase error rate of approximately 10^{-6} per . Membrane transport processes, particularly those mediated by ATP-binding cassette (ABC) transporters, rely on coordinated domain movements visualized in high-resolution structures. ABC exporters, such as , consist of two transmembrane domains (TMDs) forming substrate pathways and two nucleotide-binding domains (NBDs) that dimerize upon ATP binding to drive alternating access. Crystal structures reveal how at the NBDs powers TMD rearrangements, alternating between inward- and outward-facing conformations to translocate diverse substrates across lipid bilayers, often against concentration gradients. Specific examples further illustrate these principles, such as the cryo-EM structures of (PSII) that have elucidated water oxidation in . In the 2010s, resolutions below 3 Å revealed the Mn4CaO5 (OEC) within PSII, showing how light-induced charge separation oxidizes the OEC through the S-state cycle, where sequential proton-coupled electron transfers split water into O2, protons, and electrons. These structures highlight calcium and roles in stabilizing the catalytic site, providing a blueprint for the enzyme's four-electron oxidation mechanism. The spliceosome's dynamics, captured in 2010s cryo-EM structures, demonstrate RNA processing intricacies. The human spliceosomal Bact complex, resolved at 3.4 Å, shows and U6 snRNPs forming a catalytic that positions intronic phosphates for branching and ligation, with protein factors like Prp8 stabilizing transient conformations. These snapshots reveal stepwise and disassembly, where ATP-dependent helicases remodel RNA-RNA and RNA-protein interactions to excise introns with near-perfect accuracy, linking structural changes to pre-mRNA maturation. Structural biology operates across scales, from atomic details of catalysis to cellular phenomena like viral entry. At the atomic level, enzyme active sites—such as the polymerase's catalytic aspartates—dictate reaction kinetics, while at the cellular scale, the SARS-CoV-2 spike protein's trimeric structure explains host cell invasion. Cryo-EM structures show the receptor-binding domain (RBD) engaging ACE2, triggering furin-mediated cleavage and TM fusion peptide exposure, facilitating membrane merger and genome delivery. This multiscale view connects individual protein motions to emergent biological outcomes. Evolutionary insights emerge from classifying protein folds in databases like and CATH, which trace functional conservation across . SCOP organizes domains into hierarchical classes, folds, superfamilies, and families based on structural similarity, revealing that ancient folds like the Rossmann fold underpin diverse enzymes from to . CATH complements this by integrating functional annotations, showing how fold repurposing—e.g., TIM barrels in unrelated hydrolases—drives evolutionary innovation while preserving core mechanisms. These classifications, encompassing over 100,000 domains, underscore structural determinism in function and adaptation.

Challenges and Future Directions

Limitations of Current Methods

Structural biology methods face significant experimental limitations that restrict their applicability to a subset of biomolecules. In , remains a primary bottleneck, as only a small of proteins—estimated at around 10%—successfully form suitable crystals for studies due to factors like protein flexibility and issues. Cryo-electron microscopy (cryo-EM) is constrained by from electron beams, which induces events that degrade sample integrity, introduce noise, and limit achievable resolution, particularly for beam-sensitive specimens. (NMR) spectroscopy struggles with protein size and dynamics; it is generally limited to molecules below 50 , as larger systems exhibit slow tumbling rates leading to broadened signals and overlap, while capturing fast dynamics requires specialized techniques that are not always feasible. Computational approaches in structural biology also encounter key challenges. Molecular dynamics (MD) simulations rely on force fields that often exhibit inaccuracies, such as over-stabilizing helical conformations or inadequately representing polarizable effects, leading to biased trajectories that deviate from experimental observations. A major hurdle is sampling rare events, like protein folding or conformational transitions, which occur on timescales exceeding milliseconds—far beyond the microsecond limits of routine all-atom MD simulations without enhanced sampling methods. Data-related issues further complicate structural determination. The phase problem in arises because diffraction experiments measure only amplitudes, requiring indirect methods like anomalous dispersion to infer s, which can fail for poor-quality crystals or weak signals. In computational structure prediction, models like can introduce biases, producing hallucinated or spurious structural features, especially in regions with low confidence scores, affecting reliability for novel sequences. Broader gaps persist across methods, including underrepresentation of certain protein classes and contexts. Membrane proteins, despite comprising 20-30% of proteomes, account for less than 10% of experimental structures in the (PDB), owing to difficulties in solubilization and stabilization outside native lipid environments. Transient protein complexes, characterized by weak or short-lived interactions, are challenging to capture at high resolution, as they resist stabilization for crystallographic or cryo-EM studies. Additionally, structures determined often differ from conditions due to the absence of cellular crowding, post-translational modifications, and molecular interactions, leading to discrepancies in conformation and dynamics. Handling large-scale data from techniques like cryo-electron tomography (cryo-ET) poses further challenges, including high computational demands for processing terabyte-scale datasets.

Emerging Technologies and Integrations

Recent advancements in structural biology are leveraging time-resolved cryo-electron microscopy (cryo-EM) to capture on to millisecond timescales, enabling the visualization of conformational changes in proteins. This technique integrates rapid sample mixing and advanced detectors to study transient states, such as intermediates. Complementing this, free-electron lasers (XFELs) facilitate ultrafast structural snapshots of biomolecular reactions, providing atomic-level insights into processes like or viral assembly on timescales. AI-hybrid approaches further enhance these methods, particularly through denoising algorithms that improve signal-to-noise ratios in low-dose cryo-EM images, allowing reconstruction of high-resolution structures from noisy datasets with improved accuracy. Integrative multi-modal strategies are fusing computational predictions with experimental data to refine structural models holistically. For instance, combining AlphaFold-predicted structures with (MD) simulations and (NMR) validation yields dynamic ensembles that account for flexibility and interactions in native environments. In situ structural biology, exemplified by cryo-electron (cryo-ET) of cellular contexts, maps protein architectures within intact s, revealing spatial organizations like nucleoprotein complexes without purification artifacts. Emerging AI tools are increasingly integrated with cryo-ET for automated in situ structure prediction as of 2025. These integrations, often supported by volume electron , correlate structural data across scales to elucidate context-dependent functions, such as assembly in organelles. Looking ahead, promises to revolutionize MD simulations by solving complex quantum mechanical interactions in pathways, potentially reducing computational times from years to hours for large systems. This could enable the design of enzymes with tailored active sites, as seen in AI-guided frameworks that generate novel folds for applications like carbon fixation catalysts. In , patient-specific structural predictions from genomic data, integrated with quantum-enhanced modeling, facilitate bespoke drug designs targeting individual mutations, as demonstrated in early inhibitors for variants. Such trends underscore the shift toward programmable biomaterials and precision therapeutics. Ethical considerations in these developments emphasize equitable access and responsible data stewardship. The (PDB) mandates deposition of structural data within one year of publication to promote , fostering global collaboration while protecting proprietary interests through embargo policies. However, disparities in AI tool access, particularly in low-resource settings, risk widening gaps in structural biology research, with calls for open-source initiatives to ensure inclusive benefits from tools like . Addressing these issues through international policies on and AI equity is crucial for sustainable progress.

References

  1. [1]
    Structural Biology: A Century-long Journey into an Unseen World - NIH
    Structural biology emerged in the 1950s using X-ray diffraction to study DNA and proteins, and has transformed our understanding of the molecular basis of life.
  2. [2]
    Advances in integrative structural biology: Towards understanding ...
    Dec 3, 2020 · The combination of multiple techniques, known as integrative structural biology, has provided insight into how large protein complexes interact ...
  3. [3]
    What is Structural Biology - Instruct-ERIC
    Structural Biology looks at proteins and nucleic acids at a molecular level, unlocking secrets of structure, function and interactions.Missing: scope | Show results with:scope
  4. [4]
    The Neurobiologist's Guide to Structural Biology: A Primer on Why ...
    X-ray crystallography has unsurpassed power to resolve the 3D configuration of amino acids within proteins and protein complexes and is the only method that can ...
  5. [5]
    Enzymes: An integrated view of structure, dynamics and function
    Jan 12, 2006 · In this review, we describe recent biochemical and theoretical investigations of internal protein dynamics linked to enzyme catalysis.<|separator|>
  6. [6]
    Structural biology of complement receptors - Frontiers
    In this review, we describe what is currently known about the structural biology of the complement receptors and their complexes with natural agonists and ...
  7. [7]
    Rational approach to AIDS drug design through structural biology
    This review presents the methods used to develop such drugs and discusses the remaining problems, such as the rapid emergence of drug resistance.
  8. [8]
    HIV-1 Protease: Structural Perspectives on Drug Resistance - PMC
    Antiviral inhibitors of HIV-1 protease are a notable success of structure-based drug design and have dramatically improved AIDS therapy.
  9. [9]
    RATIONAL APPROACH TO AIDS DRUG DESIGN THROUGH ...
    At this time, the principal targets are reverse transcriptase and protease, enzymes encoded by the human immunodeficiency virus. The introduction of protease ...
  10. [10]
    Conservation of protein structure over four billion years - PMC
    Little is known with certainty about the evolution of protein structures in general and the degree of protein structure conservation over planetary time scales ...
  11. [11]
    Correlation between evolutionary structural development ... - PNAS
    Evolution should have played important roles in determining folding mechanisms and structures of proteins. In this article we discuss how the folding ...
  12. [12]
    Protein misfolding and aggregation in Alzheimer's disease and Type ...
    Misfolded protein intermediates form large polymers of unwanted aggregates and are involved in the pathogenesis of many human diseases, including Alzheimer's ...
  13. [13]
    Protein misfolding in neurodegenerative diseases: implications and ...
    Mar 13, 2017 · A hallmark of neurodegenerative proteinopathies is the formation of misfolded protein aggregates that cause cellular toxicity and contribute to cellular ...
  14. [14]
    Structural materials meet synthetic biology in biomedical applications
    This review summarizes recent progress at the confluence of structural biomaterials research and synthetic biology, focusing on biomedical applications.
  15. [15]
    The Disruptive Impact of Structural Biology on Biopharmaceutical ...
    Mar 2, 2025 · Structural biology plays a crucial role in the development of biopharmaceuticals, providing atomic-level insights that drive the design and ...
  16. [16]
    The Nobel Prize in Chemistry 2009 - Popular information
    Some of them block the tunnel through which the growing proteins leave the ribosome, others prevent the formation of the peptide bond between amino acids.
  17. [17]
    Robert Hooke
    Hooke had discovered plant cells -- more precisely, what Hooke saw were the cell walls in cork tissue. In fact, it was Hooke who coined the term "cells": the ...
  18. [18]
    Looking Back: A Short History of the Discovery of Enzymes and How ...
    ... enzymes such as pepsin and invertase, but the term enzyme was only coined in 1877 by Wilhelm Kühne. The concept of catalysts, chemicals facilitating ...
  19. [19]
    Max von Laue – Facts - NobelPrize.org
    In 1912, Max von Laue came upon the idea that X-rays passing through crystals might create similar patterns. That is, that a crystal's structure would ...Missing: paper | Show results with:paper
  20. [20]
    William Thomas Astbury (1898–1961) | Embryo Project Encyclopedia
    Jun 3, 2019 · To do that, Astbury obtained X-ray diffraction patterns of wool by allowing X-rays to scatter off a crystal and to strike a film. That process ...
  21. [21]
    [PDF] Frederick Sanger - The chemistry of insulin - Nobel Prize
    In 1943 the basic principles of protein chemistry were firmly established. It was known that all proteins were built up from amino acid residues bound together ...Missing: 1940s | Show results with:1940s
  22. [22]
    [PDF] *(1951).
    HELICAL CONFIGURATIONS OF THE POLYPEPTIDE CHAIN. By LINUS PAULING, ROBERT B. COREY, AND H. R. BRANSON*. GATES AND CRELLIN LABORATORIES OF CHEMISTRY ...
  23. [23]
    Photograph 51, by Rosalind Franklin (1952)
    Dec 30, 2019 · On 6 May 1952, at King's College London in London, England, Rosalind Franklin photographed her fifty-first X-ray diffraction pattern of ...
  24. [24]
    A Structure for Deoxyribose Nucleic Acid - Nature
    The determination in 1953 of the structure of deoxyribonucleic acid (DNA), with its two entwined helices and paired organic bases, was a tour de force in ...
  25. [25]
    A Three-Dimensional Fourier Synthesis at 2 Å. Resolution - Nature
    Structure of Myoglobin: A Three-Dimensional Fourier Synthesis at 2 Å. Resolution. Nature 185, 422–427 (1960).
  26. [26]
    A Three-Dimensional Fourier Synthesis at 5.5-Å. Resolution ... - Nature
    Article; Published: 13 February 1960. Structure of Hæmoglobin: A Three-Dimensional Fourier Synthesis at 5.5-Å. Resolution, Obtained by X-Ray Analysis. M. F. ...
  27. [27]
    The Quiet Renaissance of Protein NMR - PMC - NIH
    The first NMR spectrum of a protein, that of ribonuclease A, was reported in 1957 by the lab of Martin Saunders. The introduction of radiofrequency-pulsed ...
  28. [28]
    NMR evidence for tertiary structure base pair in E. coli tRNA ... - Nature
    Dec 20, 1974 · The anomalous low field resonances in E. coli tRNA might be due to ring NH protons of S 4 U 8 in a tertiary structure base pair.
  29. [29]
    The ribosome returned - PMC - PubMed Central
    Jan 26, 2009 · The first ribosome crystals, reported by Yonath, Wittmann and colleagues in 1980 [2], diffracted poorly; but, as the years went by, crystals ...
  30. [30]
    History of Synchrotron Radiation Sources
    Synchrotron-radiation research had begun at Daresbury around 1970 with the addition of a beamline to the 5-GeV NINA electron synchrotron. When NINA shut down in ...
  31. [31]
    [PDF] THE DEVELOPMENT OF CRYO-ELECTRON MICROSCOPY
    Oct 4, 2017 · The Nobel Prize was awarded for developing cryo-electron microscopy for high-resolution structure determination of biomolecules in solution.
  32. [32]
    Cryo-Electron Microscopy Reaches Resolution Milestone - PMC
    Aug 18, 2020 · Since 2010, the average resolution of a cryo-EM structure has improved from 15 Å to about 6 Å, and it is increasingly common for cryo-EM to ...
  33. [33]
    Cryo-electron microscopy wins chemistry Nobel - Nature
    Oct 12, 2017 · Jacques Dubochet, Joachim Frank and Richard Henderson were awarded the prize on 4 October for their work in developing cryo-electron microscopy (cryo-EM).
  34. [34]
    Highly accurate protein structure prediction with AlphaFold - Nature
    Jul 15, 2021 · AlphaFold greatly improves the accuracy of structure prediction by incorporating novel neural network architectures and training procedures ...Missing: impact | Show results with:impact
  35. [35]
    Accurate prediction of protein structures and interactions using a ...
    Aug 19, 2021 · The method is available as a server at https://robetta.bakerlab.org (RoseTTAFold option), and the source code and model parameters are available ...
  36. [36]
    Review: Serial Femtosecond Crystallography: A Revolution in ...
    SFX relies on exceptionally brilliant, micro-focused X-ray pulses, which are femtoseconds in duration, to probe nano/micrometer sized crystals in a serial ...Crystal Detection And... · Sample Delivery Methods · Data Acquisition And Data...
  37. [37]
    Structures of biomolecular complexes by combination of NMR and ...
    Combination of cryoEM and NMR became an established hybrid approach in structural biology that significantly contributes to our understanding of functional ...
  38. [38]
    PDB Statistics: Overall Growth of Released Structures Per Year
    Year, Total Number of Entries Available, Number of Structures Released Annually. 2025, 244,730, 15,064. 2024, 229,666, 15,474. 2023, 214,192, 14,501.
  39. [39]
    EMDB < Home - EMBL-EBI
    The Electron Microscopy Data Bank (EMDB) is a public repository for ... As of 05 November 2025, EMDB contains 50977 entries (latest entries, trends).
  40. [40]
    The Shape and Structure of Proteins - Molecular Biology of the Cell
    Biologists distinguish four levels of organization in the structure of a protein. The amino acid sequence is known as the primary structure of the protein.
  41. [41]
    Protein Structure, Modelling and Applications - NCBI - NIH
    May 1, 2006 · Typically four levels are used and referred to as the primary, secondary, tertiary and quaternary structure of a protein. It is often useful ...
  42. [42]
    Biochemistry, Secondary Protein Structure - StatPearls - NCBI - NIH
    Many globular proteins have multiple alpha-helical portions separated by long stretches of non-helical regions. Some fibrous proteins, including alpha-keratin, ...Missing: examples | Show results with:examples
  43. [43]
    Ramachandran Plot - an overview | ScienceDirect Topics
    In theory, the allowed regions of the Ramachandran plot show which values of the Phi/Psi angles are possible for an amino acid, X, in a ala-X-ala tripeptide ( ...
  44. [44]
    Biochemistry, Primary Protein Structure - StatPearls - NCBI Bookshelf
    Oct 31, 2022 · Each protein at least contains a primary, secondary, and tertiary structure. Only some proteins have a quaternary structure as well.
  45. [45]
    Protein folding funnels: a kinetic approach to the sequence-structure ...
    Convergent kinetic pathways, or "folding funnels," guide folding to a unique, stable, native conformation. Solution of the probability flow equations is ...Missing: concept | Show results with:concept
  46. [46]
  47. [47]
    DNA: Alternative Conformations and Biology - NCBI - NIH
    This chapter describes the structures of DNA forms called alternative DNA conformations that are different from the canonical B-DNA helix.<|control11|><|separator|>
  48. [48]
    The importance of being supercoiled: how DNA mechanics regulate ...
    The DNA topology is described quantitatively by the twist of double helix and by the number of times the helix crosses over on itself (plectoneme). Plectonemic ...
  49. [49]
    Large-Scale Conformational Changes and Protein Function
    Functional conformational changes usually involve either large rigid-body motions of structural elements or more local unfolding, loop fluctuation transitions.Missing: seminal | Show results with:seminal
  50. [50]
    ON THE NATURE OF ALLOSTERIC TRANSITIONS: A PLAUSIBLE ...
    Authors. J MONOD, J WYMAN, J P CHANGEUX. PMID: 14343300; DOI: 10.1016/s0022-2836(65)80285-6. No abstract available. MeSH terms. Chemical Phenomena; Chemistry ...
  51. [51]
    Probing Structure and Reaction Dynamics of Proteins Using Time ...
    Dec 9, 2019 · Time-resolved (TR) resonance Raman (RR) is a particularly versatile tool to probe processes of proteins harboring cofactors with electronic transitions in the ...
  52. [52]
    Protein Dynamics from Time-Resolved UV Raman Spectroscopy - NIH
    UVRR spectroscopy is also useful in characterizing slow protein conformation changes. Lednev and coworkers have evaluated the mechanism of lysozyme fibril ...
  53. [53]
    Synchrotron Radiation as a Tool for Macromolecular X-Ray ...
    In this review, we analyze selected aspects of the impact of synchrotron radiation on structural biology. Synchrotron beamlines have been used to determine over ...
  54. [54]
    How cryo‐electron microscopy and X‐ray crystallography ... - NIH
    Principles of X‐Ray Crystallography and Cryo‐EM. X‐ray crystallography's foundation principle lies in Bragg's Law of X‐ray diffraction by crystals, i.e. by ...
  55. [55]
    Protein X-ray Crystallography: Principles of the Technique
    Mathematically, Bragg's law looks like this: nλ = 2d sinθ, where n is an integer, d is the interplanar distance in the crystal, and θ is the incident angle of ...
  56. [56]
    Introduction to phasing - PMC - NIH
    Nowadays, methods from small-molecule crystallography can be used to find the heavy-atom substructure and the phases for the whole protein can be bootstrapped ...
  57. [57]
    As MAD as can be - ScienceDirect.com
    The traditional method of phase determination is multiple isomorphous replacement (MIR) [3]. In this method, heavy metal compounds are bound to the protein in ...
  58. [58]
    Estimation of the quality of refined protein crystal structures - PMC
    Finally, the average R-factor difference for all structures at resolutions lower than 2.85 Å is much smaller than the value predicted by the trends using ...
  59. [59]
    Validation and Quality Assessment of X-ray Protein Structures
    As mentioned earlier, for protein structures, the R-factor typically ranges from 25% to 14%, while for small-molecule structures, it is around 4% to 5%. Ideally ...
  60. [60]
    Methods for Determining Atomic Structures - PDB-101
    X-ray crystallography can provide very detailed atomic information, showing every atom in a protein or nucleic acid along with atomic details of ligands ...
  61. [61]
    X-Ray Crystallography - an overview | ScienceDirect Topics
    However, X-ray crystallography does have limitations. It requires obtaining high-quality crystals, which can be challenging for some biomolecules, particularly ...
  62. [62]
    John Kendrew and myoglobin: Protein structure determination ... - NIH
    The essay reviews John Kendrew's pioneering work on the structure of myoglobin for which he shared the Nobel Prize for Chemistry in 1962.
  63. [63]
    Structure of the SARS-CoV-2 spike receptor-binding domain bound ...
    Mar 30, 2020 · We determined the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 bound to the cell receptor ACE2.
  64. [64]
    An Introduction to Biological NMR Spectroscopy - PMC
    Abstract. NMR spectroscopy is a powerful tool for biologists interested in the structure, dynamics, and interactions of biological macromolecules.
  65. [65]
    Structural Biology by NMR: Structure, Dynamics, and Interactions - NIH
    Sep 26, 2008 · Nuclear Magnetic Resonance (NMR) spectroscopy has emerged as the method of choice for studying both protein structure and dynamics in solution.
  66. [66]
  67. [67]
    Cryo-electron microscopy: A primer for the non-microscopist - PMC
    Single-particle imaging and electron tomography of individual molecular complexes also invoke the principle of averaging multiple images collected from frozen- ...
  68. [68]
    Cryo Electron Microscopy: Principle, Strengths, Limitations and ...
    May 24, 2024 · Cryo EM is an advanced imaging technique used to elucidate the three-dimensional (3D) structure of biological molecules and complexes at near-atomic resolution.
  69. [69]
    Challenges and opportunities in cryo-EM single-particle analysis
    Cryo-EM enables structure determination of monodisperse macromolecular assemblies imaged at cryogenic temperatures in a transmission electron microscope. Cryo- ...
  70. [70]
    Cryo-electron tomography: A long journey to the inner space of cells
    Jul 21, 2022 · Cryogenic electron tomography (cryo-ET) is the application of tomographic principles of data acquisition and reconstruction to frozen-hydrated biological ...
  71. [71]
    Cryo-electron tomography | Nature Methods
    Dec 29, 2016 · In a cryo-ET study, a biological sample—a cell, tissue, or organism—is flash frozen, thinned to an appropriate thickness, and then imaged using ...
  72. [72]
    Present and Emerging Methodologies in Cryo-EM Single-Particle ...
    Cryo-EM single-particle analysis uses frozen-hydrated macromolecules to visualize unrestrained targets, enabling high-resolution structural analysis of ...
  73. [73]
    Collection, pre-processing and on-the-fly analysis of data for high ...
    The use of cryo-EM to determine near-atomic-resolution structures of macromolecular complexes has grown dramatically in the past 5 years, led by improvements in ...
  74. [74]
    RELION: Implementation of a Bayesian approach to cryo-EM ...
    RELION implements a modified version of the adaptive expectation maximization algorithm that was proposed by Tagare et al. (2010). For each experimental image, ...
  75. [75]
    How cryo-EM is revolutionizing structural biology - ScienceDirect.com
    For many years, structure determination of biological macromolecules by cryo-electron microscopy (cryo-EM) was limited to large complexes or low-resolution ...Missing: Strengths limitations
  76. [76]
    Cryo-electron microscopy reconstruction of a poliovirus-receptor ...
    Here we report the three-dimensional structure of a poliovirus-receptor-membrane complex solved by cryo-electron microscopy (cryo-EM) at a resolution of 32Å.Missing: milestones | Show results with:milestones
  77. [77]
    Ribosome structures to near-atomic resolution from thirty ... - eLife
    Feb 19, 2013 · The reconstructions presented in this paper are of significantly higher resolution than any prokaryotic or eukaryotic ribosome structures that ...
  78. [78]
    Ribosome structures to near-atomic resolution from thirty thousand ...
    Feb 19, 2013 · The reconstructions presented in this paper are of significantly higher resolution than any prokaryotic or eukaryotic ribosome structures that ...
  79. [79]
    About MODELLER - Andrej Sali
    MODELLER is used for homology or comparative modeling of protein three-dimensional structures (1,2). The user provides an alignment of a sequence to be modeled.Download & Installation · Tutorial · Online manual · MODELLER NewsMissing: primary | Show results with:primary
  80. [80]
    Ab initio protein structure prediction of CASP III targets using ...
    Nov 8, 1999 · Ab Initio: Prediction Report. Full Access. Ab initio protein structure prediction of CASP III targets using ROSETTA. Kim T. Simons,. Kim T ...
  81. [81]
    Optimization of the Additive CHARMM All-Atom Protein Force Field ...
    The results indicate that the revised CHARMM 36 parameters represent an improved model for modeling and simulation studies of proteins.
  82. [82]
    Protein fold recognition by prediction-based threading - PubMed
    Jul 18, 1997 · In fold recognition by threading one takes the amino acid sequence of a protein and evaluates how well it fits into one of the known three-dimensional (3D) ...
  83. [83]
    Protein Structure Prediction Center
    In CASP14, about 2/3 of the 96 targets reached GDT_TS values greater than that, and so are considered competitive with experiment in backbone accuracy. ... The ...CASP16 · CASP13 · CASP_Commons · CASP15
  84. [84]
    AlphaFold Protein Structure Database: massively expanding the ...
    Nov 17, 2021 · The data set contains proteins with sequence lengths of 16–2700 and excludes isoforms and sequences with unknown or non-standard amino acids.
  85. [85]
    Design of a Novel Globular Protein Fold with Atomic-Level Accuracy
    Here, we used a general computational strategy that iterates between sequence design and structure prediction to design a 93-residue α/β protein called Top7 ...
  86. [86]
    Molecular Dynamics Simulations of Biomolecules - ACS Publications
    McCammon, J. A.; Gelin, B. R.; Karplus, M. Dynamics of Folded Proteins ... Journal of Chemical Information and Modeling 2025, 65 (15) , 8158-8167 ...Missing: seminal | Show results with:seminal
  87. [87]
    The Amber Biomolecular Simulation Programs - PMC - NIH
    Rather, we will focus on two more recent aspects of the Amber force fields: applications to carbohydrates, and the facilities for defining a model in which part ...
  88. [88]
    GROMACS: High performance molecular simulations through multi ...
    GROMACS is one of the most widely used open-source and free software codes in chemistry, used primarily for dynamical simulations of biomolecules.
  89. [89]
    Routine Microsecond Molecular Dynamics Simulations with AMBER ...
    The GPU accelerated version of AMBER can be used on all NVIDIA cards that support double precision in hardware, that is, those with hardware revision 1.3 or 2.0 ...<|separator|>
  90. [90]
    ANI-1: an extensible neural network potential with DFT accuracy at ...
    ANI is a new method designed with the intent of developing transferable neural network potentials that utilize a highly-modified version of the Behler and ...2. Theory And Neural Network... · 2.1. Neural Network... · 2.2. The Anakin-Me Model
  91. [91]
    The Process of Structure-Based Drug Design - ScienceDirect.com
    This review is intended to provide an overview of the process of structure-based drug design from the selection of a target to the generation and evaluation of ...
  92. [92]
    Structure and dynamics in drug discovery - Nature
    Nov 7, 2024 · Structure-based screening and docking methods are often able to sample accessible conformations of the small molecules being tested but, apart ...
  93. [93]
    Past, present, and future of Bcr-Abl inhibitors: from chemical ...
    Jun 20, 2018 · These structural developments led to the production of imatinib (STI571) (Fig. 3), the first known ATP competitor able to inhibit Bcr-Abl kinase ...
  94. [94]
    Structure-based design of antiviral drug candidates targeting the ...
    We designed and synthesized two lead compounds (11a and 11b) targeting M pro. Both exhibited excellent inhibitory activity and potent anti–SARS-CoV-2 infection ...
  95. [95]
    Structural biology in fragment-based drug design - PubMed
    Here we examine the recent literature looking at how structural biology has been used in a variety of successful fragment-screening applications.
  96. [96]
    Allosteric Modulators: An Emerging Concept in Drug Discovery - PMC
    Key Structures: The inventors reported the structures of 267 examples of formula (I) including the following compounds: graphic file with name ml-2014 ...
  97. [97]
    Structure based drug design and machine learning approaches for ...
    Sep 24, 2025 · The Discovery Studio Visualizer was used to examine atomic coordinates, types of interaction, and participation of residues in the interactions.
  98. [98]
    Target Flexibility: An Emerging Consideration in Drug Discovery and ...
    The essence of the problem for drug discovery is that for a flexible target it is not known in advance which conformation the target will adopt in response to ...
  99. [99]
    G protein-coupled receptors (GPCRs): advances in structures ...
    Apr 10, 2024 · Advances in the structural biology of GPCRs have revealed key information on ligand-receptor interactions, conformational changes, and signaling ...Missing: seminal | Show results with:seminal
  100. [100]
    GPCR Dynamics: Structures in Motion | Chemical Reviews
    Sep 13, 2016 · Structural change within a GPCR allows it to act as a molecular conduit, transmitting an extracellular signal across the cell membrane to ...Missing: seminal | Show results with:seminal
  101. [101]
    Structure and Mechanism of DNA Polymerase β | Biochemistry
    Apr 10, 2014 · Here, the authors review the current understanding of replication fidelity, with emphasis on structural and biochem. studies of DNA polymerases ...Biological Role · Structural Intermediates · DNA Polymerase Nucleotidyl... · Fidelity
  102. [102]
    Replicative DNA polymerases | Genome Biology | Full Text
    Feb 1, 2001 · Replicative DNA polymerases are essential for the replication of the genomes of all living organisms. On the basis of sequence similarities they can be ...Gene Organization And... · Characteristic Structural... · Sequence Features
  103. [103]
    Structure and mechanism of ABC transporters - PMC
    Feb 3, 2015 · ABC transporters are widespread in all forms of life and are characterized by two nucleotide-binding domains (NBD) and two transmembrane domains (TMDs).
  104. [104]
    High-resolution cryo-electron microscopy structure of photosystem II ...
    Photosystem II (PSII) is a photo-oxidoreductase that harnesses light energy to use water to make fuel. Water oxidation occurs at a metal cluster in the ...
  105. [105]
    The spliceosome: disorder and dynamics defined - PMC - NIH
    Feb 14, 2014 · Here we review recent advances in our understanding of spliceosome structural dynamics stemming from bioinformatics, deep sequencing, high ...
  106. [106]
    Cell entry mechanisms of SARS-CoV-2 | PNAS
    May 6, 2020 · A virus surface spike protein mediates SARS-CoV-2 entry into cells. To fulfill its function, SARS-CoV-2 spike binds to its receptor human ACE2 ( ...
  107. [107]
    The SCOP database in 2020: expanded classification of ... - NIH
    Nov 14, 2019 · The Structural Classification of Proteins (SCOP) database is a classification of protein domains organised according to their evolutionary and structural ...Missing: insights | Show results with:insights
  108. [108]
    CATH: Protein Structure Classification Database at UCL
    Sep 30, 2024 · CATH-Gene3D provides information on the evolutionary relationships of protein domains through sequence, structure and functional annotation data ...
  109. [109]
  110. [110]
    [PDF] Program.pdf - Brookhaven National Laboratory
    Oct 11, 2025 · We anticipate that this novel polymer-based microchip will enable time-resolved structural studies of a range of proteins at both XFEL and.
  111. [111]
    X-ray free electron lasers (XFELs) in time-resolved X-ray diffraction ...
    Nov 1, 2025 · The feasibility of time-resolved structural studies using pulsed X-ray radiation from a 3rd generation synchrotron is discussed and compared ...
  112. [112]
    Augmenting X-ray single-particle imaging reconstruction with self ...
    This work introduces an end-to-end self-supervised machine-learning method that simultaneously estimates particle orientations and reciprocal-space intensities.
  113. [113]
    AlphaFold 2 and NMR Spectroscopy: Partners to Understand ...
    AlphaFold 2 predicts protein structures, but NMR provides folding and dynamics info. They complement each other, with NMR addressing AlphaFold 2's limitations.
  114. [114]
    Integrating cellular electron microscopy with multimodal data to ...
    Feb 1, 2024 · In this Review, we detail the latest advancements in volume electron microscopy (vEM) and cryo-electron tomography (cryo-ET), which together can ...
  115. [115]
    Integrating cellular electron microscopy with multimodal data to ...
    Feb 1, 2024 · In this Review, we detail the latest advancements in volume electron microscopy (vEM) and cryo-electron tomography (cryo-ET), which together can visualize ...
  116. [116]
    Harnessing AI and Quantum Computing for Accelerated Drug ...
    AI-driven quantum computing also supports de novo drug design, generating novel compounds with desired chemical and biological properties. Additionally, quantum ...
  117. [117]
    De novo protein design—From new structures to programmable ...
    Feb 1, 2024 · De novo proteins could be engineered to sense new signals, integrate signals and perform logic, and precisely regulate downstream biological ...
  118. [118]
    Quantum-computing-enhanced algorithm unveils potential KRAS ...
    Jan 22, 2025 · This study highlights the promising intersection of quantum computing and drug discovery, marking a quantum computer's first experimental hit. ...<|separator|>
  119. [119]
    Journal policies: Structure - Cell Press
    Exceptions to these requirements can be made for individual datasets when sharing data is legally or ethically prohibited. ... Please also include a link to, or a ...Journal Policies · Authorship · Resource Availability<|control11|><|separator|>
  120. [120]
    AI-Driven Deep Learning Techniques in Protein Structure Prediction
    ... data sharing, inconsistencies and gaps in data still pose challenges. A ... Ethical concerns arise regarding equal access to AI-generated treatments.2.1. Homology Modeling · 3.1. Alphafold · 3.7. Ai Integration
  121. [121]
    Balancing ethical data sharing and open science for reproducible ...
    Apr 15, 2025 · Here we examine the balance between ethical and responsible data sharing and open science practices that are essential for reproducible research in biomedical ...Missing: PDB | Show results with:PDB