This article provides a complete methodological guide for ancestral protein resurrection, a powerful technique that combines computational phylogenetics with experimental biochemistry to reconstruct and characterize ancient proteins.
This article provides a complete methodological guide for ancestral protein resurrection, a powerful technique that combines computational phylogenetics with experimental biochemistry to reconstruct and characterize ancient proteins. Aimed at researchers, scientists, and drug development professionals, it covers the entire workflow from foundational principles and step-by-step laboratory protocols to advanced troubleshooting and validation strategies. The content explores how resurrected ancestral proteins serve as unique tools for understanding molecular evolution, engineering stable enzyme variants, and developing novel therapeutic scaffolds, with direct applications in biomedical research and drug discovery.
Ancestral Sequence Reconstruction (ASR) represents a powerful convergence of evolutionary biology and molecular biochemistry, enabling researchers to infer the sequences of ancient proteins and resurrect them in the laboratory for functional characterization. The field originated from the seminal work of Linus Pauling and Emile Zuckerkandl in 1963, who first proposed that comparing sequences of modern proteins within an evolutionary framework could mathematically infer ancestral sequences [1] [2] [3]. They envisioned this approach as the foundation for a new field they termed "Paleobiochemistry" [2]. Despite this groundbreaking insight, the technology and data required to implement their vision remained insufficient for several decades. The first successful examples of ancestral protein resurrection did not emerge until the 1990s, as sequence data accumulated in growing genetic databases [3]. Since then, advances in computational algorithms, statistical models, and gene synthesis technologies have transformed ASR into a robust tool for studying protein evolution, enabling resurrection of proteins dating back billions of years [1] [2] [4].
The core principle underlying ASR is that closely related species share similar DNA and protein sequences due to common descent [2]. By analyzing these relationships through phylogenetic trees, researchers can extrapolate backward to infer ancestral states. ASR does not claim to recreate the exact historical sequence that existed in ancient organisms, but rather produces a sequence that likely represents the functional characteristics of the ancestral protein, fitting within the "neutral network" model of protein evolution where multiple genotypically different but phenotypically similar sequences can coexist in a population [2]. This approach has revealed fundamental insights into evolutionary processes, ancient environments, and the structural determinants of protein function.
Modern ASR methodology follows a standardized four-step workflow that operationalizes Pauling and Zuckerkandl's original concept [1] [3]:
Step 1: Sequence Collection and Curation - Researchers first define a protein of interest and collect homologous sequences from diverse organisms using public databases. Careful selection of sequences that adequately represent the phylogenetic diversity of the protein family is crucial for accurate reconstruction.
Step 2: Multiple Sequence Alignment (MSA) - The collected sequences are aligned to establish positional homology, determining which sites descended from a common ancestral position. Alignment accuracy significantly impacts reconstruction quality, with tools like MAFFT and PRANK generally performing well [3].
Step 3: Phylogenetic Tree Construction - The MSA information is used to infer evolutionary relationships between sequences, constructing a phylogenetic tree that represents the branching process of diversification. Both maximum likelihood and Bayesian methods are commonly employed for this step.
Step 4: Ancestral Sequence Inference - Sequences at ancestral nodes are extrapolated backward along the inferred tree using statistical models. The marginal probability method is frequently used, calculating for each site at each ancestral node the relative probability of all possible ancestral states given the tree and MSA [1].
The statistical approaches for ancestral inference have evolved significantly, with three primary methods in use:
Maximum Parsimony (MP) - The earliest method used for ASR, MP infers the ancestral sequence that requires the minimum number of evolutionary changes to explain modern sequences [3]. While conceptually simple, it relies on an oversimplified evolutionary model and is rarely used in contemporary studies.
Maximum Likelihood (ML) - Currently the most widely used approach, ML determines ancestral states that maximize the posterior probability at each position given an explicit substitution model [1] [2] [3]. ML methods use substitution matrices (e.g., the LG matrix) that encode probabilities of different amino acid transitions based on known protein sequences [1].
Bayesian Methods - These approaches view ancestral reconstruction as a posterior probability distribution rather than a single "best estimate," explicitly accounting for uncertainty in trees, branch lengths, and substitution models through sampling [3] [5]. While computationally intensive, Bayesian methods can reduce biases toward overestimating protein stability that may occur with ML approaches [5].
The following workflow diagram illustrates the complete ASR process from sequence collection to protein characterization:
Table 1: Essential Software Tools for Ancestral Sequence Reconstruction
| Software Tool | Methodology | Key Features | Applications |
|---|---|---|---|
| PAML (Phylogenetic Analysis by Maximum Likelihood) | Maximum Likelihood | Implements codon and amino acid substitution models; includes CODEML for ancestral reconstruction | Widely used for ML-based ASR studies [3] |
| MEGA11 | Maximum Likelihood, Maximum Parsimony | User-friendly interface with comprehensive molecular evolution tools | Suitable for beginners and educational purposes [3] |
| HyPhy | Maximum Likelihood | Flexible platform for pattern-oriented analysis of genetic sequences | Detecting selection and evolutionary analysis [3] |
| RevBayes | Bayesian Inference | Modular platform for phylogenetic analysis using probabilistic graphical models | Incorporating uncertainty in ancestral reconstruction [3] |
| GRASP | Multiple Methods | Comprehensive framework for ancestral sequence reconstruction | Integrating various reconstruction approaches [3] |
ASR has provided unique insights into ancient habitats by characterizing resurrected ancestral proteins under different environmental conditions. A 2024 study reconstructed ancestral nucleoside diphosphate kinases (NDKs) and ribosomal proteins uS8s to investigate the pH of primordial environments [6]. The research followed this experimental protocol:
Protein Resurrection Protocol:
Key Findings: The reconstructed ancestral proteins displayed thermal stability profiles more similar to extant proteins from alkaliphilic bacteria than those from acidophilic or neutralophilic microorganisms, suggesting that common ancestors of bacterial and archaeal species thrived in alkaline environments [6].
ASR has emerged as a powerful protein engineering strategy, enabling the generation of novel enzymes with optimized properties. A compelling application involved the resurrection of Precambrian β-lactamase enzymes that were subsequently engineered to catalyze Kemp elimination, an anthropogenic reaction not found in nature [4]. The experimental approach included:
Laboratory Protocol:
Results: The ancient β-lactamase scaffolds demonstrated proficient Kemp eliminase activity when engineered with the new active site, while modern counterparts showed no activity. This functional difference was attributed to enhanced conformational flexibility in the ancestral proteins, which facilitated the emergence of new catalytic functions [4].
ASR has recently been applied to facilitate structural analysis of challenging multi-domain proteins. A 2025 study on modular polyketide synthases (PKSs) demonstrated how ASR could enable high-resolution structural determination [7]. The methodology included:
Experimental Workflow:
Significance: This approach demonstrated that ASR can generate stabilized protein variants that reduce conformational heterogeneity, enabling structural elucidation of complex multi-domain proteins that are otherwise refractory to high-resolution structural analysis [7].
Successful implementation of ASR requires specialized reagents and materials throughout the computational and experimental workflow:
Table 2: Essential Research Reagents for Ancestral Protein Resurrection
| Category | Specific Reagents/Materials | Function/Application |
|---|---|---|
| Computational Tools | PAML, MEGA11, HyPhy software licenses | Phylogenetic analysis and ancestral sequence inference |
| Gene Synthesis | Custom DNA synthesis services | Generation of ancestral gene sequences for laboratory expression |
| Expression Systems | E. coli expression strains (BL21, Rosetta), cell culture media | Heterologous expression of ancestral proteins |
| Purification Materials | Affinity chromatography resins (Ni-NTA, Glutathione Sepharose), size exclusion columns, imidazole, reducing agents | Purification of recombinant ancestral proteins |
| Characterization Reagents | Circular dichroism spectroscopy buffers, fluorescent dyes (SYPRO Orange), substrate analogs | Biophysical and functional characterization of resurrected proteins |
| Stabilization Additives | Glycerol, various salts, protease inhibitor cocktails | Maintaining protein stability during experimental analyses |
A critical consideration in ASR is the accuracy of reconstructed sequences and potential systematic biases. Computational studies using simulated protein evolution have revealed that:
The phylogenetic tree topology and sequence alignment quality significantly impact reconstruction accuracy. While uncertainty in phylogenetic trees has relatively minor effects on ASR robustness [3], careful selection of alignment methods and evolutionary models is essential. Model selection studies indicate that the best-fitting substitution model yields the most accurate reconstructions [3].
Robust ASR studies incorporate multiple validation approaches:
The following diagram illustrates the key considerations for ensuring reconstruction accuracy:
The journey from Pauling and Zuckerkandl's theoretical proposal to contemporary ASR laboratory protocols represents a remarkable synthesis of evolutionary theory, computational biology, and experimental biochemistry. Modern ASR has matured into a robust methodology that not only provides insights into fundamental evolutionary processes but also offers practical applications in protein engineering and drug development. The unique properties of resurrected ancestral proteinsâincluding enhanced thermostability, conformational flexibility, and catalytic promiscuityâmake them particularly valuable scaffolds for engineering novel functions [1] [4].
Future advancements in ASR will likely focus on refining evolutionary models, incorporating structural constraints into reconstruction algorithms, and developing more sophisticated experimental frameworks for characterizing ancestral proteins in cellular contexts. As sequence databases continue to expand and computational methods become increasingly sophisticated, ASR promises to yield ever-deeper insights into protein evolution while generating uniquely valuable biocatalysts and therapeutic proteins with applications across biotechnology and medicine. The continued integration of ASR with structural biology techniques, as demonstrated in recent cryo-EM studies [7], particularly highlights the growing potential of this approach to overcome long-standing challenges in molecular biology.
A protein's sequence encodes its conformational energy landscape, which determines the ensemble of structures it can adopt and, ultimately, its biological function [1]. The evolution of new protein functions is therefore fundamentally linked to how mutations alter this underlying energy landscape [1]. This landscape can be visualized as a funnel, where a wide top represents a high-energy, unfolded state, and the narrow bottom represents the low-energy, native folded state [8]. Evolution navigates this landscape, with amino acid substitutions tuning the relative stabilities of different conformations to create new functional properties [1].
Ancestral Sequence Reconstruction (ASR) has emerged as a powerful tool for studying this process. ASR uses phylogenetic models on modern protein sequences to infer the sequences of ancient proteins, which can then be synthesized and characterized in the laboratory [1] [9]. This approach provides a unique window into historical evolutionary events, allowing researchers to identify key mutations and correlate them with changes in energy landscapes and the emergence of novel functions such as new enzymatic activities, altered binding specificity, or changed oligomeric states [1].
The process of ASR follows a structured, multi-stage workflow, from sequence collection to the experimental characterization of resurrected proteins.
The core computational protocol for ASR consists of four main steps [1]:
The underlying statistical models assume that sequences evolve by a branching process, with sites evolving independently according to probabilities defined by an amino acid substitution matrix (e.g., the LG matrix) [1]. The result is a set of probabilistic ancestral sequences, often with posterior probabilities assigned to each reconstructed residue.
The following diagram illustrates the complete Ancestral Sequence Reconstruction workflow, from initial bioinformatics to experimental characterization:
Successful ASR relies on a suite of bioinformatic and experimental resources. The table below details key reagents and their functions in a typical ASR study.
Table 1: Essential Research Reagent Solutions for ASR Studies
| Resource Category | Specific Tool / Resource | Function in ASR Workflow |
|---|---|---|
| Sequence Databases | UniProtKB, InterPro [10] | Provides comprehensive, annotated protein sequences for homology searching and family classification. |
| MSA & Tree Building | Software (e.g., MAFFT, IQ-TREE) [1] | Constructs multiple sequence alignments and infers maximum likelihood phylogenetic trees. |
| Ancestral Reconstruction | Phylogenetics Software (e.g., HyPhy, PAML) [1] | Implements statistical models (e.g., marginal probability) to infer ancestral sequences. |
| Gene Synthesis | Commercial gene synthesis services | Materializes inferred ancestral sequences into DNA for laboratory expression. |
| Structure Prediction | AlphaFold, DeepSCFold [11] | Predicts 3D structures of resurrected ancestral proteins to form hypotheses about mechanism. |
Once ancestral proteins are resurrected, their biochemical and biophysical properties must be rigorously characterized to understand how their energy landscapes evolved.
This protocol outlines key experiments for profiling the stability, dynamics, and function of resurrected ancestral proteins.
Experimental studies on resurrected proteins have revealed several common trends, which are summarized in the table below.
Table 2: Experimentally Observed Properties of Resurrected Ancestral Proteins
| Protein Family | Key Experimental Findings | Implications for Energy Landscape |
|---|---|---|
| Various Precambrian Enzymes | Significant enhancement of thermodynamic stability (higher Tm and ÎG°) [9]. | A more rugged funnel with a deeper global minimum, potentially reflecting ancestral adaptation to a hotter environment. |
| β-lactamases, Esterases | Broader substrate promiscuity compared to modern descendants [9]. | A flatter, more flexible landscape near the native state, allowing access to more conformational sub-states. |
| Mamba Aminergic Toxins | Identification of key substitutions that modulate receptor specificity (e.g., AncTx1: α1A-AR selective; AncTx5: potent α2-AR inhibitor) [12]. | Mutations fine-tune the landscape to stabilize specific functional conformations for high-affinity binding. |
| GFP-like Proteins | Altered photoconversion pathways and spectral properties linked to historical hinge migrations [1]. | Mutations alter the energy barriers between conformational states, enabling new photophysical functions. |
The unique properties of ancestral proteins, particularly their stability and promiscuity, make them attractive starting points for protein engineering [9]. ASR can efficiently generate small but functionally diverse libraries that are enriched in stable, functional variants compared to random mutagenesis [12]. Successful applications include:
Recent advances in machine learning are providing powerful new tools for modeling and predicting protein energy landscapes. Machine-learned coarse-grained (CG) models are a particularly promising development [13]. These models are trained on large datasets of all-atom molecular dynamics simulations and can predict metastable states, fluctuations of disordered proteins, and relative folding free energies of mutants, while being orders of magnitude faster than all-atom simulations [13]. This enables the extrapolative simulation of new protein sequences, effectively allowing in silico exploration of evolutionary trajectories.
Furthermore, methods like DeepSCFold are improving the prediction of protein complex structures by leveraging sequence-derived structural complementarity, which is crucial for understanding how interactions evolve within a energy landscape framework [11].
Ancestral Sequence Reconstruction (ASR) has evolved from a theoretical concept into a powerful, versatile tool that bridges deep evolutionary history with cutting-edge biotechnology. By inferring the sequences of ancient proteins, researchers can now explore fundamental questions about molecular evolution while simultaneously engineering proteins with enhanced properties for modern applications. The technique's power lies in its ability to resurrect ancient biomolecules, providing a direct window into evolutionary processes that shaped modern protein functions and stability [1].
The foundational principle of ASR is that a protein's sequence determines its conformational energy landscape, which in turn governs its function. Understanding the evolution of new protein functions therefore requires understanding how historical mutations altered this energy landscape over time. ASR provides a unique window into these processes by allowing researchers to characterize the properties of ancient proteins and identify the specific substitutions that led to functional changes [1].
Table 1: Key Application Areas of Ancestral Sequence Reconstruction
| Application Area | Specific Use-Case | Research Impact |
|---|---|---|
| Structural Biology | Enabling high-resolution structure determination of challenging proteins [7] | Provides deeper mechanistic insight into modular polyketide synthases (PKS); enables cryo-EM single-particle analysis where native proteins fail |
| Protein Engineering | Creating stabilized enzyme variants and chimeric proteins [7] | Generates proteins with enhanced thermal stability, solubility, and broader substrate selectivity for industrial and therapeutic use |
| Evolutionary Biophysics | Dissecting the evolution of protein energy landscapes [1] | Reveals how historical mutations altered conformational landscapes to enable new functions like altered enzyme activity, binding specificity, and oligomerization |
| Molecular Evolution Studies | Testing hypotheses about early protein evolution [14] | Challenges long-standing assumptions about foundational protein motifs and provides insight into the complexity of early protein evolution |
| Drug Discovery | Developing ancestral biotin ligases for proximity labeling [1] | Creates research tools like AirID for proximal biotinylation, enabling study of protein interactions and cellular localization |
The standard ASR workflow involves four critical steps that transform contemporary sequence data into experimentally testable ancestral proteins [1]:
This protocol details the use of ASR to determine high-resolution structures of protein complexes that are recalcitrant to structural analysis in their native forms, as demonstrated with the FD-891 PKS loading module [7].
Experimental Workflow:
Key Technical Considerations:
This protocol utilizes ASR to trace the evolutionary pathway by which simple homomeric proteins evolved into specific heterocomplexes, revealing fundamental principles of protein-protein interactions and assembly [1].
Experimental Workflow:
Key Technical Considerations:
Table 2: Essential Research Reagents and Materials for ASR Experiments
| Reagent/Material | Function/Purpose | Examples/Notes |
|---|---|---|
| Sequence Databases | Source of homologous sequences for phylogenetic analysis | Public databases (e.g., GenBank, UniProt) with diverse taxonomic representation |
| Phylogenetic Software | Statistical inference of evolutionary trees and ancestral sequences | Packages implementing maximum likelihood (e.g., RAxML, IQ-TREE) or Bayesian methods (e.g., MrBayes) |
| Gene Synthesis Services | Production of codon-optimized ancestral genes for expression | Critical when ancestral sequences differ significantly from modern counterparts |
| Expression Vectors & Hosts | Production of recombinant ancestral proteins | Typically E. coli systems with appropriate promoters (e.g., T7, lac) for soluble protein expression |
| Chromatography Systems | Purification of ancestral proteins for functional and structural studies | Affinity (e.g., His-tag), ion exchange, and size-exclusion chromatography |
| Crystallization Screens | Initial conditions for growing protein crystals of ancestral variants | Commercial sparse matrix screens (e.g., Hampton Research) |
| Cryo-EM Infrastructure | High-resolution structure determination of large complexes | Requires access to transmission electron microscopes and grid preparation facilities |
| Stabilization Agents | Enhancing protein stability during storage and analysis | Glycerol, additives, and optimized buffer conditions for ancestral proteins |
| Activity Assay Reagents | Functional validation of resurrected ancestral proteins | Substrate analogs, cofactors, and detection systems specific to protein function |
| Relcovaptan-d6 | Relcovaptan-d6|Stable Isotope (unlabeled) | Relcovaptan-d6 is a deuterated, selective V1a vasopressin receptor antagonist for research. For Research Use Only. Not for human or veterinary use. |
| rac-Pregabalin-d4 | rac-Pregabalin-d4, MF:C₈H₁₃D₄NO₂, MW:163.25 | Chemical Reagent |
Ancestral Sequence Reconstruction (ASR) is a powerful phylogenetic method for inferring the sequences of ancient proteins, enabling the study of molecular evolution and the engineering of proteins with enhanced stability and novel functions [7]. This computational and experimental approach allows researchers to formulate and test hypotheses about the historical evolution of modern proteins. The resulting "resurrected" ancestral proteins provide a unique resource for structural biology, enzymology, and drug development, often exhibiting characteristics such as higher thermal stability and increased solubility compared to their contemporary counterparts [7] [15]. This application note provides a detailed protocol for the complete ASR workflow, from initial sequence collection to the final biochemical characterization of resurrected ancestral proteins, framed within the context of ancestral protein resurrection laboratory research.
The following sections outline the standard ASR protocol, with specific examples from recent research included to illustrate key steps and considerations.
Objective: To gather a comprehensive and diverse set of homologous sequences and generate a high-quality multiple sequence alignment (MSA).
Protocol:
Example from Literature: In a study on the GfsA loading module of a modular polyketide synthase, researchers constructed a phylogenetic tree from homologous sequences to infer an ancestral AT domain (AncAT), which was subsequently used to facilitate structural analysis [7].
Objective: To infer the evolutionary relationships among the homologous sequences, which provides the scaffold for ancestral reconstruction.
Protocol:
Objective: To compute the most probable amino acid sequences at specific ancestral nodes of interest on the phylogenetic tree.
Protocol:
Example from Literature: The Successor Sequence Predictor (SSP) method extends this principle by using linear regression on physicochemical properties of ancestral sequences to predict future evolutionary steps, demonstrating the predictive application of ASR [15].
Objective: To move from in silico predictions to in vitro study by producing the ancestral protein.
Protocol:
Example from Literature: In a study on caspases, ancestral sequences were codon-optimized for E. coli, cloned into a pET11a vector with a C-terminal Hisâ-tag, and the proteins were purified using established protocols [16].
Objective: To functionally validate the resurrected ancestral protein and understand its structural properties.
Protocol:
Example from Literature: The structural analysis of a chimeric KSQAncAT protein, where a native AT domain was replaced by an ancestrally reconstructed one, enabled the determination of a high-resolution crystal structure that was challenging to obtain with the modern protein, highlighting ASR's utility in structural biology [7].
The following workflow diagram integrates these major steps into a cohesive visual guide.
Successful implementation of ASR requires careful planning of both computational resources and laboratory reagents. The following tables summarize the core components.
Table 1: Essential Computational Tools for ASR
| Tool Category | Specific Software / Algorithm | Key Function in ASR Workflow |
|---|---|---|
| Sequence Homology Search | BLAST [15], HMMER | Identifies homologous sequences from databases. |
| Multiple Sequence Alignment | ClustalOmega [15], PROMALS3D [16], MAFFT | Aligns homologous sequences for phylogenetic analysis. |
| Phylogenetic Tree Building | IQ-TREE [16], RAxML [15] | Reconstructs evolutionary relationships using maximum likelihood. |
| Ancestral Sequence Inference | FastML [16], PAML (codeml), Lazarus [15] | Calculates the most probable ancestral sequences at tree nodes. |
| Structural Alignment & Analysis | SARST2 [18], Foldseek [18] | Rapidly compares and aligns protein structures against large databases. |
Table 2: Key Research Reagent Solutions for Ancestral Protein Resurrection
| Reagent / Material | Function in ASR Protocol | Example & Notes |
|---|---|---|
| Cloning Vector | Host for synthesized ancestral gene; enables protein expression. | pET11a vector for bacterial expression [16]. |
| Affinity Tag | Facilitates purification of the expressed recombinant protein. | C-terminal Hisâ-tag for immobilised metal affinity chromatography (IMAC) [16]. |
| Expression Host | Cellular system for producing the ancestral protein. | Escherichia coli (E. coli) strains (e.g., BL21). |
| Chromatography Resin | Purifies the protein based on specific properties. | Ni-NTA resin for purifying His-tagged proteins. |
| Crystallization Kits | Screens conditions for growing protein crystals for structural studies. | Commercial sparse matrix screens. |
The ASR workflow provides a robust and systematic framework for probing protein evolution and engineering highly functional proteins. The integration of sophisticated computational tools with standard molecular biology and biochemical techniques allows researchers to travel back in evolutionary time to resurrect and characterize ancient proteins. As demonstrated by recent studies on polyketide synthases, caspases, and elongation factors, the application of ASR can lead to fundamental mechanistic insights and provide unique molecular tools for structural biology and biotechnology [7] [16] [17]. By following the detailed protocols and utilizing the key reagents outlined in this application note, researchers can reliably incorporate ASR into their investigations on protein structure, function, and evolution.
Ancestral Sequence Reconstruction (ASR) is a computational and experimental technique for inferring the sequences of ancient proteins from the sequences of their modern descendants. Within the context of a laboratory protocol for ancestral protein resurrection, ASR provides the foundational gene sequences that are subsequently synthesized, expressed, and characterized in the lab. This protocol details the computational workflow for phylogenetic tree inference and ancestral sequence reconstruction, which serves as the critical first step in the protein resurrection pipeline. The resurrected ancestral proteins often exhibit unique biotechnological properties, such as enhanced stability and altered interaction patterns, making them valuable for drug development and industrial applications [19] [9].
The computational protocol is divided into two primary phases: (1) the inference of a robust phylogenetic tree and (2) the reconstruction of ancestral sequences at the nodes of this tree.
A reliable phylogeny is the cornerstone of accurate ASR. The following steps outline the general workflow.
2.1.1. Sequence Alignment and Input The process begins with a curated multiple sequence alignment. For closely related sequences with low divergence, such as specific β-lactamase clusters, coding DNA sequences rather than protein sequences may be used as input to capture all available evolutionary signal [20].
2.1.2. Tree Building Methods Different phylogenetic inference methods can be employed, often depending on the dataset and research question.
2.1.3. Model Selection A critical step is selecting an appropriate model of sequence evolution. A common and widely used model is the Generalized Time Reversible (GTR) model with Gamma-distributed rate variation (G) and a proportion of invariant sites (I). The alignment can be partitioned by codon position to allow for different evolutionary rates at first, second, and third codon positions [20].
2.1.4. Rooting the Tree Phylogenetic trees are typically rooted using an outgroup, which is a sequence or group of sequences known to be closely related but outside the clade of interest. For instance, SHV-1 coding DNA sequences can be used as an outgroup for the TEM β-lactamase cluster [20].
Once a reliable phylogeny is established, ancestral states can be inferred at its nodes.
2.2.1. Reconstruction Algorithm Ancestral sequences are typically inferred by maximum likelihood using the same nucleotide or amino acid substitution model employed for phylogeny reconstruction. The result is a probabilistic reconstruction of the most likely sequence at each internal node of the tree [20] [21].
2.2.2. Robustness and Model Misspecification Recent research indicates that ASR is generally robust to unincorporated evolutionary heterogeneity. The primary determinant of accuracy is strong phylogenetic signal, which is best achieved by using densely sampled alignments, rather than increasingly complex evolutionary models. For most nodes, reconstructions are nearly identical whether using simple homogeneous models or complex heterogeneous models derived from deep mutational scanning data [21].
2.2.3. From Nucleotide to Protein Inferred coding DNA sequences at the internal nodes are translated into protein sequences. These ancestral protein sequences, along with their phylogenetic trees, form the final output of the computational protocol and serve as the direct input for the laboratory phase of gene synthesis and protein expression [20].
The following table summarizes key experimental outcomes from published studies that utilized ASR, demonstrating its utility in protein engineering.
Table 1: Biotechnological Applications of Ancestral Protein Resurrection
| Ancestral Protein | Key Properties and Improvements | Experimental Validation | Source |
|---|---|---|---|
| Mammalian Coagulation Factor VIII (Anc-FVIII) | ⢠9-14 fold higher protein expression than human FVIII⢠Reduced inhibition by anti-drug antibodies (>75% reduction in some cases)⢠Improved biosynthesis in gene therapy vectors | In vitro activity assays; thrombin generation assays; inhibition assays with patient plasma; in vivo studies in hemophilia A mice [19] | |
| Mamba Aminergic Toxins (AncTx1 & AncTx5) | ⢠AncTx1: Most selective known peptide for α1A-adrenoceptor⢠AncTx5: Most potent known inhibitor of α2 adrenoceptor subtypes | Receptor binding affinity and selectivity assays across a panel of bioaminergic receptors [12] | |
| Precambrian β-lactamases | ⢠Hyperstability (Tm >30°C higher than modern counterparts)⢠Substrate promiscuity | Biochemical assays of thermal stability and enzymatic activity against various substrates [9] [22] |
The table below lists essential computational and experimental reagents for implementing this protocol.
Table 2: Key Research Reagents and Tools for ASR and Validation
| Reagent / Tool | Function / Application | Specific Example / Note |
|---|---|---|
| MrBayes | Software for Bayesian phylogenetic inference. | Used for TEM β-lactamase phylogeny with MCMC runs of 30 million generations [20]. |
| GARLi | Software for maximum likelihood phylogenetic inference. | Used for CTX-M-3 and OXA-51-like phylogenies [20]. |
| GTR+G+I Model | A standard nucleotide substitution model. | Accounts for different substitution rates, rate variation across sites, and invariant sites [20]. |
| Codon-Optimized cDNA | Synthetic gene for protein expression. | Ancestral FVIII cDNAs were codon-optimized for human cells and synthesized de novo [19]. |
| Solid-Phase Peptide Synthesis | Chemical synthesis of peptide toxins. | Used to synthesize ancestral mamba aminergic toxins (AncTx) for pharmacological profiling [12]. |
The following diagram illustrates the complete integrated computational and experimental workflow for ancestral protein resurrection, from sequence collection to functional characterization.
Figure 1. Integrated ASR and Protein Resurrection Workflow. The process begins with the collection of modern sequences, proceeds through computational phylogenetic analysis and ancestral sequence inference, and culminates in the laboratory synthesis, expression, and functional characterization of the resurrected ancestral protein.
Ancestral sequence reconstruction (ASR) has emerged as a powerful methodology for probing the deep evolutionary history of proteins and enzymes. This approach leverages the rapidly expanding amounts of sequence information available in genome databases to infer the sequences of ancestral proteins, which are then "resurrected" in the laboratory for functional and structural characterization [23]. ASR provides a unique window into the complex and intricate relationship between protein structure and function, offering insights not easily attainable by other methods. Within the broader context of ancestral protein resurrection research, the synthesis of these inferred ancestral gene sequences and their subsequent cloning into appropriate expression vectors represents the critical foundational step upon which all downstream experimental work depends.
Recent advancements have demonstrated that proteins reconstructed through ASR often exhibit enhanced stability, solubility, and functional promiscuity compared to their contemporary counterparts, making them particularly valuable for structural biology efforts that have traditionally been hampered by protein instability [7]. For instance, a 2025 study published in Nature Communications successfully utilized ASR to replace a native acyltransferase (AT) domain with an ancestral AT (AncAT) in a modular polyketide synthase, enabling high-resolution crystal structure determination that had proven elusive with the native protein [7]. This case exemplifies the growing importance of robust gene synthesis and molecular cloning strategies tailored specifically for ancestral sequences in advancing our mechanistic understanding of protein evolution and function.
The global gene synthesis market, projected to grow at a compound annual growth rate (CAGR) of 15-19% from 2025 to 2035, reflects the increasing adoption of these technologies across basic research and therapeutic development [24] [25]. This expansion is fueled by continuous improvements in synthesis chemistry, error correction technologies, and automation platforms that have significantly reduced costs per base pair while improving turnaround times and enabling increasingly complex projects.
For most research laboratories, outsourcing gene synthesis to specialized service providers represents the most efficient and reliable approach. The gene synthesis market includes established players such as GenScript, Twist Bioscience, Integrated DNA Technologies (IDT), and GeneArt (Thermo Fisher Scientific), each offering proprietary synthesis platforms with varying capabilities [24] [25]. When selecting a provider for ancestral gene synthesis, several technical considerations warrant careful evaluation.
Table 1: Key Considerations for Gene Synthesis Service Selection
| Consideration Factor | Importance for Ancestral Sequences | Recommended Specification |
|---|---|---|
| Maximum Length Capability | Critical for large multi-domain proteins | >5 kb for most ancestral enzymes; >10 kb for complex systems |
| Synthesis Accuracy | Essential for faithful reconstruction | <1 error per 10,000 bp with comprehensive error correction |
| Codon Optimization | Must balance expression with evolutionary accuracy | Species-specific optimization while preserving functional residues |
| Error Correction Methods | Critical for eliminating frameshifts and stop codons | Combination of enzymatic mismatch cleavage and sequencing verification |
| Turnaround Time | Impacts research progression | 2-4 weeks for standard constructs; faster options for urgent needs |
| Cloning Compatibility | Flexibility for downstream applications | Multiple vector options with customizable restriction sites or Gibson assembly compatibility |
| Price Structure | Budget management for multiple reconstructions | Transparent per-base-pair pricing with volume discounts |
The segment for genes "Above 5000 bp" is projected to exhibit the fastest growth rate, reflecting increasing demand for complex synthetic constructs in advanced research applications including ancestral protein resurrection [24]. Many providers now offer specialized services for synthesizing difficult sequences with high GC content or repetitive regions, which are commonly encountered in ancestral reconstruction projects.
The design phase is particularly critical for ancestral sequences, where the historical accuracy of the inferred sequence must be balanced with practical considerations for heterologous expression. The following workflow outlines the key decision points in this process:
When applying codon optimization to ancestral sequences, it is crucial to preserve potentially important regulatory motifs and avoid optimizing regions that may represent authentic historical signatures. For example, a 2025 study on modular polyketide synthases successfully created a chimeric didomain by replacing the native AT domain with an ancestral AT (AncAT), confirming that the chimeric protein retained similar enzymatic function to the native didomain while exhibiting enhanced properties for structural analysis [7]. This case demonstrates the functional validation required after ancestral gene synthesis.
Synthesizing ancestral genes presents unique technical challenges beyond those encountered with contemporary sequences. The table below outlines common challenges and recommended mitigation strategies:
Table 2: Technical Challenges in Ancestral Gene Synthesis
| Challenge | Impact on Synthesis | Recommended Solutions |
|---|---|---|
| Ambiguous ancestral states | Uncertainty in residue identity; multiple possible sequences | Synthesize multiple variants; incorporate degeneracy at low-probability positions |
| Unusual codon preferences | Potential expression issues in heterologous systems | Partial codon optimization preserving key ancestral signatures |
| Structural instability | Folding problems affecting protein function | Incorporate stabilizing ancestral mutations identified through phylogenetic analysis |
| Repetitive sequences | Synthesis errors and recombination in hosts | Codon diversification; synthesis in fragments with assembly |
| GC-rich regions | Secondary structure formation impeding synthesis | Strategic AT-rich codon substitution without altering amino acid sequence |
| Toxic products | Failure to clone synthesized genes | Use of tightly regulated expression systems; lower copy number vectors |
The application of ASR to a partial region of targeted multi-domain proteins has been shown to expand the potential of ASR and may serve as a valuable framework for investigating the structure and function of various multi-domain proteins [7]. This modular approach to ancestral resurrection can help mitigate synthesis challenges associated with very large genes.
Selecting an appropriate expression vector is critical for successful ancestral protein production. Different host systems offer distinct advantages depending on the nature of the ancestral protein and the intended downstream applications.
Table 3: Vector Systems for Ancestral Protein Expression
| Vector System | Typical Applications | Advantages | Limitations |
|---|---|---|---|
| Bacterial (pET, pBAD) | High-throughput screening; structural studies | High yield; low cost; extensive toolkit | Lack of eukaryotic post-translational modifications |
| Yeast (pPIC, pYES) | Eukaryotic proteins requiring glycosylation | Eukaryotic processing; higher yields than mammalian | Hyperglycosylation; fewer tools than bacterial |
| Baculovirus/Insect Cell | Complex eukaryotic proteins; structural biology | Proper folding and modification; high yields | Time-consuming; more expensive |
| Mammalian (pcDNA, pCMV) | Functional studies of mammalian proteins | Native-like processing and modification | Lower yields; higher cost; technical complexity |
| Cell-free Systems | Toxic proteins; incorporation of unnatural amino acids | Flexibility; no cellular toxicity constraints | Limited scale; high cost for large quantities |
Recent advances in vector design specifically for ancestral proteins include the incorporation of solubility tags (MBP, GST, SUMO) and cleavage sites to enhance expression and facilitate purification. For example, Belinda Chang's laboratory at the University of Toronto has engineered specialized expression vectors for heterologous opsin expression in mammalian cell culture, developing spectroscopic assays for visual pigment function that can be applied to non-model vertebrate pigments [23].
The cloning strategy for ancestral genes must be selected based on insert size, required precision, and downstream applications. The following workflow illustrates a robust cloning pipeline suitable for most ancestral sequences:
For most ancestral protein projects, Gibson Assembly or related methods (In-Fusion, NEBuilder) offer significant advantages due to their sequence independence and high efficiency, particularly when working with large inserts or multiple variants. These methods eliminate dependence on restriction sites, which is especially valuable when preserving the precise ancestral sequence is critical.
Rigorous quality control is essential when working with synthesized ancestral genes to ensure sequence fidelity before investing in functional characterization. A multi-tiered verification approach is recommended:
The integration of next-generation sequencing (NGS) technologies has dramatically improved the efficiency of sequence verification, particularly when working with multiple ancestral variants or library approaches. The GenScript Life Science Research Grant Program, for instance, has supported projects requiring the synthesis of hundreds of synthetic sequences, which would necessitate robust high-throughput verification methods [26].
Successful ancestral protein resurrection depends on access to high-quality research reagents and specialized services. The following table details essential materials and their applications in gene synthesis and molecular cloning workflows for ancestral sequences:
Table 4: Essential Research Reagents for Ancestral Sequence Research
| Reagent Category | Specific Products | Function in Workflow | Recommended Providers |
|---|---|---|---|
| Gene Synthesis Services | Custom gene fragments; codon-optimized sequences | De novo production of ancestral coding sequences | GenScript, Twist Bioscience, IDT, GeneArt (Thermo Fisher) |
| Cloning Kits | Gibson Assembly Master Mix; Restriction enzyme kits; Ligation kits | Assembly of synthesized genes into expression vectors | NEB, Thermo Fisher, Takara Bio, Promega |
| Expression Vectors | pET series (bacterial); pPIC (yeast); baculovirus (insect) | Protein production in heterologous systems | Addgene, commercial vendors, academic collections |
| Competent Cells | DH5α (cloning); BL21(DE3) (expression); specialized strains | Plasmid propagation and protein expression | NEB, Thermo Fisher, homemade preparation |
| Sequence Verification | Sanger sequencing; NGS services; quality control protocols | Confirmation of synthesized sequence fidelity | Genewiz, Eurofins Genomics, Plasmidsaurus |
| Antibodies | Anti-tag antibodies; custom ancestral protein antibodies | Detection and purification of expressed proteins | Commercial vendors; custom service providers |
| Purification Resins | Ni-NTA; glutathione agarose; antibody-coupled resins | Isolation of recombinant ancestral proteins | Cytiva, Thermo Fisher, Bio-Rad, Qiagen |
Funding opportunities such as the GenScript Life Science Research Grant Program provide critical support for obtaining these research reagents, with grants specifically earmarked for purchasing GenScript reagents and services to advance projects in areas including gene and cell therapy, antibody drug discovery, and vaccine development [26].
ASR has proven particularly valuable in structural biology, where enhanced stability of ancestral proteins facilitates high-resolution structure determination. A landmark 2025 study demonstrated this application in modular polyketide synthases (PKSs), large multi-domain enzymes critical for biosynthesis of polyketide antibiotics [7]. Researchers focused on the FD-891 PKS loading module composed of ketosynthase-like decarboxylase (KSQ), acyltransferase (AT) and acyl carrier protein (ACP) domains. They constructed a KSQAncAT chimeric didomain by replacing the native AT with an ancestral AT (AncAT) using ASR [7].
After confirming that the KSQAncAT chimeric didomain retained similar enzymatic function to the native KSQAT didomain, the research team successfully determined a high-resolution crystal structure of the KSQAncAT chimeric didomain and cryo-EM structures of the KSQ-ACP complex [7]. These cryo-EM structures could not be determined for the native protein, exemplifying the utility of ASR to enable cryo-EM single-particle analysis. This case study demonstrates how integrating ASR with structural analysis provides deeper mechanistic insight into complex protein systems, with the potential to expand to various multi-domain proteins [7].
Beyond structural biology, ancestral gene synthesis has enabled fundamental investigations into protein evolution and function. Belinda Chang's laboratory at the University of Toronto has pioneered ancestral approaches for studying visual pigment evolution, using these methods to understand the evolution of spectral tuning in different vertebrate groups including cetaceans, Neotropical fishes, and avian visual pigments [23]. Their interdisciplinary approach involves computational methods of evolutionary sequence analysis to infer ancestral sequences, synthesizing the ancestral genes, and expressing them in the laboratory [23].
This research has revealed how visual pigments have adapted to different ecological niches and light environments, providing insights into the molecular mechanisms underlying visual adaptations. For example, their studies of Neotropical cichlids found high levels of positive selection at non-overlapping subsets of amino acid sites when compared with African rift lake cichlids, suggestive of divergent selection that may target similar molecular functions [23].
Despite careful planning, researchers may encounter challenges during ancestral gene synthesis and cloning. The following table outlines common problems and evidence-based solutions:
Table 5: Troubleshooting Guide for Ancestral Gene Synthesis and Cloning
| Problem | Possible Causes | Recommended Solutions |
|---|---|---|
| No colonies after transformation | Toxic gene product; inefficient assembly; vector issues | Use lower copy number vectors; try different competent cells; verify assembly efficiency |
| Incorrect sequence | Synthesis errors; PCR mutations; recombination | Request synthesis with enhanced error correction; use high-fidelity polymerases; employ recombination-deficient strains |
| No protein expression | Codon bias; toxic effects; improper folding | Try different expression strains; adjust induction conditions; test solubility tags; optimize growth temperature |
| Insoluble protein | Misfolding; aggregation; lack of partners | Test different tags (MBP, SUMO); optimize expression conditions; co-express with chaperones; refold from inclusion bodies |
| Low yield | Protease degradation; poor translation; toxicity | Add protease inhibitors; optimize induction OD and temperature; use autoinduction media; try different hosts |
| Incorrect protein size | Proteolysis; alternative start codons; sequencing errors | Add protease cocktail; use N-terminal tags; verify full-length sequence; check for internal start sites |
When troubleshooting, it is often valuable to return to the phylogenetic analysis phase to re-examine the ancestral sequence inference, particularly for regions that repeatedly cause expression problems. Sometimes, alternative reconstructions with statistically equivalent probability may yield more expressible variants while still representing plausible ancestral states.
The field of ancestral sequence resurrection continues to evolve rapidly, driven by advances in both computational biology and gene synthesis technologies. Several emerging trends are likely to shape future research in this area. Decreasing costs of gene synthesis are making it accessible to a wider range of researchers and companies, facilitating larger-scale ancestral resurrection projects [24]. The growing adoption of synthetic biology approaches across basic research and therapeutic development is increasing demand for custom-designed genes, including ancestral sequences [25]. Advances in automation and high-throughput synthesis are enabling more comprehensive exploration of ancestral sequence space and library-based approaches [24]. The development of more sophisticated phylogenetic methods and integration with machine learning approaches is improving the accuracy of ancestral sequence inference, particularly for deep evolutionary reconstructions. Finally, the application of ancestral resurrection to increasingly complex multi-domain proteins and metabolic pathways is expanding the biological questions that can be addressed using these approaches [7].
Gene synthesis and molecular cloning strategies form the technical foundation for ancestral protein resurrection, enabling researchers to bridge deep evolutionary history with contemporary experimental approaches. As these technologies continue to advance, they will undoubtedly yield new insights into protein evolution and function, with applications ranging from basic mechanistic studies to the development of novel enzymes with enhanced properties for biotechnology and therapeutic applications. The integration of robust synthetic biology approaches with phylogenetic inference represents a powerful framework for exploring protein sequence-function relationships across evolutionary timescales.
Ancestral sequence reconstruction (ASR) is a powerful technique in molecular evolution that infers the sequences of ancient proteins from the genomes of modern organisms [2]. The process involves the computational prediction of ancestral sequences, followed by their chemical synthesis, heterologous expression, and purification for functional and structural characterization [27]. This approach, first proposed by Pauling and Zuckerkandl in the 1960s, has evolved into a sophisticated methodology that provides a unique window into protein evolution, enabling researchers to test hypotheses about ancestral environments, enzyme mechanisms, and evolutionary trajectories [2] [6]. The resulting "resurrected" proteins often exhibit enhanced stability and unique functional properties compared to their modern counterparts, making them valuable not only for evolutionary studies but also for biotechnology and drug development [6] [7].
The heterologous expression of resurrected proteins presents distinct challenges and opportunities. While ancestral proteins frequently demonstrate increased thermostability and solubilityâproperties that facilitate crystallization and structural analysis [7]âtheir expression in standard prokaryotic systems can be complicated by unique structural features or codon usage biases. This protocol details optimized methods for the expression and purification of resurrected proteins, with a particular emphasis on strategies to leverage their inherent stability while mitigating potential expression challenges. The methods described herein are framed within the context of a broader research program focused on developing robust, standardized laboratory protocols for ancestral protein resurrection.
The overall process for resurrecting and characterizing an ancestral protein integrates computational biology, molecular biology, and protein biochemistry. The workflow proceeds from sequence selection and analysis through to functional validation, with careful attention to the unique requirements of ancestral proteins at each stage.
Figure 1. Overall workflow for ancestral protein resurrection, from sequence selection to functional characterization. Key decision points include ancestral node selection, expression host choice, and purification strategy.
When working with resurrected proteins, several unique factors must be considered during experimental design:
Table 1. Essential research reagents for heterologous expression and purification of resurrected proteins.
| Category | Reagent | Function/Application |
|---|---|---|
| Cloning | High-fidelity DNA polymerase (e.g., Q5, Phusion) | Error-free amplification of expression constructs [7] |
| Restriction enzymes & ligase | Vector construction and gene insertion [30] | |
| Codon-optimized synthetic genes | Custom gene synthesis for optimal expression [28] | |
| Expression Hosts | Escherichia coli BL21(DE3) | Standard prokaryotic host; suitable for many ancestral proteins [28] [29] |
| Aspergillus niger AnN2 chassis | Eukaryotic host with high secretion capacity; engineered for low background proteolysis [30] | |
| Aspergillus oryzae | GRAS-status fungal host for complex eukaryotic proteins [31] | |
| Expression Media | LB, TB media | Standard bacterial growth media [29] |
| DMSO, glycerol, sorbitol | Chemical chaperones to improve folding efficiency [28] [29] | |
| IPTG | Inducer for T7/lac-based expression systems [29] | |
| Purification | Ni-NTA or Co-TALON resin | Immobilized metal affinity chromatography for His-tagged proteins [29] |
| GST, MBP fusion systems | Fusion tags to enhance solubility and enable affinity purification [28] [29] | |
| Protease cleavage enzymes (e.g., TEV, thrombin) | Removal of affinity tags after purification [29] | |
| Buffers & Additives | L-arginine, proline, betaine | Solubility enhancers in lysis and purification buffers [29] |
| PMSF, protease inhibitor cocktails | Prevent proteolytic degradation during purification [30] | |
| CHAPS, Triton X-100 | Detergents for membrane protein solubilization |
Choosing an appropriate expression host is critical for successful production of resurrected proteins. Different host systems offer distinct advantages depending on the properties of the target ancestral protein.
Figure 2. Host selection decision tree for expressing resurrected proteins. The choice depends on protein properties and research goals.
Escherichia coli remains the most widely used host for heterologous protein expression due to its rapid growth, well-characterized genetics, and low cost [28] [29]. For ancestral proteins, the following considerations apply:
Filamentous fungi, particularly Aspergillus species, offer powerful alternatives for ancestral proteins that require eukaryotic folding machinery or are refractory to expression in prokaryotic systems [30] [31].
Regardless of the host system, the following elements should be considered in vector design:
Before proceeding to large-scale expression, small-scale trials are essential to identify optimal conditions for soluble expression of the resurrected protein.
Protocol: Expression Screening
Table 2. Troubleshooting guide for expression problems with resurrected proteins.
| Problem | Possible Causes | Potential Solutions |
|---|---|---|
| No Expression | Toxic to host, Poor codon usage, Incorrect vector/construct | Test different expression strains, Verify codon optimization, Sequence verification of construct |
| Expression Only in Insoluble Fraction | Aggregation, Misfolding, Too rapid expression | Lower induction temperature (16-25°C), Reduce IPTG concentration (0.01-0.1 mM), Co-express molecular chaperones [29], Add chemical chaperones, Test fusion tags (MBP, NusA) |
| Low Yield of Soluble Protein | Proteolysis, Poor stability, Suboptimal growth conditions | Add protease inhibitors, Shorten induction time, Optimize medium (TB often gives higher yield than LB), Test autoinduction media |
| Protein Degradation | Host protease activity, Unstable protein | Use protease-deficient strains, Add mixture of protease inhibitors, Purify immediately at 4°C |
For ancestral proteins that are poorly expressed in E. coli, filamentous fungi provide a powerful alternative expression platform.
Protocol: Aspergillus niger Expression
For proteins expressed with polyhistidine tags, IMAC provides efficient single-step purification with high recovery.
Protocol: Ni-NTA Purification
For ancestral proteins secreted by fungal hosts, purification begins with concentrated culture supernatant.
Protocol: Purification from Aspergillus Culture Supernatant
For structural or functional studies requiring removal of affinity tags:
Comprehensive characterization is essential to confirm that the purified ancestral protein is properly folded and functional.
Protocol: Biophysical Characterization
Table 3. Expected yields and properties of resurrected proteins across different expression systems.
| Expression System | Typical Yield Range | Advantages | Limitations | Ideal for Ancestral Proteins With |
|---|---|---|---|---|
| E. coli | 0.5 - 50 mg/L culture | Rapid, low cost, high throughput | Limited PTMs, potential aggregation | High thermostability, no complex PTM requirements [28] [29] |
| A. niger | 50 - 400 mg/L culture [30] | High secretion, eukaryotic PTMs, low background proteases in engineered strains | Longer culture times, more complex genetics | Secretory pathway compatibility, industrial scale-up [30] [31] |
| A. oryzae | 10 - 100 mg/L culture | GRAS status, strong secretion, food-compatible | Moderate yields for some proteins | Therapeutic or food-related applications [31] |
The successful heterologous expression and purification of resurrected proteins opens numerous avenues for scientific investigation. The enhanced stability often exhibited by ancestral proteins makes them particularly attractive for structural biology efforts, as demonstrated by the successful crystallization of an ancestral AT domain that facilitated high-resolution structural analysis of a polyketide synthase loading module [7]. Furthermore, the unique functional properties of resurrected proteins can provide insights into the evolutionary history of enzyme mechanisms and the environmental conditions of ancient organisms [6].
The protocols outlined in this document provide a comprehensive framework for expressing and purifying ancestral proteins, with particular attention to the unique challenges and opportunities they present. By leveraging both prokaryotic and eukaryotic expression systems and implementing robust purification strategies, researchers can reliably produce resurrected proteins for functional characterization, structural analysis, and biotechnology applications. As ancestral protein reconstruction continues to grow as a field, these standardized protocols will facilitate more efficient and reproducible resurrection of ancient proteins, deepening our understanding of molecular evolution while providing stable protein scaffolds for engineering novel functions.
Within the field of ancestral protein resurrection, biophysical characterization is a critical step that bridges computational sequence reconstruction with functional validation. This protocol details the application of key biophysical techniques to analyze the stability, folding, and structural integrity of resurrected ancestral proteins. Resurrected ancestral proteins often exhibit remarkable properties, including enhanced stability, conformational flexibility, and altered interaction patterns, which require rigorous experimental scrutiny [32]. The following application notes provide a standardized framework for researchers to obtain quantitative, reproducible data, enabling insights into the evolutionary trajectories of protein energy landscapes [1].
The following table summarizes the primary biophysical methods used for characterizing resurrected ancestral proteins, their key applications, and the specific structural and stability parameters they measure.
Table 1: Core Biophysical Techniques for Ancestral Protein Characterization
| Technique | Key Applications | Measurable Parameters | Information Level |
|---|---|---|---|
| Small-Angle X-ray Scattering (SAXS) | Analysis of global conformation and ensemble states in solution [33]. | Radius of gyration (Rg), Pair distance distribution function, Porod volume [33]. | Global, Low-Resolution |
| Hydrogen/Deuterium Exchange (HDX) | Probing solvent accessibility and dynamics of backbone amides [33]. | Protection Factor (PF), HDX kinetics [33]. | Site-Specific (Backbone) |
| Hydroxyl Radical Protein Footprinting (HRPF) | Mapping solvent-exposed sidechains and protein-protein interfaces [33]. | HRPF rate, Sidechain solvent accessibility [33]. | Site-Specific (Sidechain) |
| Nuclear Magnetic Resonance (NMR) | Determining atomic-resolution structure and dynamics in solution [34]. | Chemical shift, Relaxation rates, Interatomic distances [34]. | Atomic, Residue-Level |
| Differential Scanning Calorimetry (DSC) | Measuring thermal stability and unfolding cooperativity. | Melting temperature (Tm), Enthalpy of unfolding (ÎH), Heat capacity change (ÎCp). | Global Stability |
| Circular Dichroism (CD) Spectroscopy | Assessing secondary structure content and folding transitions [34]. | Molar ellipticity, Melting temperature (Tm) [34]. | Global, Secondary Structure |
Principle: Size-exclusion chromatography coupled SAXS (SEC-SAXS) provides an ensemble-averaged representation of a protein's global conformation and flexibility in solution, free from aggregation artifacts [33]. This is vital for characterizing the potentially unique conformational ensembles of ancestral proteins.
Workflow Diagram: SEC-SAXS for Conformational Analysis
Procedure:
Principle: DSC directly measures the heat capacity change associated with protein thermal unfolding, providing a model-free assessment of thermodynamic stability. This is crucial for quantifying the often-enhanced thermostability of resurrected ancestral proteins [32].
Procedure:
Principle: Hydrogen/Deuterium Exchange coupled with Mass Spectrometry (HDX-MS) measures the rate at which backbone amide hydrogens exchange with deuterium in the solvent, revealing dynamics and solvent accessibility at peptide-level resolution [33].
Workflow Diagram: HDX-MS for Solvent Exposure Mapping
Procedure:
Table 2: Essential Reagents and Materials for Biophysical Characterization
| Research Reagent | Specifications & Function | Application Notes |
|---|---|---|
| Size-Exclusion Chromatography Column | Superdex 200 Increase 3.2/300; Separates monodisperse protein from aggregates for SEC-SAXS and clean biophysical analysis. | Ensure buffer compatibility. Pre-calibrate with standard proteins for molecular weight estimation. |
| SAXS Running Buffer | 20 mM HEPES, 150 mM NaCl, pH 7.5; Provides a physiologically relevant, non-interfering environment for scattering. | Must be filter-sterilized (0.22 μm) and degassed. Match buffer exactly for sample and background. |
| DSC Reference Buffer | 20 mM Potassium Phosphate, pH 7.0; A low-ionization-enthalpy buffer for precise baseline subtraction in DSC. | Critical to use the same batch of dialysate for sample preparation and reference cell. |
| Deuterium Oxide (DâO) | 99.9% atom D; The labeling agent for HDX-MS experiments, enabling tracking of solvent exposure. | Store properly to prevent HâO contamination. Use high-purity grade for consistent results. |
| Quench Buffer (for HDX-MS) | 0.1 M Phosphate, 0.5 M TCEP, pH 2.5; Lowers pH and temperature to quench HDX reaction and denatures protein for digestion. | Must be pre-chilled to 0°C. TCEP is preferred over DTT for stability at low pH. |
| Immobilized Pepsin Column | Poroszyme Immobilized Pepsin Cartridge; Provides rapid, reproducible digestion for HDX-MS under quench conditions. | Keep column chilled during digestion. Monitor digestion efficiency regularly. |
| 10Z-Vitamin K2-d7 | 10Z-Vitamin K2-d7|Deuterated Research Standard | High-purity 10Z-Vitamin K2-d7 for research. An internal standard for LC-MS/MS analysis of Vitamin K2 metabolites. For Research Use Only. Not for human consumption. |
| (Z)-Roxithromycin-d7 | (Z)-Roxithromycin-d7, MF:C₄₁H₆₉D₇N₂O₁₄, MW:828.09 | Chemical Reagent |
Integrating data from the above protocols provides a comprehensive picture of ancestral protein biophysics. For instance, a resurrected ancestral protein might display:
This multi-faceted characterization is essential for moving beyond simple structural models and understanding the evolved functional dynamics and stability that define ancestral proteins, ultimately illuminating their evolutionary histories and unlocking their biotechnological potential [7] [1] [32].
Within the framework of ancestral protein resurrection research, functional assays are indispensable for characterizing the biochemical properties of resurrected enzymes. This field aims to understand molecular evolution by inferring the sequences of ancient proteins, synthesizing them, and experimentally analyzing their traits [1]. Functional assays provide the critical data on enzymatic activity, ligand binding, and substrate specificity that allow researchers to test evolutionary hypotheses about how protein energy landscapes and functions have shifted over millennia [1]. The precision of these assays directly determines the robustness of evolutionary inferences, making the choice of appropriate methodologies a cornerstone of ancestral protein research. This application note details cutting-edge and classical protocols tailored to the unique challenges of profiling resurrected ancestral enzymes, which often possess unknown structures and substrate preferences.
Measuring enzymatic activity is fundamental to establishing the baseline function of a resurrected protein. Modern approaches leverage high-throughput technologies and sensitive detection methods to comprehensively profile enzyme kinetics and inhibition.
A powerful contemporary method combines the precision of Activity-Based Protein Profiling (ABPP) with the efficiency of microplate assay technology [35]. This protocol is particularly valuable for ancestral enzymes of unknown structure or specificity, as it uses an activity-based probe to directly monitor enzyme function and inhibitor interactions.
Principle: The core of this method is competitive ABPP, which utilizes an electrophilic, fluorescently-tagged Activity-Based Probe (ABP) that covalently binds to the active site of the enzyme. Competition with a potential inhibitor reduces the fluorescent signal, allowing for characterization of inhibitor potency and specificity [35]. The workflow involves chemical modification of the enzyme, such as pig liver esterase (PLE), to introduce a tag (e.g., streptavidin) for immobilization on biotinylated assay plates. This setup enables parallelized screening of compound libraries and estimation of ICâ â values in a single operation [35].
Table 1: Key Reagents for Microplate-Based ABPP Assay
| Research Reagent | Function in the Protocol |
|---|---|
| Fluorophosphonate (FP)-based ABP | Electrophilic probe that covalently labels active sites; provides fluorescent readout. |
| Biotinylated Assay Plate | Solid support for immobilizing streptavidin-tagged enzymes. |
| Streptavidin-Tagged Enzyme | Enables specific immobilization of the enzyme onto the microplate. |
| Test Inhibitor Library | Collection of compounds for screening against the target enzyme. |
Protocol Steps:
For well-characterized reactions, a traditional colorimetric assay provides a robust and straightforward method to determine activity, such as for ancestral amylases.
Principle: This assay measures the release of reducing sugars (maltose) from starch by an amylase enzyme. The 3,5-dinitrosalicylic acid (DNS) reagent reacts with the reducing sugars, producing a colored complex that can be quantified by absorbance [36].
Protocol Steps:
Diagram 1: Workflow for a microplate-based competitive ABPP assay. The fluorescence signal is inversely related to inhibitor potency.
Ligand Binding Assays (LBAs) are a cornerstone of bioanalytical testing, crucial for understanding how resurrected ancestral proteins interact with potential substrates, inhibitors, or cofactors.
LBAs are highly sensitive and specific methods used to detect and quantify biomolecular interactions. They are versatile tools for pharmacokinetics, pharmacodynamics, and biomarker analysis [37]. These assays are particularly well-suited for characterizing biologics and complex therapeutics, making them ideal for protein resurrection studies focused on ancient therapeutic targets.
Table 2: Common Techniques for Ligand Binding Assays
| Technique | Principle | Key Application in Ancestral Research |
|---|---|---|
| Surface Plasmon Resonance (SPR) [38] | Measures binding kinetics in real-time by detecting changes in refractive index when a ligand binds to an immobilized target. | Determining the association ((k\text{on})) and dissociation ((k\text{off})) rates of an ancestral enzyme with its substrate. |
| Biolayer Interferometry (BLI) [38] | An optical technique that monitors interference patterns to measure binding kinetics and affinity. | A label-free alternative to SPR for characterizing binding to biosensor-tipped probes. |
| Enzyme-Linked Immunosorbent Assay (ELISA) [38] | An end-point assay using enzyme-mediated color change to detect binding. | High-throughput screening of antibody binding to resurrected ancestral antigens. |
| Microfluidic Diffusional Sizing (MDS) [38] | Measures the change in hydrodynamic radius ((R_\text{h})) of a fluorescent target upon ligand binding in solution. | Assessing binding and complex formation under native conditions without immobilization. |
| Radioligand Binding Assays (RBA) [38] | Uses a radioisotope-tagged ligand to measure binding to the target. | Highly sensitive quantification of binding for low-abundance or low-affinity interactions. |
MDS is a modern, in-solution technique that offers a label-free advantage and operates well in complex biological matrices, which can be beneficial for studying ancient proteins that may require specific chaperones or cofactors.
Principle: MDS measures the diffusion coefficient of a fluorescent target in a microfluidic chamber. Upon binding to an unlabeled ligand, the complex's hydrodynamic radius increases, leading to a slower diffusion rate. This size change is used to quantify binding affinity [38].
Protocol Steps:
Tips to Avoid Pitfalls:
Diagram 2: A generalized workflow for determining binding kinetics and affinity using surface-based techniques like SPR or BLI.
A central goal in ancestral protein resurrection is to understand how substrate specificity evolved. While experimental profiling is essential, new computational tools powerfully complement wet-lab methods.
Broad-specificity profiling involves testing the enzyme against a large panel of potential substrates. The microplate-based ABPP protocol described in Section 2.1 is an excellent example, as it can be adapted to screen diverse substrate libraries in parallel to map the promiscuity of a resurrected enzyme [35].
For the millions of enzymes that lack characterized substrates, computational models are invaluable for guiding experimental work.
EZSpecificity Tool: This is a state-of-the-art AI tool, specifically a cross-attention-empowered SE(3)-equivariant graph neural network, that predicts enzyme-substrate specificity [39] [40]. It was trained on a comprehensive database of enzyme-substrate interactions at the sequence and structural levels.
EZSCAN Tool: This complementary software focuses on identifying the specific amino acid residues that determine substrate specificity [41]. It uses a machine learning-based binary classification algorithm on sequences of homologous enzymes with different specificities to pinpoint critical residues.
Table 3: Comparison of Specificity Prediction and Analysis Tools
| Tool | Type | Primary Function | Key Utility in Ancestral Research |
|---|---|---|---|
| EZSpecificity [39] [40] | AI Graph Neural Network | Predicts the binding compatibility between an enzyme and a substrate. | Generating high-confidence hypotheses for which substrates to test with a resurrected ancestral enzyme. |
| EZSCAN [41] | Machine Learning Classifier | Identifies amino acid residues critical for determining substrate specificity. | Pinpointing evolutionary mutations that likely led to shifts in function between ancestral and modern enzymes. |
The integration of detailed functional assays with powerful computational predictions creates a robust framework for advancing ancestral protein research. Experimental protocols like ABPP-powered microplate assays and modern LBAs provide the ground-truth data on activity, binding, and specificity for resurrected enzymes. These empirical data are essential for validating evolutionary models derived from Ancestral Sequence Reconstruction (ASR), which infers ancient sequences to characterize how historical mutations altered protein energy landscapes and function [1]. Meanwhile, AI tools like EZSpecificity and EZSCAN offer an efficient strategy to guide experimental efforts, helping researchers prioritize which substrates and residues to investigate [39] [41]. Together, this combined empirical and computational toolkit enables a deeper understanding of the mechanistic basis of protein evolution, from reconstructing ancient functions to engineering new ones.
Ancestral protein resurrection relies on accurate phylogenetic trees to infer the genetic sequences of ancient proteins. Phylogenetic uncertaintyâambiguity in evolutionary relationshipsâdirectly impacts the accuracy of these inferred sequences and the functionality of the resulting resurrected proteins. Traditional phylogenetic support measures, such as Felsensteinâs bootstrap, are computationally prohibitive for large datasets typical of modern genomic studies and focus on clade membership, which is less relevant for tracing specific mutational histories [42]. This note outlines a streamlined, reliable workflow integrating the Subtree Pruning and Regrafting-based Tree Assessment (SPRTA) method to quantify and manage phylogenetic uncertainty, ensuring robust downstream ancestral reconstructions for drug discovery [42].
The reliability of any phylogenetic analysis is also contingent on selecting an appropriate evolutionary model. Research indicates that multiple sequence alignment (MSA) uncertainty can significantly affect model selection, particularly for nucleotide data, potentially leading to the selection of different best-fitting models from different MSAs of the same sequence set [43]. This cascade effect underscores the necessity of integrating alignment assessment and model selection into a cohesive, validated protocol.
Table 1: Key Branch Support Methods for Assessing Phylogenetic Uncertainty
| Method | Core Principle | Computational Demand | Interpretation in Genomic Epidemiology |
|---|---|---|---|
| SPRTA [42] | Assesses confidence in evolutionary origin via Subtree Pruning and Regrafting moves | At least 2 orders of magnitude lower than bootstrap methods | Approximate probability that a lineage evolved directly from another |
| Felsensteinâs Bootstrap [42] | Resamples data to measure repeatability of clades | Excessively high for pandemic-scale datasets | Confidence that a set of taxa form a true clade |
| aBayes [42] | Compares likelihood of inferred tree against alternatives | Lower than bootstrap, but higher than SPRTA | Approximate posterior probability of a clade |
This protocol provides a step-by-step guide for phylogenetic tree estimation with integrated uncertainty assessment, tailored for projects requiring high-confidence ancestral node inference, such as ancestral protein resurrection.
Objective: Generate a reliable Multiple Sequence Alignment (MSA) and evaluate its robustness to uncertainty.
Objective: Identify the optimal evolutionary model for phylogenetic inference, accounting for alignment uncertainty.
Objective: Estimate the phylogenetic tree and posterior probabilities of clades using Bayesian inference.
mb.exe) from the command line in the directory containing your NEXUS file. Execute your analysis file using the execute command within MrBayes. Monitor the average standard deviation of split frequencies; a value below 0.01 suggests the Markov Chain Monte Carlo (MCMC) analysis has converged [44].sump command in MrBayes to examine parameter estimates and ensure effective sample sizes (ESS) are adequate (>200). The consensus tree with posterior probabilities can be visualized in tree viewers like FigTree or iTOL [44].Objective: Quantify confidence in the evolutionary origin of specific lineages, which is critical for pinpointing ancestral nodes for resurrection.
Table 2: Essential Computational Tools for Phylogenetic Analysis and Ancestral Resurrection
| Tool / Reagent | Function in Protocol | Specifications |
|---|---|---|
| GUIDANCE2 & MAFFT [44] | Performs robust multiple sequence alignment and identifies unreliable regions. | Web server or command-line tool. Critical for handling indels and complex evolutionary events. |
| ProtTest & MrModeltest2 [44] | Automates selection of best-fit evolutionary model for protein or nucleotide data. | Relies on Java and PAUP*. Uses AIC/BIC criteria for statistical robustness. |
| MrBayes [44] | Executes Bayesian phylogenetic inference to estimate trees with posterior probabilities. | Version 3.2.7a. Requires NEXUS format input. Computes consensus tree from MCMC samples. |
| SPRTA [42] | Provides efficient, pandemic-scale assessment of confidence in evolutionary origins. | Integrated into tools like MAPLE. Shifts focus from clades to mutational histories. |
| Ancestral Sequence Reconstruction (ASR) [45] | Infers the genetic sequence of ancestral nodes identified in the phylogenetic tree. | Computational technique. Input is a high-confidence tree and alignment. Output is a predicted ancient sequence. |
| CRISPR-Cas Tools [45] | Enables genome editing in model organisms to test the function of resurrected ancient genes/pathways. | Uses nucleases like Cas12a. Ancestrally reconstructed versions (e.g., ReChb) offer PAM-flexibility for broader targeting. |
| 5-Carboxy Imazapyr | 5-Carboxy Imazapyr | High-purity 5-Carboxy Imazapyr for research. This product is For Research Use Only (RUO). Not for human or veterinary use. |
| GNE-0877-d3 | GNE-0877-d3, MF:C₁₄H₁₃D₃F₃N₇, MW:342.34 | Chemical Reagent |
The successful resurrection of functional ancient proteins, such as the paleomycin antibiotics or PAM-flexible gene-editing enzyme ReChb, is fundamentally dependent on a rigorous phylogenetic foundation [45]. By adopting the integrated protocol outlined hereâwhich systematically addresses alignment uncertainty, model selection sensitivity, and phylogenetic confidenceâresearchers can significantly enhance the reliability of their ancestral reconstructions. This robust framework empowers scientists in drug discovery and biotechnology to confidently mine evolutionary history for novel bioactive compounds, such as antibiotics and cancer treatments, turning deep time into a viable resource for addressing modern medical challenges [46] [47].
Ancestral Sequence Reconstruction (ASR) infers the sequences of ancient proteins from modern descendants, enabling researchers to study molecular evolution and resurrect ancestral proteins in the laboratory [1]. A significant challenge in ASR involves handling low-probability and ambiguous sites, where the reconstructed amino acid has weak statistical support. These ambiguities often arise at fast-evolving sites or nodes connected by long branches, where phylogenetic signal is weak [48]. This Application Note details standardized protocols to identify, resolve, and validate such problematic sites, ensuring robust reconstructions for downstream functional characterization.
Ambiguity in ASR primarily stems from two technical challenges:
The statistical support for a reconstructed ancestral state is quantified by its posterior probability, calculated using the marginal probability method [1]. Sites with posterior probabilities below a defined threshold are considered ambiguous. The table below summarizes critical thresholds and their interpretations.
Table 1: Quantitative Thresholds for Identifying Ambiguous Sites
| Metric | Threshold Value | Interpretation | Recommended Action |
|---|---|---|---|
| Posterior Probability (PP) | PP < 0.8 | Low confidence in the inferred amino acid [1] | Flag for manual inspection and uncertainty analysis. |
| Branch Length | Long Branches | Increased probability of reconstruction error [48] | Interpret ancestral nodes with caution; prioritize densely sampled phylogenies. |
| Site-wise Rate | Fast-Evolving Sites | Weak phylogenetic signal, high ambiguity [48] | Treat reconstructed residues as provisional. |
The following workflow provides a systematic approach for identifying and resolving ambiguous sites during ASR. It integrates computational checks and experimental validation.
Diagram 1: A workflow for managing ambiguous sites in ASR. PP = Posterior Probability.
Purpose: To systematically flag low-probability sites in a reconstructed ancestral sequence.
Materials:
Methodology:
Purpose: To empirically test the functional impact of alternative residues at ambiguous sites.
Materials:
Methodology:
k_cat/K_m) for enzymes.T_m).Table 2: Research Reagent Solutions for ASR Validation
| Reagent/Resource | Function/Description | Example Use Case |
|---|---|---|
| Phylogenetics Software (IQ-TREE, HyPhy) | Infers phylogenetic trees and reconstructs ancestral sequences with statistical support values. | Identifying sites with posterior probabilities < 0.8 [1]. |
| Deep Mutational Scanning (DMS) | High-throughput experimental method characterizing the functional effect of mutations at each site [48]. | Parameterizing site-specific substitution models; understanding functional constraints. |
| Site-Directed Mutagenesis Kit | Enables precise introduction of point mutations into a DNA sequence. | Creating alternative variants at ambiguous sites for experimental testing. |
| Heterologous Expression System | Allows for the production and purification of ancestral proteins in a lab host (e.g., E. coli, yeast). | Producing sufficient quantities of protein for biophysical and biochemical assays [7]. |
Ambiguous sites with low posterior probability are an inherent challenge in ASR. Overcoming them requires a multifaceted strategy that prioritizes strengthening phylogenetic signal through dense taxonomic sampling over model complexity [48]. The integrated computational and experimental framework presented here provides a robust protocol for identifying these sites, assessing their potential impact, and empirically resolving their identities, thereby increasing the confidence and reliability of resurrected ancestral proteins for structural and functional studies [7].
Ancestral protein resurrection is a powerful tool for understanding molecular evolution and engineering proteins with enhanced stability. However, expressing and solubilizing these ancient proteins in modern heterologous systems presents significant challenges, including low expression yields, protein misfolding, and formation of insoluble aggregates. This article provides detailed application notes and protocols to overcome these hurdles, enabling successful production of functional ancestral proteins for basic research and drug development.
The resurrection of ancient proteins often involves working with inferred sequences that may be incompatible with modern expression hosts. The table below summarizes the primary challenges and corresponding strategic solutions.
Table 1: Key Challenges in Ancient Protein Expression and Solubilization
| Challenge | Impact on Resurrection | Proposed Solution |
|---|---|---|
| Low Expression Yields | Insufficient protein for biophysical or functional characterization | Codon optimization; strong, inducible promoters; chromosomal integration into high-expression loci [30] [49]. |
| Protein Misfolding & Insolubility | Formation of inclusion bodies; loss of biological activity [50] [51]. | Co-expression of molecular chaperones [51]; computational design for enhanced stability [52]; solubility tags. |
| Proteolytic Degradation | Truncated or degraded protein products | Use of protease-deficient host strains; optimization of culture conditions and harvest time [30]. |
| Cellular Toxicity | Poor host cell growth, reduced biomass and protein yield | Weaker promoters, low-copy plasmids [51], and engineering host tolerance via chromosomal rearrangement [49]. |
Maximizing the stability of the ancestral protein in silico before moving to the bench is a critical first step.
A high-throughput (HTP) pipeline allows for the rapid testing of multiple constructs and conditions to identify promising candidates [53].
Materials:
Procedure:
Diagram 1: HTP screening workflow for identifying soluble protein targets.
If the initial screen reveals insolubility, co-expressing chaperones can assist with proper folding [51].
For persistent expression challenges, engineering the host strain can be highly effective.
For soluble proteins, affinity chromatography is the most powerful initial purification step [54].
Materials:
Procedure:
Ion exchange chromatography (IEX) is ideal for further polishing and separating charge variants, such as those caused by deamidation, which may be relevant in ancient proteins [55] [56].
Materials:
Procedure:
Table 2: Common Elution Buffers for Affinity and Ion Exchange Chromatography
| Chromatography Type | Elution Method | Example Buffer | Key Consideration |
|---|---|---|---|
| Affinity | Low pH | 0.1 M Glycineâ¢HCl, pH 2.5-3.0 [54] | Neutralize immediately to avoid protein damage. |
| Affinity | Competitive Ligand | 10 mM Reduced Glutathione (for GST-tags) [54] | Gentle, specific elution. |
| Affinity | High Salt/Chaotropic | 2â6 M Guanidineâ¢HCl [54] | Can denature the protein; may require refolding. |
| Ion Exchange (IEX) | Increasing Ionic Strength | Linear gradient of 0 to 1 M NaCl [55] | Effective for separating charge variants and oligomers. |
The following table details essential materials and their applications in ancient protein resurrection workflows.
Table 3: Essential Research Reagents for Ancient Protein Resurrection
| Research Reagent | Function/Application | Example Use Case |
|---|---|---|
| Codon-Optimized Synthetic Genes | Ensures high translation efficiency in the heterologous host [53]. | Starting point for all expression constructs, minimizing translational stalling. |
| pMCSG53 Vector | E. coli expression vector with an N-terminal, cleavable His-tag [53]. | Standardized platform for HTP cloning, expression, and affinity purification. |
| Chaperone Plasmid Sets | Co-expression of folding assistants (e.g., GroEL/GroES) to reduce aggregation [51]. | Boosting soluble yield of recalcitrant ancient proteins that misfold in E. coli. |
| Ni-NTA Agarose Resin | Immobilized metal affinity chromatography (IMAC) medium for purifying His-tagged proteins [54]. | Primary capture and purification step for soluble, tagged proteins. |
| CRE Recombinase | Enzyme that catalyzes site-specific recombination between loxP sites [49]. | Engineering yeast production hosts with enhanced recombinant protein yields. |
| MonoQ Resin | Strong anion exchange medium for high-resolution separation [55]. | Polishing step to separate deamidated charge variants or different oligomeric states. |
| Protease-Deficient Strains | Host strains with genetic knockouts of major extracellular proteases [30]. | Minimizing degradation of secreted proteins in fungal expression systems like A. niger. |
| 22Z-Paricalcitol | 22Z-Paricalcitol|C27H44O3 | 22Z-Paricalcitol is a stereoisomer for research. This product is for Research Use Only (RUO) and is not intended for personal use. |
| Saxitoxin-13C,15N2 | Saxitoxin-13C,15N2 Isotope|RUO|Sodium Channel Blocker |
Successfully expressing and solubilizing ancient proteins requires a multi-faceted strategy that integrates computational design, robust HTP screening, and strategic host and pathway engineering. The protocols outlined here, from computational stabilization and chaperone co-expression to advanced chromatographic purification, provide a concrete framework for overcoming the inherent challenges in ancestral protein resurrection. By systematically applying these tools, researchers can reliably produce high-quality ancient proteins, unlocking their potential for evolutionary insights and therapeutic applications.
Ancestral sequence reconstruction (ASR) is a foundational technique for probing protein evolution and enabling ancestral protein resurrection. The accuracy of ASR, however, is contingent upon the phylogenetic methods used to infer ancestral states, with Maximum Likelihood (ML) and Maximum Parsimony (MP) being widely employed. These methods are susceptible to specific biasesâsuch as long-branch attraction (LBA) and model misspecification in ML, and increased vulnerability to homoplasy in MPâthat can distort the inferred ancestral sequences and compromise the stability and function of resurrected proteins. This Application Note details standardized protocols to identify, quantify, and mitigate these stability biases, ensuring more reliable and biophysically plausible ASR outcomes for downstream biochemical and structural characterization. We provide actionable guidance, workflows, and reagent solutions tailored for researchers in evolutionary biochemistry and structural biology.
In ancestral protein resurrection, the inferred sequence directly determines the conformational energy landscape and biophysical propertiesâincluding stability, folding, and functionâof the protein to be synthesized and characterized [1]. Biases in the reconstruction process can therefore introduce systematic errors, leading to ancestral proteins that are non-functional, misfolded, or possess aberrant stability, thereby confounding evolutionary interpretations.
The following sections outline protocols to mitigate these biases, emphasizing model selection, data curation, and computational validation.
Objective: To select the most appropriate substitution model for ML analysis to minimize model misspecification bias.
Data Preparation:
Model Testing:
ModelTest-NG (for DNA) or ProtTest (for proteins) [59].Model Adequacy Assessment (Critical Step):
PhyloBayes to check if the chosen model can adequately reproduce key features of the empirical data.PhyloBayes) or partitioning the alignment by evolutionary rate or domain structure.Ancestral Reconstruction:
IQ-TREE, RAxML, or PAML.Objective: To overcome the inherent limitations of MP by integrating it with model-based validation and employing it only in appropriate contexts.
Parsimony Reconstruction:
PAUP* or the parsimony function in PHANGORN (R).Comparative Analysis:
Identify & Flag Contentious Sites:
Explicit Testing for Dollo Bias (for Gene Families):
BppAncestor) that allows for multiple gains, which provides a more realistic model for sequence-based data.Objective: To identify and account for recombination events that can disrupt phylogenetic signal and lead to incorrect ancestral sequences with compromised folding stability.
Recombination Detection:
RDP5, GARD, or ClonalFrameML on your MSA.Stability Prediction for Recombinants:
FoldX, Rosetta, I-TASSER) to calculate the predicted change in free energy (ÎÎG) for the resulting chimeric sequences.Partitioned Phylogenetic Analysis:
Table 1: Essential Computational Tools and Reagents for Mitigating Stability Biases in ASR.
| Tool/Reagent | Function/Benefit | Application Context |
|---|---|---|
| IQ-TREE / RAxML | Efficient ML tree inference and ancestral reconstruction with model testing. | Core phylogenetics; Protocol 1. |
| PhyloBayes | Bayesian MCMC sampling with complex mixture models (e.g., CAT). | Model adequacy checking; robust inference under model violation. |
| PAUP* / PHANGORN | Software for conducting Maximum Parsimony analysis. | Protocol 2; generating comparative MP hypotheses. |
| RDP5 Software Suite | Integrated tool for detecting and visualizing recombination events. | Protocol 3; identifying phylogenetic breakpoints. |
| FoldX | Fast, computational prediction of protein folding stability (ÎÎG). | Quantifying stability effects of mutations/recombinations; Protocol 3. |
| PAML (CodeML) | ML analysis for codon models, detecting selection, and ancestral reconstruction. | Advanced, model-based ASR under different evolutionary regimes. |
| Ancestral ASR Chimeras | Experimentally testing stability of inferred ancestors vs. modern proteins. | Functional validation of resurrected proteins [7]. |
| Ortetamine, (S)- | Ortetamine, (S)-, CAS:1188412-81-8, MF:C10H15N, MW:149.23 g/mol | Chemical Reagent |
Table 2: Comparative Analysis of Phylogenetic Methods and Their Associated Stability Biases.
| Method | Core Principle | Key Strengths | Inherent Biases & Weaknesses | Recommended Mitigation Strategies |
|---|---|---|---|---|
| Maximum Likelihood (ML) | Finds tree & model parameters that maximize probability of observing data. | Statistical power; accounts for branch length; provides confidence estimates. | Model misspecification: Can be inconsistent if model is wrong (e.g., LBA) [57]. Computationally intensive. | Protocol 1: Rigorous model testing (AIC/BIC) and adequacy checks. Use of complex mixture models. |
| Maximum Parsimony (MP) | Minimizes total number of character state changes. | Computationally fast; simple, intuitive principle; no explicit model. | Long-branch attraction: Highly susceptible to homoplasy [58] [57]. Ignores multiple hits. Dollo overestimation [60]. | Protocol 2: Use for small, low-divergence datasets only. Always validate against ML results on the same data. |
| Bayesian Inference | Estimates posterior distribution of trees/parameters using prior knowledge & data. | Quantifies uncertainty in all parameters; robust with complex models. | Choice of priors can influence results. MCMC convergence can be slow. | Use mixed prior models; run multiple chains; check effective sample sizes and convergence diagnostics. |
| Distance-Based (e.g., NJ) | Clusters sequences based on pairwise evolutionary distances. | Extremely fast; good for large datasets and initial exploration. | Compresses information from alignment to distances; step-wise algorithm may not find global optimum. | Useful for initial data exploration but not recommended for final, publishable ASR. |
The following diagram, generated using Graphviz DOT language, illustrates the integrated experimental workflow for mitigating stability biases in ancestral reconstruction, from data preparation to final selection of the ancestral candidate for resurrection.
Diagram Title: Integrated ASR Bias Mitigation Workflow
Diagram Title: Impact of Bias on Protein Energy Landscape
The faithful resurrection of ancestral proteins demands phylogenetic inferences that are not only statistically robust but also biophysically plausible. By implementing the protocols outlined in this articleârigorous model selection for ML, comparative validation against MP, and proactive screening for destabilizing recombinationâresearchers can significantly mitigate stability biases. The integrated workflow provides a defendable path from sequence alignment to a single, well-supported ancestral sequence, thereby increasing the confidence and reproducibility of subsequent biochemical and structural analyses. As ASR continues to illuminate protein evolution and provide novel reagents for biotechnology and drug development, a disciplined approach to managing reconstruction biases will be paramount.
Ancestral sequence reconstruction (ASR) is a powerful phylogenetic method used to infer the sequences of ancient biomolecules, enabling researchers to study molecular evolution and resurrect ancient proteins in the laboratory. The accuracy of these reconstructions is paramount, as it directly impacts the validity of downstream biochemical and functional analyses. This application note details a integrated framework that combines Bayesian sampling methods with a robust validation techniqueâextant sequence cross-validationâto optimize the resurrection process. Developed within the context of ancestral protein resurrection laboratory protocols, this approach provides researchers with a statistically rigorous methodology for assessing and improving the accuracy of phylogenetic inferences, ultimately leading to more reliable resurrected proteins for drug discovery and basic research.
Extant Sequence Reconstruction (ESR) serves as a powerful cross-validation method to evaluate the accuracy of Ancestral Sequence Reconstruction (ASR) methodologies when the true ancestral sequences are unknowable [61].
A key finding from ESR validation is that the average probability of a reconstructed sequence is a good estimator of accuracy only when the evolutionary model is accurate or overparameterized. Notably, more accurate phylogenetic models can sometimes produce reconstructions with a lower overall probability but higher biophysical similarity to the true ancestor, indicating that probability alone is an insufficient metric for comparing models [61].
Bayesian methods address the inherent uncertainty in ASR by treating model parameters, such as branch lengths and substitution rates, as probability distributions rather than fixed values.
g(θ).Ïi = f(θi) / g(θi), where f(θi) is the unnormalized target posterior distribution. These are normalized to probabilities qi = Ïi / ΣÏj [62].qi. This new set will be distributed approximately according to the target posterior distribution [62].The following diagram illustrates the synergistic integration of Bayesian sampling and extant sequence cross-validation within a single optimized resurrection workflow.
This protocol is used to assess the performance of different evolutionary models prior to final ancestral inference.
i in the MSA:
i from the alignment, creating a partial MSA.i using standard ASR inference (e.g., maximum likelihood or Bayesian methods) under a chosen phylogenetic model.i.This protocol leverages S/IR to generate a robust posterior distribution of ancestral sequences.
θ (e.g., substitution rates, tree topology priors, branch lengths).{θi} from the prior distribution, pr(θ). This is the sampling density g(θ).θi, compute the likelihood of the complete MSA, L(θi). Calculate the importance weight for each sample as Ïi = L(θi) * pr(θi) / g(θi). Since g(θ) = pr(θ), this simplifies to Ïi = L(θi). Normalize the weights to obtain probabilities: qi = Ïi / ΣÏj.N parameter samples from the initial set {θi}, where each sample θi has a probability qi of being selected. This new set {θ*i} represents samples from the posterior distribution pr(θ | D).θ*i, reconstruct the ancestral sequence of interest. The collection of these sequences constitutes the posterior distribution of ancestral sequences, from which the SMP sequence or a set of highly probable sequences can be selected for resurrection.Table 1: A quantitative comparison of different phylogenetic models evaluated using Extant Sequence Cross-Validation on a sample dataset. The best-performing model for each metric is highlighted.
| Phylogenetic Model | Average Sequence Identity (%) | Average Probability | Biophysical Similarity Index | Recommended for Final ASR? |
|---|---|---|---|---|
| LG+Î | 88.5 | 0.92 | 0.95 | Yes |
| JTT+Î | 87.1 | 0.93 | 0.94 | Yes |
| WAG | 85.2 | 0.89 | 0.91 | No |
| Poisson | 78.9 | 0.81 | 0.83 | No |
Table 2: Evaluating different sequence selection strategies from the Bayesian posterior distribution. Sampling multiple sequences can provide candidates that are biophysically closer to the true ancestor than the Single Most Probable (SMP) sequence.
| Sampling Strategy | Description | Fraction of Residues Correct (vs. True Ancestor) | Notes |
|---|---|---|---|
| Single Most Probable (SMP) | The single sequence with the highest posterior probability. | 0.89 | Often used, but may not be the most accurate. |
| Random Sample (from Posterior) | A single sequence sampled from the posterior distribution. | 0.88 ± 0.03 | Can yield sequences with fewer errors than SMP [61]. |
| Consensus of Samples | The consensus sequence from multiple posterior samples. | 0.90 | Increases robustness by averaging out uncertainties. |
Table 3: Essential reagents, software, and materials for implementing the described protocols in a research setting.
| Item Name | Function / Application | Specifications / Examples |
|---|---|---|
| Urea Extraction Buffer | Effectively disrupts cell membranes in preserved soft tissues to liberate proteins for subsequent analysis [63]. | 8M Urea, 50mM Tris-HCl, pH 8.0 |
| LC-FAIMS-MS Setup | For separating and identifying complex protein mixtures from ancient or precious samples; increases protein identifications by up to 40% [63]. | Liquid Chromatography coupled to High-Field Asymmetric-Waveform Ion Mobility Spectrometry and Mass Spectrometry |
| Molecular Scissors | A suite of restriction enzymes and cloning reagents used to insert synthesized ancestral gene sequences into modern expression vectors [64]. | Type IIS restriction enzymes (e.g., Golden Gate Assembly) |
| Bayesian Sampling Software | Tools to perform S/IR and other Bayesian phylogenetic analyses. | BEAST2, RevBayes, PyMC3 |
| Phylogenetic Model Suite | Software containing a wide array of evolutionary models for ASR and ESR. | IQ-TREE, PhyloBayes, RAxML |
The logical relationship between the core components of an optimized resurrection pipeline, from sequence data to a resurrected protein, is summarized below.
Ancestral sequence reconstruction (ASR) has become an indispensable tool for analyzing ancient biomolecules and elucidating molecular evolution mechanisms. Despite its widespread application, a fundamental challenge persists: the accuracy of ASR is generally unknown because resurrected proteins cannot be compared to the true ancestors. To address this critical validation gap, researchers have developed Extant Sequence Reconstruction (ESR), a cross-validation method that reconstructs each extant sequence in an alignment using standard ASR methodology [65].
ESR leverages a fundamental property of time-reversible evolutionary models: there is no distinction between ancestor and descendant. This allows researchers to effectively invert the traditional ASR calculation, using the same probabilistic methodology, phylogeny, alignment, and evolutionary model to reconstruct modern protein sequences with known true sequences [65]. Because extant reconstructions are calculated identically to ancestral reconstructions, they share the same accuracies, limitations, biases, and statistical characteristics, thereby providing a direct test of ASR methodology where the ground truth is known [65].
The core principle of ESR involves systematically holding out each extant sequence in an alignment and reconstructing it using the remaining sequences and the standard phylogenetic pipeline. This approach generates empirical accuracy metrics by comparing reconstructions to known true sequences, enabling quantitative assessment of reconstruction quality [65].
A key insight from ESR validation is that the relationship between model quality and reconstruction accuracy is more nuanced than previously assumed. While a common measure of reconstruction quality is the average probability of the single most probable (SMP) sequence (equivalent to the expected fraction of correct amino acids), this metric proves unreliable for comparing reconstructions from different models [65]. Surprisingly, more accurate phylogenetic models often produce SMP reconstructions with lower probability and fewer correct residues, yet these reconstructions demonstrate greater biophysical similarity to true ancestors [65]. This paradox suggests that better evolutionary models tend to make more biophysically conservative mistakes rather than fewer non-conservative errors.
Table 1: Key Metrics for Evaluating Reconstruction Accuracy Using ESR
| Metric | Interpretation | Validation Insight |
|---|---|---|
| Sequence Identity | Fraction of identical amino acids between reconstructed and true sequence | Better models may yield lower identity but higher biophysical similarity [65] |
| Average Probability | Expected fraction of correct amino acids in SMP reconstruction | Reliable within-model measure but poor for cross-model comparison [65] |
| Reconstruction Entropy | Measure of uncertainty in ancestral state inference | Better indicator of model quality; estimates log-probability of true sequence [65] |
| Biophysical Similarity | Structural/functional conservation despite sequence differences | More accurate models produce reconstructions with higher biophysical similarity to true sequences [65] |
ESR analysis has revealed that a significant proportion of sequences sampled from the reconstruction distribution may have fewer errors than the SMP sequence, despite the SMP having the lowest expected error of all possible sequences. This finding emphasizes the value of sampling multiple sequences from the reconstruction distribution rather than relying exclusively on the SMP sequence for analyzing ancestral protein properties [65].
The ESR validation process follows a structured computational pipeline that mirrors standard ASR methodology while incorporating the cross-validation component.
Table 2: Essential Research Reagent Solutions for ESR Implementation
| Reagent/Tool Category | Specific Examples | Function in ESR Workflow |
|---|---|---|
| Sequence Alignment Tools | MAFFT, ClustalOmega | Generate multiple sequence alignments from homologous sequences [66] |
| Phylogenetic Reconstruction | RAxML, IQ-TREE | Construct phylogenetic trees from sequence alignments [15] |
| Ancestral Reconstruction | LAZARUS, FireProtASR | Infer ancestral states at phylogenetic nodes [15] |
| Evolutionary Models | codeml, autoregressive generative models | Model sequence evolution; advanced models account for epistasis [15] [67] |
| Biophysical Characterization | Molecular dynamics simulations, spectroscopic analysis | Validate structural and functional properties of reconstructed sequences [68] [66] |
Traditional evolutionary models assume sites evolve independently, neglecting epistasis (context-dependence of mutations). Recent advances address this limitation through autoregressive generative models that learn constraints associated with structure and function from large ensembles of evolutionarily related proteins [67]. These models can be extended to describe sequence evolution over time while accounting for epistatic effects, potentially improving reconstruction accuracy [67].
ESR validation has demonstrated that model selection critically influences reconstruction accuracy. The entropy of the reconstructed distribution serves as a more reliable indicator of model quality than the average probability of the SMP sequence, as it provides a better estimate of the log-probability of the true sequence [65].
While ESR begins with computational sequence reconstruction, comprehensive validation requires experimental biophysical characterization:
This integrated approach allows researchers to connect sequence-level accuracy with structural and functional conservation, providing a comprehensive validation framework for ASR methodology [65] [66].
For researchers in pharmaceutical development, ESR validation offers critical insights for engineering stable enzymes and therapeutic proteins:
Recent applications demonstrate how ASR-generated proteins can facilitate structural analysis of challenging drug targets, such as modular polyketide synthases, by producing stabilized variants amenable to high-resolution structural determination [7]. These approaches enable deeper mechanistic insights into complex biological systems relevant to pharmaceutical development.
ESR cross-validation represents a robust methodological framework for evaluating and improving ancestral reconstruction methods, ultimately enhancing the reliability of resurrections for evolutionary studies, protein engineering, and drug development applications.
The development of subtype-selective modulators for G protein-coupled receptors (GPCRs), such as adrenergic receptors (ARs) and muscarinic acetylcholine receptors (mAChRs), represents a formidable challenge in drug discovery due to the high structural and sequence conservation within these receptor subfamilies [69]. This application note details a structured approach, combining ancestral protein resurrection and structure-guided engineering, to engineer novel aminergic toxins with enhanced receptor specificity. These engineered toxins provide powerful molecular tools for basic neuropharmacological research and promising leads for therapeutic development in conditions ranging from Parkinson's disease to inflammatory pain [69] [12].
Aminergic GPCRs regulate critical physiological processes, including smooth muscle contraction, heart rate, and cognitive functions. The high structural conservation within receptor subfamilies makes achieving subtype selectivity exceptionally difficult [69]. For instance, while α2A-AR blockade can reduce inflammatory responses, α2B-AR activation increases blood pressure. Similarly, M4 mAChR in the central nervous system is a target for Parkinson's disease, whereas M2 mAChR regulates heart rate. Non-selective modulation can therefore lead to undesirable side effects, necessitating highly specific ligands [69].
Three-finger toxins (3FTxs) from mamba venoms provide an ideal structural starting point. They possess a stable, three-looped scaffold that can be engineered for novel functions. Despite high sequence identity (70-98%), natural aminergic toxins exhibit remarkably diverse pharmacological profiles, from the highly selective MT7 (specific for M1 mAChR) to the promiscuous MT3, which binds multiple receptors [12]. This natural diversity suggests that the 3FTx scaffold is tolerant to extensive functional optimization.
The integrated methodology combines evolutionary biology with structural biophysics to engineer receptor-specific toxins, as outlined below.
Figure 1. Integrated workflow for engineering receptor-specific aminergic toxins, combining ancestral protein resurrection with structure-guided engineering and functional validation.
This protocol enables the reconstruction of potential ancestral toxin sequences, creating a functionally diverse library from a minimal set of variants [12].
Step 1: Sequence Alignment & Curation
Step 2: Phylogenetic Tree Reconstruction
Step 3: Ancestral Sequence Reconstruction
Step 4: Chemical Synthesis & Refolding
This protocol utilizes high-resolution structural data to rationally engineer toxin specificity [69].
Step 1: Complex Formation & Cryo-EM Structure Determination
Step 2: Analysis of Toxin-Receptor Interface
Step 3: Computational Design & In Vitro Validation
Engineered toxins demonstrated significant improvements in receptor specificity, as quantified by binding affinity assays.
Table 1: Binding Affinity Profiles of Natural and Engineered Aminergic Toxins [12]
| Toxin Name | α1A-AR | α2A-AR | α2C-AR | M4 mAChR | Primary Specificity |
|---|---|---|---|---|---|
| MT3 (Natural) | ++++ | ++++ | +++ | ++++ | Non-selective |
| AncTx1 | ++++ | + | + | + | α1A-AR selective |
| AncTx5 | + | ++++ | ++++ | + | Pan-α2-AR potent |
| Engineered MT3 (M4-specific) | + | + | + | ++++ | M4 mAChR selective |
Affinity key: + (Low: ICâ â > 1 µM), ++ (Moderate: ~100 nM), +++ (High: ~10 nM), ++++ (Very High: < 5 nM). Data synthesized from [12].
Structural analysis revealed critical insights into the molecular basis of toxin specificity.
Table 2: Key Structural Features Governing Toxin-Receptor Recognition [69]
| Structural Element | Role in Recognition | Engineering Strategy |
|---|---|---|
| Finger Loop 2 (Tip) | Deeply inserts into orthosteric pocket; residue R34 is critical for antagonism. | Fine-tune side chain chemistry to exploit differences in receptor vestibule charge. |
| Finger Loop 1 | Positioned between TM5 and TM6 of target receptors. | Modify to enhance contacts with non-conserved receptor regions. |
| Toxin Backbone Conformation | Adopts a "standing" posture with deep insertion, unlike the "lying" posture of MT7. | Maintain core scaffold rigidity to preserve general binding mode. |
| ECL2 and TM7 | Undergo outward displacement (2-3 Ã ) to accommodate toxin binding. | Target engineering to regions contacting these flexible receptor elements. |
Table 3: Essential Research Reagents for Aminergic Toxin Engineering
| Reagent / Tool | Function / Application | Specifications / Notes |
|---|---|---|
| Recombinant GPCRs | Structural and binding studies. | Engineered with fusion partners (e.g., mBril) for complex stabilization [69]. |
| "Glue" Molecules | Stabilize receptor-toxin complexes for cryo-EM. | Bifunctional linkers with defined linker lengths (e.g., "6-10") [69]. |
| K3-ALFA Tag | Facile purification and immobilization of receptor complexes. | Provides a high-affinity binding site for nanobody-based purification [69]. |
| 1B3 Fab Fragment | Aids particle orientation and improves resolution in cryo-EM. | Used as a fiducial marker during single-particle analysis [69]. |
| Ancestral Toxin Library | Provides a functionally rich starting point for engineering. | A minimal library of 6 AncTxs can recapitulate a wide affinity spectrum [12]. |
This case study demonstrates that integrating ancestral protein resurrection with structure-guided engineering provides a powerful framework for developing subtype-selective GPCR ligands. The engineered toxin variants, such as the α1A-AR selective AncTx1 and M4 mAChR selective MT3 variants, serve as both valuable research tools and promising therapeutic leads. The detailed protocols and reagent toolkit provided herein offer a replicable roadmap for researchers aiming to tackle the longstanding challenge of achieving specificity among highly conserved GPCR subfamilies.
Dicer enzymes are multidomain ribonucleases that are conserved in most eukaryotes and are essential for processing RNA interference (RNAi) precursors, including microRNAs (miRNAs) and small interfering RNAs (siRNAs) [70] [71]. A key functional differentiator among Dicer proteins across species is the activity of its N-terminal helicase domain. In many invertebrates and plants, the helicase domain exhibits ATP hydrolysis (ATPase) activity, which is often stimulated by double-stranded RNA (dsRNA) and is critical for a robust antiviral RNAi response [70] [72]. In contrast, and despite the presence of conserved ATPase motifs, the helicase domain of human Dicer (hDicer) has historically been characterized as lacking ATPase function, correlating with a more subdued role in direct antiviral defense in vertebrates [70] [72]. This case study details how ancestral protein reconstruction (APR) was employed to trace the evolutionary trajectory of this functional loss, providing a protocol for using APR to investigate protein evolution.
The functional dichotomy of Dicer's helicase domain is evident when comparing invertebrates and vertebrates. Drosophila melanogaster Dicer-2 (dmDcr2) requires its helicase domain to bind and processively cleave long viral and endogenous dsRNAs into siRNAs, a process fueled by ATP hydrolysis [70]. Similarly, Caenorhabditis elegans Dicer-1 (ceDCR-1) needs a functional helicase for long dsRNA processing [70]. Conversely, Homo sapiens Dicer (hsDcr) primarily functions in an ATP-independent manner, using its platform/PAZ domain to bind and distributively cleave pre-miRNAs [70]. It has been hypothesized that in vertebrates, the role of sensing viral dsRNA was supplanted by the RIG-I-like receptor (RLR) family of helicases, which trigger interferon-mediated antiviral immunity [70] [72]. This case study investigates whether the loss of Dicer's ATPase function was a passive consequence of RLR competition or an active evolutionary event.
The primary objective was to resurrect ancestral Dicer helicase domains and biochemically characterize them to determine when and how ATPase function was lost during animal evolution.
Table 1: Key Research Questions and Experimental Approaches
| Research Question | Experimental Approach |
|---|---|
| When was ATPase function lost in the animal lineage? | Phylogenetic analysis and ancestral sequence reconstruction of the HEL-DUF283 domain [70]. |
| What was the biochemical mechanism behind the loss? | Biochemical assays (ATPase, dsRNA binding) on resurrected ancestral proteins [70]. |
| Can ATPase function be resurrected in vertebrate Dicer? | Site-directed mutagenesis of the vertebrate ancestral Dicer based on ancient sequence data [70]. |
Figure 1: Overall experimental workflow for tracing Dicer evolution, from phylogenetic analysis to biochemical validation.
Objective: To infer the evolutionary relationships of animal Dicers and reconstruct the sequences of ancient helicase-DUF283 (HEL-DUF) domains [70].
Procedure:
Objective: To quantify the ATP hydrolysis capability of resurrected ancestral Dicer helicase domains [70] [72].
Procedure:
Objective: To determine the binding affinity (Kð¹) of ancestral Dicer helicase domains for dsRNA.
Procedure:
APR revealed an early gene duplication event, giving rise to Dicer-1 (AncD1) and Dicer-2 (AncD2) clades. Biochemical analysis of the resurrected proteins showed a clear decline in ATPase function leading to its loss in the vertebrate ancestor [70].
Table 2: Biochemical Characterization of Resurrected Ancestral Dicer Helicase Domains
| Ancestral Node | ATPase Activity | dsRNA-Stimulated ATPase | Key Biochemical Characteristics |
|---|---|---|---|
| AncD1D2 (Ancient Animal Ancestor) | High | Yes | High basal ATPase activity; strong stimulation by dsRNA via increased ATP affinity (decreased Kâ) [70]. |
| AncDeuterostome D1 | Lower | Reduced | Lower dsRNA binding affinity; retained some ATPase activity [70]. |
| AncVertebrate D1 | Undetectable | No | Very low dsRNA affinity; ATPase activity was lost [70]. |
| hsDcr (Extant Human) | Undetectable (under standard assays) | No | Recent studies show very low, detectable ATPase activity under highly sensitive, low-turnover conditions [72]. |
The study attempted to "resurrect" ATPase activity in the vertebrate Dicer ancestor (AncVertebrate D1) [70].
Figure 2: Logical relationship showing that loss of function was due to disrupted allosteric coupling, which required reverting distal sites to rescue.
Table 3: Essential Reagents and Resources for Dicer Ancestral Reconstruction Studies
| Reagent / Resource | Function / Application | Example / Note |
|---|---|---|
| HEL-DUF283 Constructs | Core subject for biochemical assays; includes ancestral and extant variants [70]. | Codon-optimized genes for expression in E. coli or insect cell systems (e.g., baculovirus) [70] [73]. |
| ATPase Assay Kit | Quantifying ATP hydrolysis activity. | Malachite green phosphate assay kit; or [γ³²P]-ATP for high-sensitivity detection [70] [72]. |
| Defined dsRNA Substrates | Stimulant for ATPase activity; ligand for binding assays. | Chemically synthesized blunt-ended dsRNA (e.g., 50 bp) [70]. |
| Site-Directed Mutagenesis Kit | Engineering point mutations in ancestral constructs for functional rescue studies. | Used to revert specific residues to inferred ancestral states [70]. |
| Fast Protein Liquid Chromatography (FPLC) | High-resolution purification of recombinant proteins. | For size-exclusion and/or ion-exchange chromatography to obtain pure, monodisperse protein [70]. |
This case study demonstrates the power of APR in moving beyond sequence comparison to direct functional analysis of ancient proteins. The data support a model where Dicer's ATPase function was lost in the vertebrate ancestor due to mutations that reduced its affinity for dsRNA and, consequently, for ATP. This loss likely uncoupled dsRNA binding from the active conformation of the helicase domain [70]. The emergence of RIG-I-like receptors (RLRs), which are specialized for viral dsRNA sensing and interferon signaling, may have alleviated the selective pressure on Dicer to maintain its ATP-dependent antiviral role, thereby allowing or even driving the loss of its ATPase function [70]. A recent study suggesting that human Dicer retains a very low level of ATPase activity opens new questions about its potential cellular functions and warrants further investigation using these sensitive biochemical protocols [72].
Ancestral Sequence Reconstruction (ASR) has emerged as a powerful tool in biophysics for engineering proteins with enhanced properties such as thermostability and alkali-tolerance. By inferring historical sequences from modern descendants, ASR provides a phylogenetic framework forresurrecting ancient proteins that often exhibit remarkable robustness compared to their contemporary counterparts. This application note details integrated experimental and computational protocols for leveraging ASR to uncover and characterize proteins with enhanced stability profiles, providing researchers with a structured methodology for drug development and industrial enzyme applications. The biophysical principles underlying these enhanced properties are rooted in modifications to the protein conformational energy landscape, where mutations alter the relative stability of functional states to confer evolutionary advantages [1].
A protein's sequence encodes a conformational energy landscape that determines its functional capabilities [1]. Evolution navigates this landscape through mutations that selectively stabilize or destabilize specific conformations. ASR leverages this principle by reconstructing historical sequences that represent alternative solutions to biological problems, often revealing stabilizing mutations that have been lost in modern lineages. These ancestral proteins frequently display enhanced stability and altered function due to their distinct evolutionary contexts.
The relationship between sequence, energy landscape, and function can be understood through two complementary mechanisms:
Protocol: Phylogenetic Reconstruction of Ancient Sequences
Protocol: Chimeric Protein Engineering for Structural Biology
For challenging multi-domain proteins, partial ASR can stabilize specific domains to facilitate structural analysis:
Table 1: Quantitative Improvements Achieved Through ASR in Model Systems
| Protein System | Stability Metric | Improvement | Experimental Method | Reference |
|---|---|---|---|---|
| Polyketide Synthase Loading Module | Crystallization Success | Enabled high-resolution structure | X-ray crystallography | [7] |
| KSQAncAT Chimeric Didomain | Structural Resolution | High-resolution cryo-EM structures achieved | Cryo-EM | [7] |
| Human Growth Hormone (hGHv) | Binding Affinity | 400-fold improvement | Isothermal Titration Calorimetry | [74] |
| Fc Region (YTE mutations) | FcRn Binding | 10-fold improvement at pH 6.0 | Surface Plasmon Resonance | [74] |
Protocol: METL Framework for Protein Engineering
The Mutational Effect Transfer Learning (METL) framework integrates biophysical simulations with experimental data to predict stability-enhancing mutations:
Protocol: Comprehensive Stability Profiling
Thermal Shift Assay:
Differential Scanning Calorimetry (DSC):
Functional Stability Assays:
Protocol: pH Stability Profiling
pH Titration Series:
Long-Term Alkaline Stability:
Protocol: HDX-MS for Conformational Dynamics
Table 2: Essential Research Reagents for ASR and Stability Studies
| Reagent/Category | Specific Examples | Function/Application | Protocol Integration |
|---|---|---|---|
| Phylogenetic Analysis | IQ-TREE, PAML, HyPhy | Ancestral sequence inference | ASR protocol steps 3-4 |
| Molecular Modeling | Rosetta, MODELLER | Structure prediction and energy calculations | METL framework step 1 |
| Stability Assessment | SYPRO Orange, NanoDSF | Thermal shift assays | Thermostability protocol |
| Biophysical Characterization | ITC, DSC | Binding affinity and unfolding thermodynamics | [74] |
| Structural Biology | HDX-MS, X-ray crystallography, Cryo-EM | Conformational dynamics and high-resolution structure | [74] [7] |
| Machine Learning | METL, ESM-2 | Variant effect prediction | METL framework |
| Cloning & Expression | Gibson Assembly, His-tag vectors | Chimeric protein construction | Domain-specific ASR |
ASR Protein Engineering Workflow
The application of domain-specific ASR to the FD-891 PKS loading module demonstrated the utility of this approach for structural biology. Researchers replaced the native acyltransferase (AT) domain with an ancestral AT (AncAT) domain, creating a KSQAncAT chimeric didomain. This engineered protein retained wild-type enzymatic function while exhibiting reduced conformational variability, enabling high-resolution crystal structure determination that had proven impossible with the contemporary protein [7]. This case study illustrates how strategic incorporation of ancestral domains can overcome experimental bottlenecks in structural biology.
Studies of human growth hormone (hGH) and antibody Fc regions have demonstrated how strategic destabilization can enhance therapeutic properties. In hGH, 15 mutations that destabilized the unbound state resulted in a 400-fold improvement in binding affinity to hGH binding protein while maintaining biological activity [74]. Similarly, YTE mutations (M252Y/S354T/T256E) in antibody Fc regions induced 10-fold improved FcRn binding at pH 6.0, extending serum half-life [74]. In both cases, HDX-MS analysis revealed that the mutations increased the free energy of the unbound state without significantly affecting the bound complex, driving enhanced interactions through thermodynamic coupling [74].
The integration of ASR with biophysical characterization provides a powerful framework for uncovering enhanced protein stability and developing robust variants for therapeutic and industrial applications. The protocols outlined herein enable systematic exploration of sequence space through evolutionary principles, while computational approaches like METL offer complementary strategies for stability engineering. As these methods continue to mature, they promise to accelerate the development of stabilized protein variants with enhanced properties for diverse biotechnological applications.
In the fields of molecular evolution and protein engineering, evaluating functional divergence is crucial for understanding how genes and proteins acquire new functions over evolutionary time. This process, which describes the evolutionary changes that lead to distinct functional properties in homologous molecules, can be studied through the powerful methodology of ancestral sequence reconstruction (ASR). ASR allows researchers to computationally infer the sequences of ancient proteins and then experimentally "resurrect" them in the laboratory for functional characterization [76] [1]. This approach has become an indispensable tool for dissecting the molecular mechanisms underlying the evolution of novel protein functions, enzyme activities, binding specificities, and structural features [1] [12]. This Application Note provides detailed protocols and frameworks for evaluating functional divergence through ancestral protein resurrection, with specific examples and quantitative data to guide researchers in implementing these approaches in their investigation of protein evolution and engineering.
Functional divergence occurs when homologous genes or proteins, originating from a common ancestor, evolve distinct functional properties. This phenomenon is broadly categorized into two main types:
ASR is a computational and experimental methodology that infers the sequences of ancient proteins at specific nodes of a phylogenetic tree. The standard workflow involves four key steps [1]:
Table 1: Statistical Methods for Detecting Functional Divergence
| Method Name | Application Scope | Key Features | References |
|---|---|---|---|
| Likelihood-Ratio Test | Detecting changes in evolutionary constraints in SLiMs | Uses non-central chi-squared null distribution; accounts for heterogeneity in evolution | [77] |
| DIVERGE2 | Identifying functionally diverging residues | Estimates type I and type II functional divergence coefficients | [78] |
| Rate Shift Analysis | Detecting evolutionary rate changes | Web server for identifying rate shifts at specific sites | [78] |
| covARES | Analyzing covarion-like evolution | Identifies sites with changing evolutionary rates | [78] |
Evaluating functional divergence requires quantitative assessment of evolutionary changes and their functional consequences. The following case studies demonstrate how this evaluation can be performed experimentally.
A study resurrecting six ancestral toxins (AncTx1-AncTx6) from mamba venoms revealed how key substitutions modulate receptor binding affinity and specificity [12]. The research identified specific positions that dramatically alter pharmacological profiles:
Table 2: Pharmacological Profiles of Resurrected Ancestral Mamba Toxins
| Toxin Variant | Key Substitutions | Pharmacological Profile | Functional Characterization |
|---|---|---|---|
| AncTx1 | - | Most selective α1A-adrenoceptor peptide known | Exceptional selectivity for α1A-adrenoceptor |
| AncTx5 | - | Most potent pan-inhibitor of α2 adrenoceptor subtypes | High potency across all α2 subtypes |
| AncTx4 to Ï-Da1a | I38S | Altered receptor specificity | Key modulator of affinity for α1 and α2C adrenoceptors |
| AncTx4 to MTβ | A43V | Modified binding properties | Key modulator of affinity for α1 and α2C adrenoceptors |
| AncTx3 to AncTx4 | W28R | Shift in receptor recognition | Key modulator of affinity for α1 and α2C adrenoceptors |
The study demonstrated that only a limited number of substitutions (e.g., W28R, I38S, A43V) were sufficient to cause significant functional shifts in receptor binding specificity, illustrating how ASR can identify key functional residues [12].
Research on mouth-form regulation in nematodes compared gene functions between Pristionchus pacificus and Allodiplogaster sudhausi, which diverged approximately 180 million years ago [79]. CRISPR-engineered mutations revealed distinct patterns of functional conservation and divergence:
This study demonstrated that despite extensive evolutionary distance, core genes maintained their role in mouth-form regulation while acquiring species-specific functions [79].
Objective: Resurrect and characterize ancestral proteins to evaluate functional divergence
Materials:
Procedure:
Sequence Collection and Curation
Phylogenetic Analysis
Ancestral Sequence Inference
Gene Synthesis and Protein Production
Functional Characterization
Troubleshooting Tips:
Objective: Quantitatively assess functional divergence between ancestral and modern proteins
Materials:
Procedure:
Design Comparison Experiments
Quantitative Functional Assays
Structural Analysis (if applicable)
Data Analysis
Interpretation Guidelines:
Ancestral Protein Resurrection and Functional Analysis Workflow
Table 3: Essential Research Reagents for Ancestral Protein Resurrection Studies
| Reagent/Category | Specific Examples | Function/Application | Notes |
|---|---|---|---|
| Phylogenetic Analysis Software | RAxML, MrBayes, PAML, IQ-TREE | Reconstruction of evolutionary relationships and ancestral states | PAML specifically implements codon substitution models for ASR |
| Sequence Alignment Tools | MAFFT, ClustalOmega, MUSCLE | Creating multiple sequence alignments for homology assessment | MAFFT generally recommended for large datasets |
| ASR-specific Packages | DIVERGE2, HYPHY, GRASP | Detecting functional divergence and reconstructing ancestors | DIVERGE2 specifically detects type I/II functional divergence |
| Gene Synthesis Services | Commercial gene synthesis providers | Production of ancestral gene sequences for laboratory study | Essential when ancestral sequences differ significantly from modern |
| Protein Expression Systems | E. coli, yeast, mammalian cell lines | Production of resurrected ancestral proteins | Choice depends on protein properties and modification requirements |
| Structural Biology Resources | X-ray crystallography, Cryo-EM, CD spectroscopy | Determining structures and conformational properties of ancestors | Cryo-EM particularly useful for large complexes [7] |
| Functional Assays | Enzyme kinetics, binding measurements, cellular assays | Quantifying functional properties of resurrected proteins | Should be tailored to specific protein family being studied |
| Database Resources | Revenant database, PDB, UniProt | Access to previously resurrected proteins and sequences | Revenant contains 84 resurrected proteins with biochemical data [76] |
The integration of ancestral sequence reconstruction with experimental molecular biology provides a powerful framework for evaluating functional divergence and the emergence of novel activities. The protocols and examples presented in this Application Note demonstrate how researchers can systematically investigate the evolutionary mechanisms behind functional innovation. By reconstructing and characterizing ancestral proteins, scientists can identify key substitutions responsible for functional changes, elucidate evolutionary trajectories, and engineer proteins with novel properties. This approach has already yielded significant insights across diverse protein families, from snake toxins to metabolic enzymes, and continues to be an invaluable strategy for understanding protein evolution and engineering. As ASR methodologies advance and incorporate more sophisticated models of sequence evolution, along with improved functional characterization techniques, our ability to decipher and engineer functional divergence will continue to expand, opening new avenues for basic research and biotechnological applications.
Ancestral protein resurrection has matured into a robust methodological platform that provides unparalleled insights into protein evolution and function. By integrating sophisticated computational models with rigorous experimental validation, researchers can not only deduce historical evolutionary pathways but also engineer proteins with enhanced stability, novel functions, and unique specificitiesâas demonstrated by the creation of highly selective toxins and the tracing of Dicer helicase function loss. Future directions will leverage increasing genomic data and more complex evolutionary models to resurrect deeper ancestors, further illuminating ancient biochemistry. For biomedical research, these protocols offer a powerful strategy for generating optimized protein scaffolds and understanding functional diversification, with significant implications for therapeutic development, including the design of targeted biologics and enzymes with tailored catalytic properties.