This article provides a comprehensive analysis of Ribosomally synthesized and post-translationally modified peptides (RiPPs) as versatile platforms for peptide engineering.
This article provides a comprehensive analysis of Ribosomally synthesized and post-translationally modified peptides (RiPPs) as versatile platforms for peptide engineering. Targeting researchers and drug development professionals, we explore the foundational biosynthetic logic of RiPP pathways, detail cutting-edge methodologies for genome mining and heterologous expression, and address key challenges in optimizing these systems. By examining validation strategies and comparative analyses of engineered RiPPs, we synthesize a framework for exploiting RiPP modularity to create novel bioactive peptides with enhanced therapeutic potential, particularly against pressing global threats like antimicrobial resistance.
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a rapidly expanding superfamily of natural products with significant pharmaceutical potential, including applications as antibiotics, anticancer agents, and treatments for neurological disorders [1] [2]. The biosynthesis of RiPPs follows a conserved logic, beginning with the ribosomal synthesis of a precursor peptide that serves as the scaffold for all subsequent enzymatic modifications [3]. This precursor peptide is organized into distinct functional regionsâthe leader, core, and sometimes follower peptidesâthat work in concert to guide the maturation process. Understanding the precise roles of these domains is fundamental to leveraging RiPP pathways for peptide engineering, enabling researchers to rationally design novel bioactive compounds with enhanced stability and targeted activities [4]. The modular nature of this biosynthetic system, combined with advancing genome mining tools, positions RiPPs as ideal candidates for synthetic biology approaches to drug discovery.
The leader peptide is an N-terminal region that acts as a recognition signal and binding platform for the biosynthetic enzymes. It is typically removed proteolytically after modifications are complete [3] [1]. Its primary function is to ensure the correct enzymatic modifications are installed on the core peptide.
The core peptide is the C-terminal region that undergoes extensive post-translational modifications (PTMs) to become the mature RiPP natural product [4] [3]. The core peptide's sequence determines the final chemical structure and biological activity.
Some RiPP precursor peptides contain an additional C-terminal region, known as a follower peptide [4] [7]. Not universal across RiPP classes, the follower peptide's roles are less defined but involve recognition and proteolytic removal.
Table 1: Summary of RiPP Precursor Peptide Domains and Their Functions
| Peptide Domain | Location | Primary Function | Fate in Maturation | Example RiPP Classes |
|---|---|---|---|---|
| Leader Peptide | N-terminal | Enzyme recognition and activation [5]; Substrate delivery [5] | Proteolytically removed [3] | Lanthipeptides [6]; Lasso peptides [5]; Cyanobactins [3] |
| Core Peptide | C-terminal | Template for post-translational modifications; Determines final structure and activity [4] [3] | Becomes the mature natural product [3] | All RiPP classes |
| Follower Peptide | C-terminal (less common) | Recognition signal for biosynthetic machinery [1] | Proteolytically removed [7] | Bottromycins [1] |
Figure 1: Functional Workflow of RiPP Precursor Peptide Domains - This diagram illustrates how leader, core, and follower peptides interact with biosynthetic machinery to produce a mature RiPP.
This protocol, based on the study of the lanthionine synthetase DprM, tests whether a leader peptide is essential for enzyme activity and can be used to probe enzyme specificity [6].
Step 1: Gene Co-expression
dprM) and the gene for its putative precursor peptide (e.g., dprE1) into compatible expression plasmids. Ensure the precursor gene includes its native leader sequence.Step 2: Protein Expression and Purification
Step 3: Mass Spectrometric Analysis
This protocol details a colorimetric method to quantify leader peptidase activity and its dependence on the RRE domain, as demonstrated for lasso peptide biosynthesis [5].
Step 1: Protein and Substrate Preparation
FusB), its cognate RRE domain, and the precursor peptide or a synthetic leader peptide fused to a colorimetric tag like para-nitroanilide (pNA).Step 2: Reaction Setup
Step 3: Kinetic Measurement and Analysis
Table 2: Key Reagents for RiPP Functional Analysis
| Reagent / Tool | Function / Utility | Example Use Case |
|---|---|---|
| Codon-Optimized Synthetic Genes | Enables high-yield heterologous expression in hosts like E. coli [6] | Expression of dprM and dprE genes in E. coli for in vivo assays [6] |
| p-nitroanilide (pNA) Tagged Substrate | Provides a colorimetric readout for protease/peptidase activity [5] | Quantifying leader peptidase (FusB) kinetics in the presence and absence of the RRE domain [5] |
| RiPP Recognition Element (RRE) | Binds leader peptide with high affinity and specificity; required for full peptidase activity [5] | Studying allosteric activation of lasso leader peptidases |
| Sequence Similarity Network (SSN) | Bioinformatic tool to visualize evolutionary relationships and identify new enzyme subfamilies [6] | Discovering novel NHLP-selective LanM enzymes (e.g., DprM) [6] |
Figure 2: Experimental Workflows for Functional Analysis - This diagram outlines the key steps for conducting in vivo modification and in vitro peptidase activation assays.
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a rapidly expanding superfamily of natural products unified by a conserved biosynthetic logic. The peptide backbone is genetically encoded, and the translated precursor peptide undergoes a series of post-translational modifications (PTMs) catalyzed by maturase enzymes to produce the final bioactive compound [8]. This pathway offers significant advantages for bioengineering due to its modular nature and the frequent substrate promiscuity of its biosynthetic enzymes [9]. RiPPs have garnered substantial interest in drug discovery and development due to their biosynthetic plasticity and ability to generate diverse bioactive structural scaffolds, with approximately 6% of all US FDA-approved drugs being peptides and over 15 peptide-based drugs approved in the last five years alone [9].
The unified biosynthetic logic of RiPPsâmodifying ribosomally produced peptides using substrate-tolerant enzymesâpositions this class as highly engineerable for creating new molecules and enzyme-based tools [9]. Current successes in RiPP pathway construction, screening, and enzyme development have established a strong foundation for engineering complex peptide therapeutics, addressing limitations in bioavailability and proteolytic stability that have historically restricted their clinical potential [9] [10]. This application note details the core principles and practical methodologies for leveraging RiPP biosynthetic logic in peptide engineering research.
The biosynthesis of all RiPPs follows a conserved, sequential pathway comprising three principal stages: ribosomal translation of the precursor peptide, post-translational modification of the core peptide, and proteolytic maturation to release the bioactive final product.
Biosynthesis initiates with the ribosomal synthesis of a precursor peptide, which typically contains an N-terminal leader peptide and a C-terminal core peptide [9] [11]. The leader peptide serves as a recognition and binding domain for the maturase enzymes, while the core peptide ultimately becomes the final mature product after processing [9]. This architecture is crucial for guiding modifications, though alternative architectures also exist [8]. The genes encoding these precursors are often found in compact biosynthetic gene clusters (BGCs) alongside the genes for their modifying enzymes [8].
The core peptide undergoes extensive enzymatic tailoring by maturase enzymes recognized through their interaction with the leader peptide [9]. RiPP maturase enzymes are remarkably diverse and often promiscuous, offering significant biotechnological potential [8]. These modifications can include cyclization, crosslinking, methylation, glycosylation, and the installation of structurally complex heterocycles, dramatically increasing structural diversity and often enhancing stability and bioactivity [9] [11]. A key recognition mechanism involves the RiPP precursor peptide recognition element (RRE), a domain present in many tailoring enzymes that specifically binds the leader peptide [9].
The final stage involves proteolytic cleavage to remove the leader sequence, which is sometimes accompanied by additional steps such as N-to-C cyclization, to yield the mature bioactive product [11]. This step is essential for activating the RiPP, as the leader peptide often sterically shields the core peptide's activity during the modification process [9].
Table 1: Core Stages of RiPP Biosynthetic Logic
| Biosynthetic Stage | Key Components | Function in Biosynthesis |
|---|---|---|
| Translation | Precursor Peptide (Leader + Core) | Provides genetically encoded scaffold for modifications [9] [11] |
| Post-Translational Modification | Maturase Enzymes (e.g., RRE-containing) | Installs chemical diversity & bioactivity [9] |
| Proteolytic Maturation | Peptidases | Releases mature, active peptide product [11] |
The following diagram illustrates the logical sequence and key components of this core biosynthetic pathway:
Diagram 1: The core RiPP biosynthetic logic, showing the sequential progression from gene cluster to mature bioactive peptide via translation, modification, and proteolytic maturation.
The modularity of RiPP biosynthesis enables multiple strategic approaches for engineering novel bioactive peptides. The most prominent strategies involve manipulating the leader peptide, diversifying the core peptide, and employing enzyme engineering.
The leader peptide is not part of the final product but is essential for guiding biosynthesis. Engineering this domain allows for the redirecting of modifications to non-cognate core peptides.
Rationale: Many RiPP tailoring enzymes possess an RRE domain that recognizes and binds specific motifs within the leader peptide [9]. By swapping leader domains or modifying their sequences, the specificity of the biosynthetic machinery can be altered. This enables the modification of non-native core peptides or the creation of chimeric systems where enzymes from one RiPP pathway are used to modify the core peptide from another [9].
Protocol 3.1.1: Leader Peptide Swapping for Heterologous Core Modification
The core peptide is the most targeted region for engineering, as its sequence determines the final product. Both natural and non-proteinogenic amino acids can be incorporated to create novel analogs.
Rationale: The core peptide is the most targeted region for engineering, as its sequence determines the final product [9]. Maturase enzymes often exhibit substrate tolerance, allowing for the site-specific modification of core peptides that have been mutated or expanded [9]. This strategy is used to generate "libraries" of modified peptides to screen for improved properties like activity, stability, or selectivity.
Protocol 3.2.1: Saturation Mutagenesis of Core Residues
Engineering the maturase enzymes themselves or utilizing high-throughput cell-free systems can overcome bottlenecks in RiPP production and characterization.
Rationale: Some RiPP biosynthetic enzymes have been engineered to function without their leader peptides, simplifying production [9]. Furthermore, Cell-Free Protein Synthesis (CFE) systems offer a rapid, high-throughput platform for expressing and testing RiPP pathways, bypassing the constraints of cellular physiology [12].
Protocol 3.3.1: High-Throughput Screening of RiPP Variants using CFE
Successful engineering of RiPP pathways relies on a suite of specialized reagents and tools. The following table details key resources for designing and executing RiPP engineering campaigns.
Table 2: Essential Research Reagents for RiPP Engineering
| Reagent / Tool | Category | Function & Application |
|---|---|---|
| LassoESM [13] | Computational Model | A specialized language model for predicting lasso peptide properties, substrate compatibility, and bioactivity, enhancing rational design. |
| Cell-Free Expression System [12] | Expression Platform | Enables rapid, high-throughput expression and testing of RiPP precursors and enzymes without using living cells. |
| RODEO [13] | Bioinformatics Tool | A genome mining algorithm that identifies precursor peptides associated with lasso peptide and other RiPP biosynthetic enzymes. |
| AlphaLISA [12] | Detection Assay | A bead-based proximity assay for high-throughput, no-wash detection of post-translational modifications in peptides. |
| Chimeric Leader Peptides [9] | Engineering Tool | Synthetic leaders created by fusing parts of different leaders to redirect enzyme specificity and create hybrid RiPP products. |
| Promiscuous Lasso Cyclases (e.g., FusC, McjC) [13] | Enzymatic Tool | Enzymes with broad substrate tolerance used to cyclize a wide range of non-native core peptide sequences into lasso structures. |
| ThiF-like Adenylyltransferases (TLATs) [11] | Enzymatic Tool | A superfamily of adenylyltransferases that install diverse modifications in RiPP core peptides, such as in Microcin C biosynthesis. |
| N-Dodecanoyl-d23-glycine | N-Dodecanoyl-d23-glycine Deuterated Reagent | N-Dodecanoyl-d23-glycine, 98 atom % D min. For Research Use Only. Not for human or veterinary diagnostic or therapeutic use. |
| D-Glycero-D-guloheptonate-d7 | D-Glycero-D-guloheptonate-d7, MF:C7H13NaO8, MW:255.21 g/mol | Chemical Reagent |
The discovery and engineering of lipolanthinesâRiPP-derived lipopeptidesâexemplify the power of combining multiple RiPP engineering strategies. The following workflow details the process from genome mining to analog generation.
Diagram 2: A generalized workflow for the discovery and engineering of novel RiPP-derived lipopeptides, such as lipolanthines.
Background: Lipolanthines are a class of RiPPs bearing fatty acyl groups, representing hybrid natural products with significant antimicrobial activity [8]. Examples include microvionin and solabiomycin A, which exhibit potent activity against methicillin-resistant Staphylococcus aureus (MRSA) and Mycobacterium tuberculosis, respectively [8].
Engineering Data: The following table quantifies the antimicrobial activity of selected natural and engineered lipolanthines, demonstrating the potential of this RiPP class.
Table 3: Bioactivity of Selected RiPP-Derived Lipopeptides
| Compound Name | Producing Strain / Engineering Context | Bioactivity Profile (Minimum Inhibitory Concentration) |
|---|---|---|
| Microvionin [8] | Microbacterium arborescens (Natural Product) | MRSA: 0.46 µg/mL; MSSA: 0.1 µg/mL |
| Albopeptin B [8] | Streptomyces albofaciens (Natural Product) | Antifungal: 12.5 µg/mL; Gram-positive bacteria: 12.5 µg/mL |
| Solabiomycin A [8] | Streptomyces lydicus (Natural Product) | M. tuberculosis H37Rv: 3.125 µg/mL |
| Darobactin Analogs [9] | Biosynthetic Pathway Engineering | Improved broad-spectrum activity against Gram-negative pathogens |
Outlook: The integration of computational tools like LassoESM for predicting substrate compatibility, combined with high-throughput cell-free screening platforms, is poised to significantly accelerate the design-build-test cycle for RiPP engineering [12] [13]. This will enable researchers to more efficiently navigate the complex sequence-fitness landscape and generate novel RiPP-based therapeutics with tailored properties.
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a major superfamily of natural products with immense structural diversity and pharmaceutical potential [4]. These gene-encoded peptides undergo extensive enzymatic tailoring to achieve their final bioactive conformations, making them attractive targets for bioengineering [14]. The biosynthesis follows a common logic: a precursor peptide containing an N-terminal leader peptide and a C-terminal core peptide is recognized by modification enzymes that install various post-translational modifications (PTMs) on the core region, followed by leader peptide cleavage to yield the mature natural product [15]. This biosynthetic logic provides remarkable predictability from genetic information and enables engineering through manipulation of the precursor peptide sequence and modification enzymes.
Table 1: Major RiPP Classes and Their Characteristics
| RiPP Class | Characteristic Modifications | Key Enzymes | Bioactivities |
|---|---|---|---|
| Lanthipeptides | Lanthionine (Lan), methyllanthionine (MeLan) rings formed via Ser/Thr dehydration and Michael addition | LanB/LanC or LanM synthetases, LanT proteases | Antimicrobial (nisin), antiviral, anticancer [16] [14] |
| Cyanobactins | Heterocyclization (thiazoline/oxazoline), prenylation, oxidation | Heterocyclase (PatD), proteases (PatA/PatG), oxidase, prenyltransferase | Cytotoxic (patellamide A), protease inhibitory [15] |
| Lasso Peptides | N-terminal macrolactam ring formation followed by C-terminal tail threading | B enzyme (protease), C enzyme (cyclase) | Antimicrobial (microcin J25), receptor antagonism [17] |
| Linear Azol(in)e-containing Peptides (LAPs) | Azole/azoline ring formation from Cys, Ser, Thr residues | YcaO heterocyclase, dehydrogenase | Cytotoxic, antibacterial (streptolysin S) [18] |
Lanthipeptides are characterized by thioether cross-links called lanthionine (Lan) and methyllanthionine (MeLan) that form rigid cyclic structures [16]. These modifications proceed via a two-step process: first, Ser/Thr residues in the core peptide are dehydrated to dehydroalanine (Dha) and dehydrobutyrine (Dhb), respectively; second, cysteine thiol groups undergo Michael-type addition to the dehydroamino acids to form the characteristic (methyl)lanthionine bridges [14]. The biosynthetic machinery varies between different classes of lanthipeptides, with Class I systems employing separate dehydratases (LanB) and cyclases (LanC), while Class II systems utilize a single bifunctional enzyme (LanM) that catalyzes both dehydration and cyclization [16].
Figure 1: Lanthipeptide Biosynthetic Pathway
The UniBioCat system provides a cell-free platform for rapid biosynthesis and engineering of lanthipeptides [16].
Materials:
Procedure:
Cyanobactins represent a fascinating RiPP class characterized by heterocyclization and frequent multicore precursor peptides [15]. These natural products are produced by cyanobacteria and exhibit diverse bioactivities including cytotoxicity, protease inhibition, and metal chelation [15]. The biosynthetic pathway involves a series of enzymatic modifications that can generate remarkable chemical diversity from a single precursor peptide.
Figure 2: Cyanobactin Biosynthetic Pathway
Materials:
Procedure:
Table 2: Cyanobactin Modification Enzymes and Functions
| Enzyme | Function | Recognition Site | Key Cofactors |
|---|---|---|---|
| Heterocyclase (PatD) | Cyclizes Cys, Ser, Thr to thiazolines/oxazolines | RSI (LAELSEE) | ATP |
| N-terminal Protease (PatA) | Cleaves leader peptide | RSII (Gx(E/D)xS) | None |
| C-terminal Protease/cyclase (PatG) | Cleaves follower and catalyzes macrocyclization | RSIII ((A/S)YD) | None |
| Oxidase (ThcOx) | Oxidizes azolines to azoles | None specific | FMN |
| Prenyltransferase (LynF/TruF) | Adds prenyl groups to Tyr or Ser/Thr | None specific | Dimethylallyl PP |
Lasso peptides feature a unique threaded rotaxane structure where an N-terminal macrolactam ring is formed between the N-terminal amine and a side chain carboxylate of Asp or Glu, through which the C-terminal tail is threaded [17]. This topology confers exceptional stability against thermal and proteolytic degradation [17]. The biosynthesis requires two key enzymatic activities: proteolytic cleavage by B enzymes to remove the leader peptide, and macrolactam ring formation by C enzymes [19].
Materials:
Procedure: Leader peptide binding assay:
In vitro lasso peptide formation:
Linear azol(in)e-containing peptides (LAPs) incorporate thiazole/oxazole rings formed through heterocyclization of Cys, Ser, and Thr residues [18]. A recently discovered class termed "dehydrazoles" exemplifies hypermodified RiPPs containing both azole rings and dehydroamino acids [18]. The carnazolamide pathway demonstrates exceptional efficiency, with just five enzymes installing 18 distinct post-translational modifications on a 30-residue peptide [18].
Figure 3: LAP/Dehydrozole Biosynthetic Pathway
Materials:
Procedure:
Table 3: Key Research Reagents for RiPP Biosynthesis Studies
| Reagent/Category | Specific Examples | Function/Application |
|---|---|---|
| Expression Systems | E. coli BL21(DE3) cell extracts, PURE system | Cell-free protein synthesis and modification [16] |
| Chaperone Systems | DnaK-DnaJ-GrpE, GroEL-GroES | Enhance soluble expression of modification enzymes [16] |
| Affinity Tags | Polyhistidine (His-tag), Maltose-binding protein (MBP) | Protein purification and binding studies [17] [19] |
| Proteases | TEV protease, Factor Xa | Cleavage of affinity tags and leader peptides [17] |
| Mass Spectrometry | MALDI-TOF MS, LC-MS/MS | Modification verification and structural characterization [16] [18] |
| Binding Assay Systems | Biolayer interferometry (BLI), Isothermal titration calorimetry (ITC) | Protein-peptide interaction studies [17] [19] |
| Specialized Enzymes | YcaO heterocyclases, LanM synthetases, RRE domains | Installation of specific post-translational modifications [20] [18] |
| Activity Assays | Agar diffusion assays, microbroth dilution | Antimicrobial activity evaluation [16] |
| (3S)-3-Hydroxy Quinidine-vinyl-d3 | (3S)-3-Hydroxy Quinidine-vinyl-d3, MF:C20H24N2O3, MW:343.4 g/mol | Chemical Reagent |
| Anti-inflammatory agent 9 | Anti-inflammatory agent 9, MF:C18H15N5O2S, MW:365.4 g/mol | Chemical Reagent |
The modular nature of RiPP biosynthesis enables sophisticated engineering approaches for generating novel analogs with improved or altered bioactivities [14]. Key strategies include:
Precursor peptide engineering:
Enzyme engineering:
Combinatorial biosynthesis:
These engineering approaches facilitate the generation of RiPP analogs that mimic complex non-ribosomal peptides (NRPs) such as daptomycin and teixobactin, combining the biosynthetic flexibility of RiPPs with the potent bioactivities of NRPs [14].
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a rapidly expanding superfamily of natural products with remarkable structural diversity and significant pharmaceutical potential [22] [1]. These compounds are unified by a common biosynthetic logic: a genetically encoded precursor peptide is first synthesized by the ribosome and then extensively modified by dedicated maturase enzymes to produce the final bioactive compound [23]. The RiPP biosynthetic pathway, often referred to as Post-Ribosomal Peptide Synthesis (PRPS), begins with a precursor peptide typically comprising an N-terminal leader peptide and a C-terminal core peptide, though alternative architectures like C-terminal follower peptides also exist [22] [24]. The core peptide undergoes a series of enzymatic transformations that dramatically alter its chemical structure, imparting conformational restraint, metabolic stability, and biological activity [22] [25]. The resulting natural products exhibit an impressive range of bioactivities, including antibiotic, antifungal, antiviral, antitumor, and antiparasitic properties, making them attractive targets for drug development [26] [8].
The evolutionary advantage of RiPP biosynthesis lies in its genetic economy and high evolvability. The biosynthetic gene clusters (BGCs) are relatively compact, and the maturase enzymes often display significant substrate promiscuity, recognizing the conserved leader peptide while tolerating extensive variation in the core peptide sequence [22]. This modularity enables rapid generation of chemical diversity from simple genetic changes, a feature that peptide engineers are increasingly exploiting to create novel compounds with tailored properties [27]. As the falling cost of genome sequencing continues to reveal countless uncharacterized RiPP BGCs, understanding the key post-translational modificationsâparticularly cyclization, methylation, dehydration, and heterocycle formationâbecomes crucial for unlocking their biotechnological and therapeutic potential [23] [25] [1].
The biosynthesis of all RiPPs follows a conserved pattern initiated by the ribosomal production of a precursor peptide. This precursor is organized into distinct regions: most commonly an N-terminal leader peptide and a C-terminal core peptide [22]. The leader peptide serves as a recognition sequence for the maturase enzymes and typically remains unchanged during modification of the core peptide [22]. In some rare cases, such as the bottromycins, a C-terminal "follower" peptide performs this recognition function instead [22] [24]. For eukaryotic RiPPs like cyclotides and conopeptides, an additional signal sequence may direct cellular localization [22]. The extensive post-translational modifications installed by the maturase enzymes include cyclizations, crosslinks, heterocycle formations, and various side-chain modifications that collectively transform the linear peptide into a structurally complex natural product [22] [28]. The final maturation step involves proteolytic removal of the leader peptide (and/or follower peptide) to release the bioactive mature RiPP [22].
A key feature of RiPP biosynthesis is the division of labor between recognition and modification. The leader peptide ensures that the correct core peptide is modified by the appropriate enzymes, while the core peptide itself can tolerate significant sequence variation, enabling rapid evolution of chemical diversity [22]. This biosynthetic logic facilitates genome mining approaches, as the presence of a precursor peptide gene near maturase enzymes often signals a RiPP pathway [1]. The resulting natural products typically fall within the molecular weight range of 1,000-5,000 Da, occupying a valuable chemical space between small molecules and large biologics, with enhanced binding specificity and ability to inhibit protein-protein interactions [26].
The field of RiPP research has established uniform nomenclature to address previously confusing and contradictory terminology [22]. The biosynthetic pathway is designated Post-Ribosomal Peptide Synthesis (PRPS), drawing a parallel to Non-Ribosomal Peptide Synthesis (NRPS) [22]. The final products are termed RiPPs, specifically excluding larger post-translationally modified proteins through an approximate 10 kDa molecular weight limit [22].
Table 1: Standardized Nomenclature for RiPP Biosynthesis
| Term | Definition | Previous Designations |
|---|---|---|
| Precursor peptide | Initial ribosomal translation product containing leader and core regions | Prepeptide, prepropeptide, structural peptide |
| Leader peptide | N-terminal recognition sequence for maturase enzymes | Propeptide, pro-region, intervening region |
| Core peptide | Region transformed into the final natural product | Propeptide, structural peptide, toxin region |
| UCP | Unmodified core peptide in the precursor peptide | N/A |
| MCP | Modified core peptide after post-translational modifications | N/A |
| Follower peptide | C-terminal recognition sequence (rare) | N/A |
Residue numbering follows a specific convention where the last residue of the leader peptide (not incorporated in the final RiPP) is numbered -1, with counting proceeding -2, -3, etc., toward the N-terminus [22]. Similarly, C-terminal recognition sequences are numbered with a plus sign from the cleavage site (+1, +2, etc.) [22]. This standardized terminology facilitates clear communication and computational annotation of RiPP biosynthetic gene clusters across the diverse subclasses [22].
Macrocyclization represents one of the most common and impactful modifications in RiPP biosynthesis, serving to constrain peptide conformation, enhance metabolic stability, improve target affinity, and increase specificity [28]. This transformation is so valuable that medicinal chemists frequently employ similar strategies in peptide drug development [28]. The mechanisms of macrocyclization in RiPPs are remarkably diverse, involving different chemical reactions and resulting in various cyclic architectures that define several RiPP subclasses [28] [24].
The biological advantages of cyclization are profound. Constraining the peptide backbone into a specific conformation reduces the entropic penalty upon binding to biological targets, significantly enhancing binding affinity [28]. Additionally, the cyclic structure confers resistance to exoprotease degradation, extending the peptide's half-life in biological environments [24]. These properties make macrocyclic RiPPs particularly attractive as therapeutic leads, as they combine the specificity of peptides with improved pharmacokinetic properties [28]. The ribosomal origin of these compounds further facilitates engineering approaches, as sequence modifications can be introduced at the genetic level rather than requiring complex synthetic chemistry [24].
Table 2: Major Cyclization Mechanisms in RiPP Biosynthesis
| Cyclization Type | Chemical Basis | Representative RiPP Classes | Key Features |
|---|---|---|---|
| Lanthionine/MeLan formation | Michael-type addition of Cys thiols to dehydroamino acids | Lanthipeptides [28] | Thioether crosslinks; multiple classes based on synthetase enzymes |
| Head-to-tail cyclization | Amide bond between N- and C-termini | Cyanobactins, cyclotides, orbitides [22] [24] | N-to-C macrocyclization; diverse ring sizes |
| Azole/azoline formation | ATP-dependent heterocyclization | TOMMs, LAPs, thiopeptides, cyanobactins [24] | Oxazoles/thiazoles from Ser/Thr/Cys; requires dehydrogenase for aromatization |
| Radical-mediated cyclization | Carbon-carbon bond formation via radical SAM enzymes | Sactipeptides, ranthipeptides, streptide [28] | Unactivated carbon centers; unusual crosslinks |
| [4+2] Cycloaddition | Diels-Alder reaction | Thiopeptides [28] | Complex pyridine formation; pericyclic reaction |
| Lasso peptide formation | Isopeptide bond between N-terminus and Asp/Glu side chain | Lasso peptides [1] | Mechanically interlocked topology; protease resistance |
Figure 1: Cyclization Mechanisms and Applications in RiPP Biosynthesis
Purpose: To reconstitute the macrocyclization activity of cyanobactin enzymes in vitro for the production of cyclic peptide libraries [27].
Materials:
Method:
Enzyme preparation: Express and purify TruD1 and TruG1 as recombinant His-tagged proteins from E. coli using standard nickel affinity chromatography.
Heterocyclization reaction: Combine 50 μL precursor peptide (1 mM), 10 μL TruD1 (0.1 mM), 10 μL ATP (100 mM), and 25 μL ATP regeneration system in reaction buffer to a total volume of 500 μL. Incubate at 30°C for 2 hours with gentle agitation.
Macrocyclization and cleavage: Add 10 μL TruG1 (0.05 mM) to the heterocyclized peptide mixture. Incubate at 30°C for an additional 3 hours.
Product analysis: Quench the reaction with 50 μL 10% trifluoroacetic acid (TFA). Separate by reverse-phase HPLC using a water-acetonitrile gradient (5-95% acetonitrile + 0.1% TFA over 30 minutes). Collect eluted peaks and analyze by MALDI-TOF MS.
Cyclic peptide verification: Compare HPLC retention times and mass spectra to linear controls. Confirm cyclization through MS/MS fragmentation and resistance to exoprotease treatment (aminopeptidase M and carboxypeptidase Y).
Notes: The order of enzyme addition is criticalâheterocyclization must precede macrocyclization for efficient processing. This protocol can be adapted for one-pot reactions with multiple modification enzymes by optimizing enzyme ratios and reaction timing [27].
Methylation represents a prevalent post-translational modification in RiPP biosynthesis, dramatically altering peptide properties through the addition of methyl groups to various acceptor sites [26]. These transformations are catalyzed by S-adenosylmethionine (SAM)-dependent methyltransferases (MTs) that exhibit remarkable chemo-, regio-, and stereoselectivity [26]. The introduction of methyl groups can significantly enhance a compound's biological activityâa phenomenon known as the "magic methyl effect"âwhere a single methyl group improves drug efficacy by orders of magnitude [26]. Methylation modulates peptide properties by increasing hydrophobicity, altering hydrogen bonding capacity, providing steric bulk, and stabilizing specific conformations, all of which can profoundly influence bioactivity and metabolic stability [26].
RiPP methyltransferases target diverse positions within peptide substrates, including nitrogen atoms (N-terminus, lysine side chains, histidine imidazole, backbone amides), oxygen atoms (glutamate/aspartate side chains, serine/threonine side chains, C-terminus), carbon atoms (unactivated sp³ carbons), and sulfur atoms (cysteine side chains, methionine) [26]. This chemical versatility enables extensive diversification of RiPP scaffolds from common precursor peptides. The MTs achieve their impressive selectivity through precise recognition of both the leader peptide sequence and specific features of the core peptide, allowing them to modify particular residues while ignoring chemically similar alternatives [26]. This specificity, combined with their catalytic efficiency, makes RiPP methyltransferases valuable tools for biocatalytic applications.
Purpose: To employ RiPP methyltransferases for selective in vitro methylation of peptide substrates [26].
Materials:
Method:
SAM regeneration system preparation: Combine methionine adenosyltransferase (0.01 mM), L-methionine (10 mM), and ATP (5 mM) in methyltransferase assay buffer. Pre-incubate at 30°C for 15 minutes to generate SAM in situ.
Methylation reaction: Combine 50 μL peptide substrate (0.5 mM), 25 μL SAM regeneration system, 10 μL methyltransferase (0.05 mM), and assay buffer to a total volume of 250 μL. Incubate at 30°C for 4-16 hours with gentle mixing.
Reaction monitoring: Remove 10 μL aliquots at 0, 2, 4, and 8 hours. Quench with 10 μL 10% TFA and analyze by LC-MS. Monitor for mass increase of +14 Da per methylation event.
Product purification: Pool completed reactions and desalt using spin columns according to manufacturer's instructions. Further purify by semi-preparative HPLC if necessary.
Methylation site mapping: For unknown substrates, perform MS/MS fragmentation to identify specific methylation sites. Compare fragmentation patterns to unmethylated controls.
Troubleshooting:
Applications: This protocol enables selective introduction of methyl groups at specific positions in complex peptides, offering advantages over chemical methods that often require protecting groups and produce toxic waste [26].
Dehydration of serine and threonine residues to form dehydroalanine (Dha) and dehydrobutyrine (Dhb) represents a fundamental transformation in multiple RiPP classes [25]. This process typically begins with phosphorylation or glutamylation of the side chain hydroxyl group, followed by elimination to generate the dehydrated residue [22] [25]. These dehydroamino acids serve as crucial intermediates for subsequent modifications, particularly in lanthipeptides where they undergo Michael-type additions with cysteine thiols to form lanthionine and methyllanthionine crosslinks [28] [24]. Recent research has revealed unusual dehydration enzymes like CcaM that function independently of leader peptides and specifically target serine residues preceding azole moieties, demonstrating the sophisticated regulatory mechanisms controlling modification timing in complex RiPP pathways [25].
Heterocycle formation through cyclodehydration of cysteine, serine, and threonine residues produces thiazolines and (methyl)oxazolines, which can be further oxidized to the aromatic thiazoles and (methyl)oxazoles [25] [24]. This transformation is catalyzed by YcaO-domain proteins that utilize ATP to activate the backbone amide carbonyl for nucleophilic attack by the side chain heteroatom [24]. The resulting azol(in)es constrict peptide conformation and enhance stability against proteolytic degradation [24]. In remarkable examples of biosynthetic efficiency, some RiPP pathways install both dehydroamino acids and azol(in)es on the same precursor peptide, as demonstrated in the dehydrazoles like carnazolamide, where 18 post-translational modifications are installed by just five enzymes [25].
Purpose: To reconstitute the coordinated activities of YcaO heterocyclase and LanMbC dehydratase in the biosynthesis of dehydrazole-class RiPPs [25].
Materials:
Method:
Heterocyclization reaction: Combine 50 μL precursor peptide (0.2 mM), 10 μL CcaB (0.05 mM), 10 μL ATP (10 mM), and reaction buffer to 200 μL total volume. Incubate at 30°C for 2 hours.
Oxidation step: Add 5 μL CcaC (0.1 mM) and 5 μL NAD⺠(5 mM) to the heterocyclized peptide. Incubate at 30°C for 1 hour to convert azolines to azoles.
Dehydration reaction: Add 10 μL CcaM (0.05 mM) and additional 10 μL ATP (10 mM) to the oxidized peptide. Incubate at 30°C for 3 hours.
Process monitoring: Analyze 10 μL aliquots after each step by LC-MS. Heterocyclization is indicated by mass decrease of -18 Da per cyclization, oxidation shows no mass change, and dehydration shows additional -18 Da per dehydration event.
Order dependence testing: To confirm the core-dependence of CcaM, set up parallel reactions where dehydration is attempted before heterocyclization. Compare efficiency by product yield.
Product characterization: Purify the fully modified peptide by HPLC. Verify modification sites using MS/MS fragmentation and compare to unmodified controls.
Key Insights: This protocol demonstrates the importance of modification order in complex RiPP pathways. The core-dependence of CcaM ensures that heterocyclization precedes dehydration, preventing competition for unmodified serine residues [25]. This biosynthetic logic enables the efficient installation of multiple modification types without cross-interference.
Table 3: Essential Research Reagents for RiPP Pathway Reconstitution
| Reagent Category | Specific Examples | Function in RiPP Research | Key Considerations |
|---|---|---|---|
| Precursor Peptides | Synthetic CcaA, McbA, NisA | Substrate for PTM enzymes; engineering template | Include leader sequence for enzyme recognition; core sequence can be varied |
| Core Modification Enzymes | YcaO proteins, LanM synthetases, Radical SAM enzymes | Install primary post-translational modifications | Often require cofactors (ATP, SAM, NADâº); may need anaerobic handling |
| Processing Enzymes | Leader peptidases, Macrocyclases | Release mature RiPP from precursor | Specific to leader peptide sequence; timing affects final product |
| Cofactor Systems | ATP regeneration, SAM regeneration | Sustain enzymatic activity in vitro | Crucial for multi-enzyme reconstitution; reduces product inhibition |
| Analytical Tools | LC-MS with C18 columns, MALDI-TOF MS | Monitor reaction progress; verify structures | High resolution needed for mass accuracy; MS/MS for modification mapping |
The modular nature of RiPP biosynthetic enzymes enables sophisticated engineering approaches for generating novel peptide architectures [27]. Successful engineering requires careful consideration of enzyme compatibility, reaction order, and substrate recognition requirements. The workflow typically begins with bioinformatic identification of candidate BGCs, followed by heterologous expression and in vitro characterization of individual enzymes to define their substrate scope and specificity [25] [8]. Once the fundamental properties are established, pathway reassembly can be pursued through either genetic engineering of producer strains or in vitro reconstitution with purified components [27].
Figure 2: Integrated Workflow for RiPP Pathway Discovery and Engineering
Table 4: Kinetic Parameters of Key RiPP Modification Enzymes
| Enzyme Class | Representative Enzyme | Reaction Catalyzed | Key Cofactors | Reported Turnover | Engineering Applications |
|---|---|---|---|---|---|
| YcaO heterocyclase | CcaB [25] | Azoline formation from Cys/Ser/Thr | ATP | Multiple turnovers per peptide | Broad substrate tolerance; core diversification |
| LanM dehydratase | CcaM [25] | Ser/Thr dehydration to Dha/Dhb | ATP (Glu-tRNA for some) | 8+ dehydrations per peptide [25] | Leader-independent activity; unusual specificity |
| Radical SAM MT | CcaH [25] | C-methylation of unactivated carbons | SAM, redox center | Single methylation per SAM | Unique chemoselectivity; difficult chemical mimicry |
| Lanthipeptide cyclase | ProcM [28] | Thioether ring formation | Zn²⺠(some classes) | Multiple cyclizations per peptide | Remarkable promiscuity; library generation |
| Macrocyclase | TruG1 [27] | Head-to-tail cyclization + cleavage | None | Single cyclization per peptide | Biocompatible macrocyclization tool |
Purpose: To simultaneously employ multiple RiPP modification enzymes in a single reaction vessel for efficient synthesis of complex peptide natural products [27].
Materials:
Method:
Enzyme titration: Determine the minimal effective concentration for each enzyme through individual reactions. Aim for enzyme:substrate ratios between 1:10 and 1:100.
One-pot reaction assembly: Combine precursor peptide (0.1 mM final concentration) with cofactor master mix in reaction buffer. Add all modification enzymes simultaneously at their predetermined optimal concentrations. Adjust final volume with reaction buffer.
Reaction incubation: Incubate at 30°C with gentle agitation for 6-16 hours. Monitor progress by LC-MS at 2-hour intervals.
Product isolation: Once conversion plateaus, quench reaction with 0.1% TFA. Purify product by preparative HPLC. Lyophilize pure fractions for characterization.
Yield optimization: If yield is suboptimal, vary enzyme addition order or implement staggered addition based on known biosynthetic logic. For example, in dehydrazole synthesis, add heterocyclation enzymes before dehydration enzymes [25].
Applications: This approach has been successfully demonstrated for cyanobactin synthesis, where up to five different post-translational enzymes were combined in a single tube to generate highly unnatural peptide derivatives, including an N-C peptide macrocycle of 22 amino acids in length [27]. The method capitalizes on the modularity and substrate tolerance of RiPP enzymes to create diverse peptide libraries.
The systematic investigation of key post-translational modifications in RiPP biosynthesisâcyclization, methylation, dehydration, and heterocycle formationâhas revealed fundamental principles of enzymatic catalysis and provided powerful tools for peptide engineering. The modular nature of RiPP pathways, combined with the remarkable substrate tolerance of many maturase enzymes, enables sophisticated engineering approaches that were previously unimaginable [27]. As genome mining continues to reveal novel RiPP classes with unusual modifications [23] [25] [8], and as our understanding of enzyme mechanisms deepens [28] [26] [24], the toolkit for peptide engineering will expand accordingly.
The future of RiPP engineering lies in the intelligent combination of modifications to create peptides with tailored properties. The emerging paradigm of "biosynthetic code" decipheringâunderstanding how enzyme specificity is encoded in leader peptides and substrate contextsâwill enable more predictive engineering [22] [25]. Additionally, the discovery of hybrid systems like RiPP-derived lipopeptides [23] [8] points toward integration with other biosynthetic paradigms (PKS, NRPS) for further structural diversification. As biochemical reconstitution protocols become more sophisticated and high-throughput [25] [27], the pace of discovery and engineering will accelerate, potentially yielding new therapeutic classes to address pressing medical challenges. The protocols and analyses presented here provide a foundation for these future advances in RiPP-based peptide engineering.
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a rapidly growing class of natural products characterized by their genetically encoded precursor peptides and extensive enzymatic tailoring. These compounds are produced across all domains of lifeâprokaryotes (bacteria and archaea) and eukaryotesâexhibiting remarkable structural diversity and potent biological activities that have attracted significant interest for therapeutic development. RiPP biosynthesis follows a conserved logic: a ribosomally produced precursor peptide containing an N-terminal leader peptide and a C-terminal core peptide is modified by various enzymes, after which the leader peptide is proteolytically removed to yield the mature natural product [22]. The core peptide undergoes diverse post-translational modifications (PTMs), including cyclization, methylation, and the introduction of thioether crosslinks, which greatly expand their structural complexity and functional versatility [22] [29].
The ecological functions of RiPPs are increasingly recognized as crucial in mediating microbial interactions, including prokaryote-phage relationships and host-microbiome homeostasis [30] [31]. For peptide engineering research, the genetic encodability of RiPP precursors combined with the relaxed substrate specificity of many RiPP biosynthetic enzymes makes this class of natural products particularly amenable to bioengineering approaches [9] [29]. This application note provides a comprehensive overview of RiPP distribution across life domains, summarizes quantitative biosynthetic data, details experimental protocols for RiPP discovery, and visualizes key biosynthetic pathways to support research in this rapidly advancing field.
Bacterial RiPPs represent the most extensively characterized group, with biosynthetic gene clusters (BGCs) widespread across numerous phyla. Large-scale genomic analyses reveal that bacterial RiPPs are particularly abundant in human-associated microbiomes. A recent study of 306,481 microbial genomes from human body sites identified RiPPs in up to 95.3% of genomes using precursor-centric detection methods, with the highest enrichment observed in the gut and oral microbiomes [31]. The phyla Bacteroidota and Firmicutes, dominant in the human gut, harbored the most abundant RiPPs, with radical SAM (rSAM)-modified RiPPs being the most prevalent class [31]. Genome mining has revealed extensive diversity, with 12,076 putative RiPP families identified in human microbiomes alone, demonstrating the vast untapped biosynthetic potential of bacterial RiPPs [31].
Archaeal RiPPs are less extensively characterized but show intriguing distributions and unique features. While archaeal ribosomes share protein components with eukaryotes, including archaea/eukaryote-specific ribosomal proteins (r-proteins) [32], the distribution of RiPP pathways in archaea appears more limited compared to bacteria. However, specific archaeal lineages produce specialized RiPP classes, such as thioamidated peptides catalyzed by YcaO-TfuA protein pairs [33]. Global genome mining efforts have identified putative thioamidated RiPP BGCs in various archaeal phyla, suggesting these organisms possess unique RiPP biosynthetic capabilities [33]. Archaeal RiPP pathways often represent simplified versions of eukaryotic systems, providing models for understanding functional essentials of RiPP biosynthesis that evolved before the diversification of archaea and eukaryotes [34].
Eukaryotic RiPPs have been identified in fungi, plants, and animals, with distinctive biosynthetic features. Unlike prokaryotic systems, eukaryotic precursor peptides often contain an N-terminal signal sequence that directs the peptide to specific cellular compartments where post-translational modifications occur [22]. Well-characterized eukaryotic RiPP classes include cyclotides from plants, conopeptides from cone snails, and amatoxins from mushrooms, all featuring complex cyclization patterns and disulfide crosslinking [22]. The ribosome biogenesis machinery in eukaryotes shares significant homology with archaeal systems, particularly in the archaea/eukaryote-specific ribosomal proteins, suggesting evolutionary conservation of fundamental ribosomal processes that may extend to RiPP biosynthesis [32] [34].
Table 1: Distribution of RiPP Biosynthetic Potential Across Domains of Life
| Domain | Representative Organisms | Key RiPP Classes | Unique Features | Biosynthetic Potential |
|---|---|---|---|---|
| Bacteria | Human gut microbiota (Bacteroidota, Firmicutes) | Lanthipeptides, Thiopeptides, Sactipeptides | Most diverse RiPP classes; High prevalence in microbiome | 78.3-95.3% of genomes encode RiPPs [31] |
| Archaea | Methanogens, Asgard group | Thioamidated peptides | Simplified assembly pathways; TfuA-YcaO systems | Limited but distinct (e.g., 613 TfuA-related GCFs) [33] |
| Eukarya | Plants, Fungi, Animals | Cyclotides, Conopeptides, Amatoxins | Signal sequences for compartmentalization; Complex cyclization | Widespread but less systematically characterized |
Large-scale genomic analyses have enabled systematic quantification of RiPP biosynthetic potential across diverse taxa. In human microbiomes, precursor-centric genome mining approaches have identified 423,831 RiPP precursors confirmed by at least two independent detection strategies, distributed across 21 phyla, 1,014 genera, and 3,369 species [31]. The biosynthetic potential varies considerably at the genus level, with Elizabethkingia encoding up to 33 RiPP precursors per genome, Chryseobacterium encoding 18, and Tissierella encoding 15 [31]. This extensive diversity underscores the role of RiPPs in mediating ecological interactions within complex microbial communities.
Global mining of specific RiPP classes has revealed substantial untapped chemical space. For thioamidated RiPPs, analysis of 162,672 bacterial and archaeal genomes identified 613 unique TfuA-related gene cluster families and 797 precursor peptide families, extending across phyla where these clusters had not been previously described [33]. Similarly, analysis of ocean microbiome datasets using the deep learning tool TrRiPP uncovered a diverse array of previously uncharacterized putative RiPP families with high novelty and diversity [30]. These findings highlight that current knowledge of RiPP diversity represents only a fraction of their true biosynthetic potential in nature.
Table 2: Quantitative Distribution of RiPPs in Human Microbiomes
| Body Site | Genomes with RiPPs | Most Abundant RiPP Classes | Noteworthy Producers | Potential Health Relevance |
|---|---|---|---|---|
| Gut | High enrichment | rSAM-modified RiPPs, Lanthipeptides | Bacteroidota, Firmicutes | 30 RiPP families inversely related to IBD and CRC [31] |
| Oral | High enrichment | Thiopeptides, Lanthipeptides | Streptococcus spp. | Antimicrobials against oral pathogens |
| Skin | Moderate | Autoinducing peptides (AIPs) | Staphylococcus spp. | Maintain skin barrier homeostasis [31] |
| Vaginal | Moderate | Lanthipeptides | Lactobacillus gasseri | Antibacterial against vaginal pathogens [31] |
Purpose: To identify novel RiPP precursors from highly fragmented metagenomic data using deep learning approaches.
Materials and Reagents:
Procedure:
Applications: This protocol successfully identified novel RiPP families in ocean microbiomes and revealed their potential roles in prokaryote-phage interactions through correlation analysis with antiphage defense-related proteins [30].
Purpose: To rapidly discover and validate therapeutic RiPP candidates through biosynthesis-guided chemical synthesis.
Materials and Reagents:
Procedure:
Applications: This approach identified five AIPs that inhibited biofilm formation of disease-associated pathogens and two AIPs that regulated microbial community and reduced harmful species in IBD mouse models [31].
The unified biosynthetic logic of RiPP pathways enables predictable engineering approaches. All RiPPs originate from a ribosomally synthesized precursor peptide containing a leader region and core region. Modification enzymes recognize the leader peptide and install diverse structural features onto the core peptide, followed by proteolytic removal of the leader and export of the mature product [22]. This conserved framework facilitates bioengineering strategies targeting each biosynthetic step.
Diagram 1: Core RiPP biosynthetic pathway. PTM: Post-translational modification.
RiPP engineering leverages the modularity of biosynthetic components to generate novel analogs with optimized properties. Three primary engineering strategies have emerged:
Leader peptide manipulation: Creating chimeric leader peptides enables redirecting modification enzymes to non-cognate core peptides, generating hybrid natural products [9]. This approach capitalizes on the leader peptide's role in enzyme recognition while swapping core peptides to introduce structural diversity.
Core peptide diversification: Mutating core peptide residues within enzyme tolerance regions generates analogs with modified bioactivities [9]. Successful examples include incorporating non-proteinogenic amino acids and generating lanthipeptide libraries with enhanced antimicrobial properties.
Enzyme engineering: Modifying RiPP tailoring enzymes expands their substrate scope and enables installation of non-natural modifications [9] [29]. Combining enzymatic transformations with synthetic chemistry approaches further diversifies the accessible chemical space.
Table 3: Key Research Reagents for RiPP Discovery and Characterization
| Reagent/Tool | Type | Function | Example Applications |
|---|---|---|---|
| TrRiPP | Software tool | Identifies RiPP precursors using deep learning | Ocean metagenome mining [30] |
| antiSMASH | Software tool | Identifies biosynthetic gene clusters | RiPP BGC detection in human microbiome [31] |
| RiPPER | Software tool | Detects RiPP precursor peptides | Thioamidated RiPP discovery [33] |
| YcaO-TfuA system | Enzymatic pair | Catalyzes thioamide formation | Thioamidated RiPP biosynthesis [33] |
| Radical SAM enzymes | Enzyme family | Installs thioether crosslinks | Sactipeptide biosynthesis [29] |
| Heterologous expression systems | Method | Produces RiPPs in surrogate hosts | Thuricin CD characterization [29] |
| BRD4 ligand-Linker Conjugate 1 | BRD4 Ligand-Linker Conjugate 1 | PROTAC Intermediate | BRD4 ligand-Linker Conjugate 1 is a key intermediate for constructing PROTAC degraders. This product is For Research Use Only (RUO). Not for human or veterinary use. | Bench Chemicals |
| Chlormadinone Acetate-d3 | Chlormadinone Acetate-d3, MF:C23H29ClO4, MW:407.9 g/mol | Chemical Reagent | Bench Chemicals |
RiPPs demonstrate significant potential for therapeutic development, particularly in managing microbial infections and microbiome-associated diseases. Protective roles of RiPPs in human health are increasingly recognized, with specific RiPP families showing inverse relationships to disease states such as inflammatory bowel disease and colorectal cancer [31]. Autoinducing peptides (AIPs) from human microbiomes have demonstrated efficacy in inhibiting biofilm formation of pathogens and regulating microbial communities in disease models [31].
The bioengineering potential of RiPP pathways further enhances their therapeutic appeal. Successful engineering examples include darobactin biosynthetic pathway engineering to generate broad-spectrum antibiotics against Gram-negative pathogens [9] and creation of hybrid lanthipeptides with selective antimicrobial activity against Staphylococcus aureus [9]. The genetic encodability of RiPP precursors combined with increasing understanding of their biosynthetic logic positions RiPPs as ideal targets for rational design of peptide-based therapeutics.
RiPP natural products represent a vast and largely untapped resource for drug discovery and development, with diverse representatives distributed across prokaryotes, eukaryotes, and archaea. The unified biosynthetic logic of RiPP pathways, combined with their genetic encodability and enzymatic plasticity, makes them particularly amenable to engineering approaches. Quantitative genomic analyses reveal extensive RiPP biosynthetic potential in environmental and host-associated microbiomes, highlighting opportunities for future discovery. As research continues to elucidate the ecological functions, biosynthetic mechanisms, and therapeutic potential of RiPPs, these versatile molecules are poised to make significant contributions to peptide engineering and therapeutic development.
Biosynthetic Gene Clusters (BGCs) are genomic loci that encode the production of microbial natural products, which form the basis for nearly 75% of human medicines, including antimicrobials and immunosuppressants [35] [29]. Bioinformatics tools have become indispensable for identifying these clusters, transforming natural product discovery from a purely activity-based screening process to a targeted, sequence-driven endeavor [35]. This application note details the integrated use of three powerful bioinformatics toolsâantiSMASH, PRISM, and RODEOâfor BGC identification, with specific focus on their application to ribosomally synthesized and post-translationally modified peptides (RiPPs). RiPPs represent a promising superfamily of natural products characterized by their genetically encoded precursor peptides that undergo extensive enzymatic tailoring to produce structurally complex bioactive compounds [29] [9]. Their unified biosynthetic logic and the genetic encodability of their substrates make them particularly amenable to bioengineering for pharmaceutical development [9].
antiSMASH (antibiotics & Secondary Metabolite Analysis Shell) is the most widely used platform for detecting and characterizing BGCs in bacterial and fungal genomes [36]. It functions by identifying conserved biosynthetic genes through profile hidden Markov models (pHMMs) and then analyzing domain architecture to predict cluster type and potential chemical product [35]. The recently released antiSMASH 6.0 supports 71 different cluster typesâa significant increase from previous versionsâand offers enhanced detection of tailoring enzymes in RiPP clusters [36].
PRISM (PRediction Informatics for Secondary Metabolites) is a genome mining tool with a strong emphasis on predicting chemical structures of biosynthetic pathways, particularly for nonribosomal peptides, polyketides, and RiPPs [37] [35]. PRISM is closely integrated with the Genomes-to-Natural Products platform (GNP), which enables matching of genomic predictions with LC-MS/MS data for compound identification [35]. This structural prediction capability makes PRISM particularly valuable for hypothesis generation in drug discovery pipelines.
RODEO (Rapid ORF Description and Evaluation Online) specializes in the detection and analysis of RiPP biosynthetic gene clusters [37] [35]. Unlike general-purpose tools, RODEO combines genome mining with motif-based analysis to identify RiPP precursor peptides and predict post-translational modifications, making it uniquely suited for discovering novel ribosomally synthesized peptides [37] [9].
Table 1: Comparative Analysis of BGC Identification Tools
| Tool | Primary Function | Strengths | RiPP-Specific Features | Access |
|---|---|---|---|---|
| antiSMASH | Comprehensive BGC detection & classification | Broadest cluster type coverage (71 types); most widely used; integrates multiple algorithms | RiPP-specific HMM profiles; RODEO integration; tailoring enzyme detection [36] [35] | Web server & standalone [35] |
| PRISM | Chemical structure prediction | Predicts complete chemical structures; links BGCs to MS/MS data | RiPP cluster detection & structure prediction; handles complex modifications [37] | Web server [37] |
| RODEO | RiPP cluster identification | Specialized for RiPP discovery; identifies precursor peptides | Motif-based precursor peptide identification; comprehensive RiPP analysis [37] [9] | Web server & standalone [37] |
Several specialized tools complement these primary platforms for specific research applications. BAGEL is dedicated to mining ribosomally synthesized and post-translationally modified peptides (RIPPs), including lanthipeptides and bacteriocins, making it particularly valuable for antimicrobial discovery [37]. ARTS (Antibiotic Resistant Target Seeker) integrates with antiSMASH to prioritize BGCs based on potential antibiotic activity and identifies resistance genes that may indicate novel mechanisms of action [37] [38]. DeepBGC employs machine learning for BGC classification and can predict bioactivity (antibacterial, antifungal, cytotoxic) directly from BGC sequences, though with currently limited accuracy for some activity classes [38].
Table 2: Specialized Tools for Advanced BGC Analysis
| Tool | Application | Utility in RiPP Research |
|---|---|---|
| BAGEL | RIPP identification & analysis | Identifies and classifies RIPP clusters in genomic data [37] |
| ARTS | BGC prioritization & resistance gene identification | Identifies potential antibiotic targets and resistance mechanisms [37] |
| DeepBGC | Machine learning-based BGC classification & activity prediction | Predicts antibacterial/antifungal activity from BGC features [38] |
| BiG-SCAPE | Gene cluster family analysis & dereplication | Groups BGCs into families; identifies novel clusters [37] |
Principle: antiSMASH identifies BGCs through a multi-step process beginning with gene annotation, followed by core biosynthetic gene detection using pHMMs, and finally cluster boundary prediction and annotation [35].
Procedure:
Troubleshooting Tip: For genomes with poor annotation quality, provide pre-annotated GenBank files rather than relying on automated gene calling to improve prediction accuracy [35].
Principle: PRISM predicts the chemical structures of natural products from genomic data by correlating biosynthetic logic with structural motifs, then enables matching these predictions to experimental MS/MS data [35].
Procedure:
Application Note: PRISM is particularly valuable for identifying RiPP-derived lipopeptides and other hybrid natural products that combine RiPP biosynthetic logic with additional chemical moieties [39].
Principle: RODEO combines genome mining with heuristic analysis of RiPP biosynthetic genes to identify precursor peptides and predict post-translational modifications, specializing in the discovery of novel ribosomally synthesized peptides [37].
Procedure:
Advanced Application: For radical SAM enzymes involved in RiPP biosynthesis (such as thuricin CD), RODEO can help identify unusual biosynthetic mechanisms requiring enzyme complexes rather than single enzymes [29].
The integration of antiSMASH, PRISM, and RODEO provides a powerful framework for RiPP pathway engineering, enabling rational design of novel bioactive peptides. Three primary engineering strategies have emerged:
Many RiPP pathways employ a leader-peptide-guided biosynthetic logic where tailoring enzymes contain RiPP precursor peptide recognition elements (RREs) that bind to leader peptides, directing catalytic domains to modify core peptide regions [9]. Bioinformatics tools enable the identification of these leader-receptor pairs, facilitating the creation of chimeric systems where native leader peptides are replaced with alternative sequences to redirect biosynthetic enzymes toward non-cognate core peptides [9]. This approach was successfully demonstrated in the engineering of cyanobactins, where leader peptide swapping enabled the production of diverse macrocyclic peptides [9].
Bioinformatic analysis of RiPP enzymes' substrate tolerance enables the rational design of modified core peptides. By identifying positions with relaxed specificity, researchers can introduce non-proteinogenic amino acids, sequence variations, or chemically modified residues to generate "new-to-nature" RiPP compounds [9]. For instance, bioengineering of the lantibiotic nisin incorporated methionine analogs for subsequent click chemistry modification, generating derivatives with altered biological activity [9]. The substrate promiscuity of many RiPP maturase enzymes, particularly those in radical SAM families, makes them ideal candidates for such engineering approaches [29] [39].
BGC mining tools facilitate the identification of promiscuous enzymes that can be repurposed for peptide modification. For example, the combination of RiPP biosynthetic enzymes with established synthetic chemistry approaches has enabled the production of hybrid peptides with non-natural linkages and modifications [9]. Additionally, the discovery of leader-independent enzymes through genome mining has simplified pathway engineering by eliminating the requirement for specific leader sequences [9].
Table 3: Essential Research Reagents and Resources for BGC Analysis
| Resource | Type | Function in BGC Research | Access |
|---|---|---|---|
| MIBiG Database | Reference Data | Curated repository of known BGCs for comparison & validation [35] | http://mibig.secondarymetabolites.org [35] |
| antiSMASH DB | BGC Database | Database of pre-computed antiSMASH results for public genomes [35] | http://antismash-db.secondarymetabolites.org [35] |
| Norine | Nonribosomal Peptide DB | Database of nonribosomal peptides for structure comparison [35] | http://bioinfo.lifl.fr/NRP [35] |
| RRE Toolkits | Enzyme Reagents | RiPP Recognition Element domains for leader peptide binding studies [9] | Custom construction required |
| Chimeric Leader Peptides | Engineering Reagents | Hybrid leader sequences for redirecting biosynthetic enzymes [9] | Synthetic gene construction |
The integrated application of antiSMASH, PRISM, and RODEO provides a comprehensive bioinformatics pipeline for BGC identification and characterization, with particular utility for RiPP biosynthetic pathway engineering. While these tools have dramatically accelerated natural product discovery, important challenges remain, including accurate prediction of novel BGC classes and reliable determination of cluster boundaries [35]. Future developments in machine learning approaches for activity prediction [38] and improved integration with metabolomic data [35] will further enhance our ability to mine microbial genomes for the next generation of therapeutic compounds. For RiPP researchers, these tools collectively enable the transition from discovery to engineering, facilitating the rational design of bioactive peptides with tailored pharmaceutical properties.
The successful engineering of ribosomally synthesized and post-translationally modified peptides (RiPPs) hinges on the selection and optimization of an appropriate heterologous production host. These hosts provide the cellular machinery necessary for the biosynthesis of complex peptide natural products, which often possess promising pharmaceutical properties. Among the most prevalent hosts, the Gram-negative bacterium Escherichia coli and members of the Gram-positive Streptomyces genus each offer distinct advantages and present unique challenges. E. coli is prized for its rapid growth, well-understood genetics, and extensive toolkit for molecular manipulation [40] [41]. In contrast, Streptomyces species, being native producers of numerous clinical drugs, offer a naturally optimized metabolic background for the production of secondary metabolites and possess inherent capabilities for secreting complex, correctly folded proteins [42] [40]. This application note provides a comparative analysis and detailed protocols for utilizing these two powerful hosts within the context of RiPP biosynthetic pathway engineering.
The choice between E. coli and Streptomyces involves careful consideration of their inherent characteristics, which can significantly impact the success of a heterologous expression project. The following table summarizes the key attributes of each host system.
Table 1: Comparison of E. coli and Streptomyces as Heterologous Hosts for RiPP Production
| Feature | E. coli | Streptomyces |
|---|---|---|
| Genetic Manipulation | Highly tractable; vast array of vectors and tools; rapid cloning [41] | More complex; tools available but slower than E. coli; conjugative transfer often required for large DNA [43] [40] |
| Growth Characteristics | Very fast growth (doubling time ~20 min); high-density cultivation well-established [41] | Slow, filamentous growth; complex developmental cycle; industrial fermentation processes available [40] |
| Codon Usage | AT-rich; codon optimization often essential for GC-rich genes from actinobacteria [44] [45] | GC-rich; naturally compatible with actinobacterial genes, reducing need for optimization [42] [40] |
| Protein Secretion | Limited native secretion; requires signal peptides for Sec/Tat pathways; periplasmic production can aid disulfide bond formation [46] [47] | High native secretion capacity; Gram-positive structure (no outer membrane) facilitates release into medium; oxidizing extracellular environment promotes disulfide bond formation [40] [47] |
| Post-Translational Modifications | Limited native capacity for complex PTMs; may lack necessary chaperones or tailoring enzymes for RiPP maturation [40] [41] | Rich in tailoring enzymes (e.g., methyltransferases, oxidases); more likely to possess compatible machinery for RiPP biosynthesis [42] [48] |
| Metabolic Precursor Availability | May lack specialized precursors (e.g., non-proteinogenic amino acids) required for certain RiPPs [40] | Native producer of diverse secondary metabolites; often has inherent supply of key precursors [42] [40] |
| Key Advantage | Speed, ease of use, and high yields for soluble, uncomplexed proteins. | Compatibility for expressing functional biosynthetic enzymes and complex pathways from actinobacteria. |
| Primary Challenge | Inefficient secretion, potential for inclusion body formation, and lack of specialized PTM machinery. | Slow growth, genetic intractability, and complex cellular physiology. |
The following protocol, adapted from successful production of a chimeric human β-defensin, outlines key steps for expressing and purifying a disulfide-bonded antimicrobial peptide in E. coli [45].
Principle: To overcome the toxicity of antimicrobial peptides to the host and the challenge of disulfide bond formation, the peptide of interest is expressed as a fusion partner with a solubility-enhancing tag (Thioredoxin A, TrxA) via a cleavable linker. This promotes solubility, shields the host from the peptide's toxicity, and allows for cytoplasmic expression in a reducing environment. The tag is removed after purification under denaturing conditions.
Materials:
Procedure:
This protocol describes the use of the "Micro-HEP" platform for transferring and integrating a biosynthetic gene cluster (BGC) into an engineered Streptomyces coelicolor chassis for expression [43].
Principle: The BGC is first cloned and engineered in an E. coli strain equipped with a recombineering system. Subsequently, the modified BGC, outfitted with an origin of transfer (oriT) and recombination sites, is conjugally transferred from E. coli to the Streptomyces chassis. Finally, the BGC is integrated site-specifically into the chromosome of the chassis strain via recombinase-mediated cassette exchange (RMCE), enabling stable expression and copy number control [43].
Materials:
Procedure:
Table 2: Essential Research Reagents for Heterologous Expression
| Reagent / Tool | Function | Example Host(s) |
|---|---|---|
| pET Vector Series | High-copy-number expression vectors utilizing the strong T7 promoter for controlled, high-level protein production. | E. coli [45] |
| CASPON-Tag | A fusion tag system that enhances solubility and allows for ultrafast, scarless cleavage and purification of recombinant peptides. | E. coli [49] |
| Micro-HEP Platform | A comprehensive system using engineered E. coli strains for BGC modification/conjugation and optimized Streptomyces chassis for high-efficiency heterologous expression. | E. coli, S. coelicolor [43] |
| RMCE Cassettes (Cre-lox, Vika-vox, etc.) | Modular genetic elements that enable precise, backbone-free integration of large DNA constructs into specific chromosomal loci of the chassis. | Streptomyces spp. [43] |
| S. coelicolor M1146 / A3(2)-2023 | Genetically minimized chassis strains with deletions of multiple endogenous BGCs to reduce metabolic burden and background interference. | Streptomyces spp. [43] [48] |
| Inducible MicL sRNA System | A tool for in-process knockdown of the outer membrane lipoprotein Lpp, increasing membrane permeability and facilitating peptide secretion. | E. coli [49] |
| Elongation factor P-IN-1 | Elongation factor P-IN-1, MF:C14H31N3O2, MW:273.41 g/mol | Chemical Reagent |
| Mono(2-hydroxyisobutyl)phthalate-d4 | Mono(2-hydroxyisobutyl)phthalate-d4, MF:C12H14O5, MW:242.26 g/mol | Chemical Reagent |
The following diagram illustrates the key steps and optimization strategies for producing recombinant peptides in E. coli.
This diagram outlines the multi-step process of expressing a biosynthetic gene cluster in a Streptomyces chassis, highlighting the role of specialized platforms like Micro-HEP.
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a major class of natural products with diverse chemical structures and potent biological activities, including antimicrobial, antiviral, and anticancer properties [50]. The biosynthetic logic of RiPPs is characterized by a conserved pathway: a ribosomally synthesized precursor peptide, typically comprising an N-terminal leader peptide and a C-terminal core peptide, undergoes extensive post-translational modifications (PTMs) catalyzed by specific enzymes before the leader peptide is proteolytically removed to yield the mature natural product [51] [50]. This fundamental architecture provides a direct genetic link between the peptide sequence and the final chemical structure, making RiPPs exceptionally attractive for bioengineering.
Precursor peptide engineering exploits this genetic encodability to generate novel RiPP analogs with enhanced or entirely new functions. The leader peptide plays a critical role in biosynthetic pathways by serving as a recognition motif for the PTM enzymes, often functioning as an allosteric regulator that binds the active form of the biosynthetic enzymes in a conformational selection process [51]. The core peptide, which is modified and becomes the final product, is often tolerant of sequence variations, providing a flexible scaffold for engineering [52]. Two primary strategies have emerged for manipulating this system: site-directed mutagenesis of the core peptide to alter its sequence and properties, and leader-core fusion strategies to redirect enzymatic specificity and enable the modification of non-native substrates. This application note details practical protocols for both approaches, providing researchers with a toolkit for the targeted engineering of RiPP natural products.
Site-directed mutagenesis of the core peptide is the most straightforward method for generating RiPP analogs. This approach capitalizes on the frequent observation that the enzymes responsible for post-translational modifications often exhibit significant substrate tolerance toward the core peptide sequence while maintaining strict recognition of the leader peptide [52]. By systematically altering residues in the core region, researchers can probe structure-activity relationships (SAR), enhance bioactivity, improve stability, or introduce novel chemical functionalities.
The methodology has been successfully applied across multiple RiPP classes. In lasso peptides, such as the RNA polymerase inhibitor microcin J25 (MccJ25), comprehensive scanning mutagenesis has been employed where every residue in the ring (positions 1-7) and tail (positions 9-21) regions was individually substituted with all proteinogenic amino acids [52]. This systematic approach identified residues critical for biosynthesis (e.g., the lactam-forming Gly1 and Glu8) versus those essential for bioactivity (e.g., Tyr9), revealing the differential sequence requirements for production versus function [52]. Similarly, mutagenesis of the anti-mycobacterial lasso peptide lariatin A identified specific residues (Tyr6, Gly11, and Asn14) as crucial for its antibacterial activity, while others (Val15, Ile16, and Pro18) enhanced this activity, leading to the development of analogs with improved potency [52].
Objective: To generate a comprehensive library of core peptide variants for functional screening.
Materials:
Procedure:
Target Selection: Identify the residue(s) in the core peptide for mutagenesis based on structural data or homology analysis.
Primer Design: Design mutagenic primers containing degenerate codons (e.g., NNK, where N=A/T/C/G, K=G/T) to cover all possible amino acid substitutions at the target position. Ensure primers have sufficient flanking sequences (typically 15-20 bp) for efficient annealing.
PCR Amplification: Set up the PCR reaction as follows:
Cycling conditions:
Template Digestion: Add 1 µL of DpnI restriction enzyme directly to the PCR product and incubate at 37°C for 1-2 hours to digest the methylated template DNA.
Transformation: Transform 2-5 µL of the DpnI-treated DNA into competent E. coli cells following standard heat-shock or electroporation protocols.
Library Validation: Plate transformed cells on selective media and incubate overnight. Pick multiple colonies for sequencing to confirm the diversity and representation of the mutant library.
Screening: Express the mutant library and screen for desired properties (e.g., antimicrobial activity, enzyme inhibition, or improved stability) using appropriate functional assays.
Table 1: Key Residues for Mutagenesis in Model RiPPs
| RiPP Class | Example | Critical Residues | Mutagenesis Effect | Citation |
|---|---|---|---|---|
| Lasso Peptide | Microcin J25 | Gly1, Glu8 | Essential for biosynthesis | [52] |
| Lasso Peptide | Microcin J25 | Tyr9, Phe10, Phe19 | Essential for bioactivity | [52] |
| Lasso Peptide | Lariatin A | Tyr6, Gly11, Asn14 | Crucial for anti-mycobacterial activity | [52] |
| Lasso Peptide | Lariatin A | Val15, Ile16, Pro18 | Enhanced antibacterial activity | [52] |
| Lanthipeptide | Salivaricin B | Core Ser/Thr/Cys | Affects dehydration and cyclization pattern | [16] |
The following diagram illustrates the complete site-directed mutagenesis workflow for RiPP engineering:
Leader-core fusion strategies involve appending a recognition leader peptide to a non-cognate core peptide sequence, thereby enabling the modification of heterologous peptides by RiPP biosynthetic enzymes. This approach exploits the modular nature of precursor peptides, where the leader peptide primarily dictates enzyme recognition while the core peptide serves as the modification substrate [51] [50]. The strategy is particularly powerful for introducing complex PTMs into biologically active peptides that lack them naturally, thereby enhancing their stability, activity, or conferring new properties.
The effectiveness of this strategy depends on the specific RiPP class and the compatibility between the leader peptide and the modification enzymes. In class II lanthipeptides, the leader peptide of the precursor peptide SboA was shown to effectively guide the modification enzyme SboM, enabling the installation of thioether rings in the core peptide [16]. Similarly, in lasso peptide biosynthesis, the leader peptide is recognized by the B protein (a peptidase) and C protein (a cyclase), which work in concert to cleave the leader and form the characteristic macrolactam ring [51] [52]. Engineering efforts have demonstrated that fusing different core sequences to native leaders can successfully produce chimeric lasso peptides, provided the essential residues for enzymatic processing (e.g., the lactam-forming Glu/Asp at position 8 or 9) are preserved [52].
Objective: To create chimeric precursor peptides by fusing a leader sequence to heterologous core peptides.
Materials:
Procedure:
Vector Design: Clone the leader peptide sequence into a plasmid flanked by appropriate Type IIS restriction sites (e.g., BsaI) in a destination vector containing the RiPP BGC without the native core sequence.
Insert Preparation: Design double-stranded DNA fragments encoding the core peptide(s) of interest with flanking Type IIS sites creating complementary overhangs to the vector. These can be generated via gene synthesis or PCR assembly.
Golden Gate Assembly: Set up the reaction mixture:
Cycling conditions (25 cycles):
Transformation and Screening: Transform 2-5 µL of the assembly reaction into competent E. coli cells. Screen colonies by colony PCR or restriction digest to identify correct constructs.
Heterologous Expression: Introduce the verified plasmid into an appropriate expression host (e.g., E. coli BL21) containing the necessary modification enzymes.
Product Analysis: Express the engineered gene cluster and analyze the products using mass spectrometry (MALDI-TOF or LC-MS/MS) to confirm successful modification.
Table 2: Leader Peptide Types and Their Applications in Fusion Strategies
| Leader Type | RiPP Class | Key Features | Engineering Application | Citation |
|---|---|---|---|---|
| Canonical Leader | Lanthipeptides, Lasso peptides | Short (<50 aa), unstructured | Guiding PTMs of cognate and non-cognate cores | [6] |
| NHLP (Nitrile Hydratase-like Leader Peptide) | Proteusins | Long (>80 aa), structured tertiary fold | Engages enzymes via large protein-like interfaces | [6] |
| Follower Peptide | Cyanobactins | C-terminal recognition sequence | Directs proteolytic cleavage and cyclization | [51] |
The following diagram illustrates the leader-core fusion strategy for creating chimeric RiPPs:
Cell-free gene expression (CFE) systems provide a powerful platform for rapid prototyping of engineered RiPPs, bypassing the time-consuming steps of molecular cloning and in vivo cultivation [16]. The UniBioCat system, for instance, has been successfully used for the cell-free biosynthesis of lanthipeptides like salivaricin B, where precursor peptides and modification enzymes are co-expressed in vitro to produce mature RiPPs within hours [16]. This approach is particularly valuable for screening large libraries of engineered precursor peptides, as it eliminates potential host cytotoxicity issues and allows direct control over reaction conditions.
Protocol Highlights for Cell-Free RiPP Production:
Recent advances in machine learning have produced computational tools like PepMLM that can design peptide binders to target proteins based solely on sequence information [53]. While not specifically trained for RiPP engineering, such tools can be repurposed to design core peptides with desired binding specificities, which can then be incorporated into leader-core fusion constructs for post-translational modification.
Table 3: Key Research Reagents for Precursor Peptide Engineering
| Reagent / Tool | Category | Function | Example Application | Citation |
|---|---|---|---|---|
| Golden Gate Assembly System | Molecular Cloning | Modular assembly of leader-core fusions | Creating chimeric precursor peptides | [16] |
| ESM-2 Protein Language Model | Computational Design | Peptide binder design via masked language modeling | Designing target-specific core peptides | [53] |
| Cell-Free Gene Expression System | Expression Platform | Rapid in vitro synthesis of RiPPs | High-throughput screening of mutant libraries | [16] |
| DIA-NN with Skyline | Analytical Software | Peptide identification and quantification | Analyzing engineered RiPP products | [54] |
| Chaperone-Enriched E. coli Extracts | Expression Enhancement | Improving solubility of modification enzymes | Cell-free production of lanthipeptides | [16] |
| Riboflavin-5-Phosphate-13C4,15N2-1 | Riboflavin-5-Phosphate-13C4,15N2-1, MF:C17H21N4O9P, MW:462.30 g/mol | Chemical Reagent | Bench Chemicals | |
| Azido-PEG1-Val-Cit-PABC-PNP | Azido-PEG1-Val-Cit-PABC-PNP, MF:C30H39N9O10, MW:685.7 g/mol | Chemical Reagent | Bench Chemicals |
Precursor peptide engineering through site-directed mutagenesis and leader-core fusion strategies represents a powerful and versatile approach for expanding the chemical diversity of RiPP natural products. The protocols outlined in this application note provide researchers with practical methodologies for generating and screening engineered RiPP variants, from targeted single mutations to comprehensive leader-core fusions. When combined with emerging technologies like cell-free biosynthesis and computational design tools, these strategies enable the rapid exploration of RiPP sequence-function relationships and the development of novel peptides with enhanced therapeutic potential. As our understanding of leader peptide recognition and enzyme specificity continues to grow, so too will the precision and sophistication of RiPP engineering platforms.
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a rapidly expanding family of natural products characterized by their genetically encoded precursor peptides and extensive enzymatic tailoring [55]. This biosynthetic logic makes RiPP pathways exceptionally amenable to engineering for generating non-natural analogues and hybrid molecules. The exploitation of enzyme promiscuityâthe ability of enzymes to accept non-native substratesâserves as the fundamental principle enabling the creation of structurally diverse peptide libraries with potential applications in drug discovery and basic research [56]. This document outlines standardized protocols and application notes for harnessing RiPP biosynthetic enzymes to produce novel peptide analogues and hybrids, providing a framework for researchers working at the intersection of chemical biology and natural product discovery.
Table 1: Promiscuous RiPP Enzymes for Peptide Engineering
| Enzyme | Source Pathway | Reaction Catalyzed | Substrate Tolerance | Key Applications | Limitations |
|---|---|---|---|---|---|
| PatA Protease | Cyanobactin (Patellamide) | N-terminal leader peptide cleavage | Broad tolerance to N-methylated residues [57] | Liberation of modified core peptides | Requires conserved GVDAS recognition sequence |
| PsnB Macrocyclase | Plesiocin | Side-chain to side-chain macrolactone formation | Accepts synthetic N-methylated peptides [57] | Cyclization of constrained peptides | Requires Thr-Glu/Asp recognition for lactonization |
| PCY1 Macrocyclase | Orbitide | Head-to-tail macrocyclization | Tolerates single amide methyl group [57] | Backbone cyclization of peptides | May require optimization for multiple N-methylations |
| SpyB/C/D Complex | Streptamidine/Pyrimidone | Pyrimidone ring formation from Asn | Novel nucleobase formation on peptide [55] | Peptide-nucleobase hybrid generation | Requires specific His residue in substrate |
| OphMA Methyltransferase | Omphalotin | Backbone Nα-methylation | Narrow substrate range; auto-methylation [57] | Installation of N-methyl groups | Limited promiscuity; requires engineering |
The strategic application of these enzymes enables the modular construction of complex peptide architectures. For instance, the combination of OphMA-mediated N-methylation with PatA cleavage and PsnB or PCY1 macrocyclization provides a route to generate N-methylated cyclic peptides, which often exhibit improved membrane permeability and metabolic stability [57]. Similarly, the recently discovered pyrimidone-forming enzymes (SpyB/C/D) enable the creation of unprecedented peptide-nucleobase hybrids, expanding the chemical space accessible through RiPP engineering [55].
Table 2: Enzymatic Efficiency with Natural and Non-Natural Substrates
| Enzyme | Substrate Type | Reaction Conditions | Conversion Yield | Key Structural Determinants |
|---|---|---|---|---|
| PatA Protease | Unmethylated peptide (GFPGVDAS/TVAT) | Standard buffer, overnight, 37°C | 65% [57] | GVDAS recognition sequence |
| PatA Protease | N-methylated peptide (GFPGVDAS/TVAT) | Standard buffer, overnight, 37°C | 32% [57] | GVDAS recognition sequence with steric hindrance |
| PsnB Macrocyclase | Unmethylated linear peptide | ATP, Mg2+, 37°C | ~80% (estimated) [57] | TTxxxxEE recognition motif |
| PsnB Macrocyclase | N-methylated linear peptide | ATP, Mg2+, 37°C | ~60% (estimated) [57] | Tolerant to backbone modification |
| SpyB/C/D Complex | Native SpyA precursor | In vivo co-expression in E. coli | Complete conversion [55] | Conserved His for substrate-assisted catalysis |
| YcaO-DHF Enzymes | Thuricin CD precursor | In vivo pathway expression | Unexpected cleavage side-reactions [58] | Context-dependent functionality |
To bioinformatically identify candidate RiPP biosynthetic gene clusters (BGCs) with potential for engineering novel hybrid natural products through enzyme promiscuity.
Identification of candidate hybrid BGCs (e.g., lipoavitide-type RiPP-fatty acid hybrids) [59] and design of non-natural precursor peptides amenable to modification by promiscuous RiPP enzymes.
To clone and express engineered RiPP pathways in heterologous hosts (E. coli) for production of novel peptide analogues and hybrids.
Successful production of modified peptide intermediates and final products, detectable by LC-MS analysis. Typical yields for modified peptides range from 0.1-10 mg/L culture depending on the pathway efficiency [55].
To quantitatively evaluate the substrate promiscuity of RiPP biosynthetic enzymes using synthetic peptide substrates.
Quantitative assessment of enzyme activity with non-natural substrates. For example, PatA maintains 32-65% activity with N-methylated substrates compared to unmethylated controls [57].
To create diverse libraries of RiPP analogues and identify variants with enhanced bioactivity.
Identification of novel RiPP analogues with enhanced or altered bioactivity profiles. Typical success rates range from 0.1-5% of library variants showing interesting bioactivity.
Table 3: Essential Research Reagents and Solutions for RiPP Engineering
| Category | Item | Specification | Application | Key Considerations |
|---|---|---|---|---|
| Cloning Tools | CAPTURE System | Cas12a-assisted cloning | Hybrid cluster capture [59] | Efficient for large gene clusters |
| Golden Gate Assembly | Type IIS restriction enzymes | Library construction [60] | Modular and scalable | |
| Gibson Assembly | Exonuclease, polymerase, ligase | Pathway construction [60] | Seamless and efficient | |
| Expression Systems | E. coli BL21(DE3) | T7 RNA polymerase expression | Heterologous production [60] | High yield, well-characterized |
| Interchangeable Leader Peptides (ILP) | Modular recognition sequences | Mix-and-match pathway engineering [56] | Enables pathway hybridization | |
| Analytical Tools | LC-MS System | High-resolution mass spectrometer | Modification verification [57] [55] | Essential for PTM detection |
| MALDI-TOF MS | Matrix-assisted laser desorption | Rapid screening of modifications [55] | High-throughput capability | |
| Enzyme Resources | PatA Protease | Cyanobactin N-terminal protease | Leader peptide cleavage [57] | Tolerant to N-methylation |
| PsnB Macrocyclase | ATP-grasp ligase | Side-chain cyclization [57] | Compatible with solid-phase synthesis | |
| PCY1 Macrocyclase | Serine protease | Head-to-tail cyclization [57] | Broad substrate tolerance | |
| YcaO Enzymes | Heterocomplex with RRE/DHF | Diverse backbone modifications [55] | Novel chemistry (e.g., pyrimidone) |
The successful implementation of RiPP engineering strategies requires careful integration of these protocols into a cohesive workflow. Begin with comprehensive bioinformatic analysis (Protocol 1) to identify the most promising enzyme candidates and design appropriate precursor peptides. Move to heterologous expression (Protocol 2) to validate pathway functionality, then characterize individual enzyme components in vitro (Protocol 3) to establish substrate scope and limitations. Finally, implement library generation and screening approaches (Protocol 4) to identify novel bioactive compounds.
Throughout this process, maintain rigorous analytical validation using LC-MS and other appropriate techniques to confirm structural features and quantify yields. Note that enzyme promiscuity can sometimes lead to unexpected results, as recent studies have revealed unforeseen cleavage side-reactions and context-dependent enzyme functionalities in RiPP pathways [58]. These observations highlight the importance of detailed biochemical characterization as a prerequisite for successful engineering applications.
The genomic era has revealed a vast untapped resource for natural product discovery: silent (or cryptic) biosynthetic gene clusters (BGCs). These clusters encode the potential for novel bioactive compounds but are not expressed under standard laboratory conditions [61]. This is particularly relevant for Ribosomally synthesized and post-translationally modified peptides (RiPPs), a major class of natural products with diverse therapeutic applications [62]. Unlocking these silent clusters is essential for expanding the pipeline of new drugs, especially amid rising antibiotic resistance [63]. Two powerful, complementary strategies for activating these silent genetic treasures are microbial co-culture, which mimics ecological interactions to trigger natural product synthesis, and genetic refactoring, which involves the rational redesign and reconstruction of BGCs for heterologous expression [62]. This application note provides detailed protocols and frameworks for implementing these strategies within RiPP-focused peptide engineering research.
Microbial co-culture leverages the natural communications and competitive interactions between different microorganisms to activate silent BGCs. This approach is physiologically driven, often leading to the production of novel secondary metabolites that are not observed in axenic cultures [63].
The following workflow, also depicted in Figure 1, outlines a standardized method for conducting a bacteria-fungi co-culture experiment to activate silent fungal BGCs.
Diagram Title: Co-culture Experimental Workflow
Figure 1: A generalized workflow for a microbial co-culture experiment aimed at activating silent biosynthetic gene clusters.
Materials:
Procedure:
Table 1: Essential reagents and materials for microbial co-culture experiments.
| Item | Function/Application | Example/Description |
|---|---|---|
| Actinobacterial Strains | Co-culture partner known to trigger SM production in fungi. | Streptomyces rapamycinicus, S. lividans [63]. |
| Fungal Strains | Target organism with silent/cryptic BGCs. | Aspergillus nidulans, A. fumigatus [63]. |
| Solid Co-culture Media | Provides a surface for microbial interaction and metabolite exchange. | Aspergillus Minimal Medium (AMM), DNase agar (DNM) [63]. |
| Ethyl Acetate | Organic solvent for broad-spectrum extraction of secondary metabolites from agar. | Used for metabolite extraction from the interaction zone [63]. |
| LC-HRMS System | High-resolution metabolomic profiling and dereplication of extracts. | Used to compare co-culture vs. mono-culture metabolite profiles [62]. |
Genetic refactoring involves the complete redesign and synthesis of a silent BGC to optimize its expression in a heterologous host. This strategy circumvents native regulatory constraints and simplifies the genetic architecture for reliable production [62].
The process of refactoring a RiPP BGC for expression in a model host like E. coli involves a multi-stage workflow, as illustrated in Figure 2.
Diagram Title: RiPP Refactoring Workflow
Figure 2: A standard workflow for the genetic refactoring and heterologous expression of a silent RiPP biosynthetic gene cluster.
Materials:
Procedure:
Table 2: Key reagents, tools, and hosts for the genetic refactoring of RiPP BGCs.
| Item | Function/Application | Example/Description |
|---|---|---|
| Bioinformatics Tools | In silico identification and analysis of RiPP BGCs. | AntiSMASH, RODEO, RRE-Finder [62] [64]. |
| Heterologous Hosts | Production chassis for refactored gene clusters. | Escherichia coli [62], Streptomyces species [62]. |
| Standardized Genetic Parts | Modular promoters, RBS, and terminators for predictable expression. | T7/lac promoter, strong E. coli RBS [62]. |
| Assembly System | Cloning methodology for multi-gene constructs. | Golden Gate assembly [62]. |
| Analytical MS | Detection and structural confirmation of produced RiPPs. | LC-MS/MS for mass determination and PTM analysis [62]. |
The strategic choice between co-culture and genetic refactoring depends on the research goals. Co-culture is a powerful, unbiased method for discovering novel metabolites and understanding ecological interactions, making it ideal for initial discovery phases [63]. Genetic refactoring provides a more controlled, scalable, and engineerable system for robust production and detailed mechanistic studies of a specific BGC, which is crucial for applied peptide engineering [62].
For a targeted RiPP engineering project, these strategies can be combined: a silent RiPP BGC first discovered and activated via co-culture can subsequently be refactored for high-yield, reproducible production in a heterologous host. This integrated approach maximizes the potential of microbial genomes to yield new and therapeutic peptides, effectively bridging the gap between natural product discovery and biotechnological application.
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a promising class of natural products for drug development due to their diverse bioactivities and genetically encodable scaffolds. A significant challenge in harnessing these pathways for peptide engineering lies in the inherent enzyme-substrate specificity of RiPP biosynthetic enzymes, which must precisely recognize their precursor peptides among the thousands of other cellular proteins. This specificity is largely mediated by a conserved structural domain known as the RiPP Recognition Element (RRE), a ~90-residue domain that facilitates binding between precursor peptides and their modifying enzymes [65]. The RRE solves a fundamental paradox in RiPP biosynthesis: how enzymes can exhibit strict specificity for their native substrates while displaying remarkable promiscuity toward the core peptide regions that become the final bioactive products [66]. This application note details practical strategies for leveraging RRE domains and chimeric enzyme design to overcome specificity barriers, enabling the production of novel peptide therapeutics with customized modifications.
The RRE domain is structurally homologous to PqqD, a peptide chaperone in pyrroloquinoline quinone biosynthesis, and has been identified in over half of all known prokaryotic RiPP classes [65]. These domains function as essential recognition modules that bind specifically to leader peptide regions of precursor peptides, typically through interactions with short, conserved recognition sequences such as the FNLD motif in lanthipeptides or the YxxP motif in lasso peptides [67]. In radical SAM enzymes such as those found in ranthipeptide pathways, the RRE domain is frequently fused to the catalytic enzyme, as observed with PapB, where deletion of the RRE results in complete loss of function that can only be restored by supplying the RRE in trans [68]. Structural studies of enzymes like SuiB have revealed that while RRE domains facilitate initial engagement, the leader peptide ultimately binds within the catalytic barrel, positioning the core peptide for modification [69].
Table 1: Experimental Validation of RRE Dependence Across RiPP Classes
| RiPP Class | Modifying Enzyme | RRE Dependency | Key Experimental Evidence |
|---|---|---|---|
| Ranthipeptides | PapB | Essential | RRE deletion causes complete loss of function; activity restored with RRE in trans [68] |
| Lanthipeptides | NisB/C | Essential | Leader peptide binding via RRE necessary for dehydration and cyclization [66] |
| Sactipeptides | AlbA | Essential | RRE domain required for radical SAM-mediated thioether formation [66] [65] |
| Lasso Peptides | Specific cyclases | Essential | RRE mediates recognition prior to cyclization [65] |
| Linear Azole-Containing Peptides | BalhC/D | Essential | RRE in C protein guides peptide to active site [65] |
The chimeric leader peptide approach represents a powerful method for redirecting enzyme specificity by concatenating recognition sequences (RS) from multiple RiPP pathways into a single leader peptide [66]. This design creates a multifunctional leader that can recruit enzymes from unrelated RiPP classes to modify a single core peptide. The experimental workflow involves:
Identification of Recognition Sequences: Determine essential RS motifs within native leader peptides through sequence alignment and mutational analysis. For example, the NisA leader contains critical residues between positions -22 and -1 for NisB recognition [66].
Strategic Concatenation: Fuse RS elements while maintaining appropriate spacing between each RS and its corresponding modification sites in the core peptide. The successful Hyp1.1 chimera incorporated the HcaA RS followed by the essential region of the NisA leader [66].
Core Peptide Optimization: Engineer core regions with appropriate spacing between modification sites for different enzyme classes, typically including a "GGRCG" motif for thiazoline formation followed by lanthipeptide core sequences [66].
Table 2: Chimeric Leader Design Specifications for Hybrid RiPP Production
| Design Parameter | Technical Considerations | Validated Examples |
|---|---|---|
| Recognition Sequence Selection | Conserved motifs essential for RRE binding (e.g., FNLD for lanthipeptides) | HcaA and NisA recognition sequences [66] |
| Linker Design | Maintain natural spacing between RS and core modification sites (typically 4-8 residues) | GGRCG motif positioned at appropriate distance from HcaA RS [66] |
| Core Region Engineering | Include substrate sequences for multiple enzyme classes with compatible spacing | Combination of thiazoline-forming and lanthipeptide core sequences [66] |
| Expression System | Co-expression with modifying enzymes in heterologous hosts (e.g., E. coli) | Plasmid-based expression in E. coli [66] |
Materials Required:
Procedure:
Diagram 1: Chimeric leader strategy for hybrid RiPP production. Recognition sequences from different pathways enable single precursor to recruit multiple enzymes.
Table 3: Essential Research Tools for RiPP Engineering Studies
| Reagent/Tool | Specific Application | Function in Experimental Workflow |
|---|---|---|
| RODEO Bioinformatic Tool | RRE domain identification and RiPP BGC annotation | Predicts precursor peptides and identifies RRE-containing enzymes in genomic data [67] |
| Heterologous Expression Systems (e.g., E. coli) | Pathway reconstruction and engineering | Enables expression of RiPP pathways from unculturable organisms or engineered variants [68] [66] |
| MALDI-TOF Mass Spectrometry | Modification tracking and product verification | Monitors mass shifts corresponding to specific PTMs (e.g., -12 Da for thioether formation) [68] |
| Leader Peptide Binding Assays | RRE recognition specificity validation | Measures binding affinity between RRE domains and leader peptides (SPR, ITC, etc.) [65] |
| Radical SAM Reconstitution Systems | In vitro activity assays for rSAM enzymes | Provides controlled conditions for studying RRE-dependent enzymes (SAM, reductant) [68] |
Diagram 2: Engineering framework for addressing enzyme-substrate specificity. Complementary strategies expand RiPP biosynthetic capabilities.
RRE Swapping and Engineering: The modular nature of RRE domains enables their transplantation between enzyme systems to redirect specificity. Experimental evidence confirms that RRE domains maintain function when provided in trans, as demonstrated by the restoration of PapB activity when its RRE was supplied separately [68]. This approach allows researchers to:
Optimization Considerations: Successful implementation requires maintaining critical recognition residues while optimizing interfacial compatibility between fused domains. Structural studies indicate that leader peptides primarily contact the third alpha helix and third beta strand of the RRE [67], highlighting these as key regions for engineering efforts.
Objective: Determine whether a putative RiPP-modifying enzyme requires its RRE domain for activity.
Procedure:
Expected Results: For PapB, RRE deletion abolished thioether formation, but activity was restored when the RRE was provided in trans [68].
Objective: Produce a hybrid RiPP containing modifications from two different classes.
Procedure:
Troubleshooting: Incompatibility issues may arise from competing for Cys residues; address by strategic spacing of modification sites or sequential expression strategies [66].
RRE domains and chimeric enzyme design represent complementary, powerful strategies for overcoming the inherent enzyme-substrate specificity limitations in RiPP engineering. By understanding the molecular basis of RRE-mediated recognition and applying rational design principles for chimeric leaders, researchers can significantly expand the biosynthetic landscape accessible for peptide-based therapeutic development. The protocols and approaches outlined herein provide a framework for designing and producing novel RiPP variants with customized modification patterns and enhanced bioactivities. As these engineering strategies continue to evolve, they promise to unlock the full potential of RiPP pathways for generating next-generation peptide therapeutics.
This application note provides a structured framework for optimizing post-translational modification (PTM) efficiency in ribosomally synthesized and post-translationally modified peptide (RiPP) biosynthesis. Focusing on the coordination of multi-enzyme complexes and strategic reaction order, we present experimental protocols and analytical methodologies to enhance the yield and fidelity of complex bioactive peptides. The strategies outlined herein enable researchers to overcome significant challenges in natural product engineering, particularly in activating silent biosynthetic gene clusters (BGCs) and optimizing modification sequences. By integrating machine learning prediction with high-throughput experimental validation, this guide supports accelerated development of novel peptide therapeutics with potential applications against antimicrobial resistance, cancer, and metabolic disorders.
RiPPs represent a diverse superfamily of natural products with immense potential for drug development, featuring complex structures generated through enzymatic modifications of ribosomal precursor peptides [4] [9]. The biosynthesis follows a conserved logic: a ribosomally synthesized precursor peptide typically containing an N-terminal leader peptide and a C-terminal core peptide is modified by specialized enzymes, after which the leader peptide is proteolytically removed to yield the mature product [4]. The core challenge in RiPP engineering lies in controlling the efficiency of multiple PTMs, as the order of enzymatic actions significantly impacts final product yield and bioactivity. Research indicates that enzyme-specific PTM patterns govern substrate recognition and modification kinetics, creating complex interdependencies that must be understood for effective pathway optimization [70] [71]. This document establishes standardized protocols for deciphering and optimizing these complex enzymatic networks to maximize RiPP production and engineer novel bioactive compounds.
Recent advances in machine learning (ML) have enabled more accurate prediction of enzyme-substrate relationships, providing critical data for planning modification sequences. The following table summarizes performance metrics for established PTM prediction methods:
Table 1: Performance Metrics of PTM Prediction Methods
| Method | Application | Validation Rate | Key Performance Features |
|---|---|---|---|
| ML-hybrid ensemble models [70] | SET8 methyltransferase substrate prediction | 37-43% | Marks important performance increase over traditional in vitro methods |
| Permutation array-based prediction [70] | SET8 methylation site identification | Low precision (26/346 hits validated) | Limited by inability to capture structural features |
| PeSA2.0 motif generation [70] | Enzyme recognition motif characterization | Normalized score cutoff >0.5 | Identifies weighted AA preferences at each position |
| SCASP-PTM multi-enrichment [72] | Simultaneous PTM profiling (phosphorylation, ubiquitination, acetylation, glycosylation) | Desalting-free sequential enrichment | Streamlined workflow for comprehensive PTM analysis |
The quantitative data demonstrates that ML-driven approaches significantly outperform conventional methods, providing more reliable substrate predictions for planning efficient modification sequences. The 37-43% validation rate for ML-hybrid models represents a substantial improvement in accurately identifying enzyme-specific PTM sites, enabling better planning of multi-enzyme reaction sequences [70].
The following table details essential research reagents and their applications in RiPP engineering and PTM optimization studies:
Table 2: Essential Research Reagents for RiPP Pathway Engineering
| Reagent/Category | Function/Application | Specific Examples |
|---|---|---|
| Temporary Protecting Groups [73] | N-terminus protection during synthetic peptide production | Boc (tert-butoxycarbonyl), Fmoc (9-fluorenylmethoxycarbonyl) |
| Permanent Protecting Groups [73] | Side chain protection during peptide synthesis | Bzl (benzyl), tBu (tert-butyl) derivatives |
| Coupling Reagents [74] [73] | Activate carboxyl groups for peptide bond formation | DCC (dicyclohexylcarbodiimide), DIC (diisopropylcarbodiimide), HOBt (1-hydroxybenzotriazole) |
| Cell-Free Protein Synthesis Systems [75] | In vitro transcription/translation for pathway prototyping | E. coli extracts, yeast cell-free systems |
| Solid Supports [74] [73] | C-terminal protection and simplified purification in SPPS | Polystyrene, polyacrylamide resins |
| Cleavage Reagents [73] | Final deprotection after peptide synthesis | TFA (trifluoroacetic acid), HF (hydrogen fluoride) |
| RiPP Modification Enzymes [9] | Install complex PTMs on precursor peptides | Lanthipeptide dehydratases, cyclodehydratases, cytochrome P450 enzymes |
Purpose: To identify potential modification sites for specific enzymes to inform reaction order planning.
Background: ML-hybrid approaches combine high-throughput in vitro peptide array experiments with computational modeling to predict enzyme-substrate relationships with higher accuracy than traditional methods [70].
Procedure:
Applications: This protocol successfully identified 64 unique deacetylation sites for SIRT2 and revealed changes in SET8-regulated substrate networks among breast cancer missense mutations [70].
Purpose: To optimize enzyme recognition and modification efficiency through strategic leader peptide manipulation.
Background: Most RiPP tailoring enzymes contain RiPP precursor peptide recognition elements (RREs) that bind leader peptides, positioning catalytic domains to modify core peptide regions [9].
Procedure:
Applications: Leader peptide engineering has enabled production of hybrid, new-to-nature ribosomal natural products and improved modification efficiency across diverse RiPP classes [9].
Purpose: To rapidly test multi-enzyme reaction orders and coordination without cellular constraints.
Background: Cell-free gene expression (CFE) systems provide a quasi-chemical bioreactor platform for rapid prototyping of natural product biosynthetic pathways, enabling precise control over enzyme and substrate concentrations [75].
Procedure:
Applications: CFE technology has been successfully applied to characterize biosynthetic pathways for ribosomal peptides and engineer novel antimicrobial compounds [75].
Diagram 1: RiPP PTM Optimization Workflow
Diagram 2: Enzyme Engineering Strategies
The strategic coordination of multi-enzyme complexes and optimization of reaction order represents a critical advancement in RiPP bioengineering. By integrating computational prediction with high-throughput experimental validation, researchers can systematically overcome the historical challenges of PTM efficiency in complex peptide synthesis. The methodologies outlined in this application note provide a structured framework for elucidating and optimizing enzyme-substrate relationships, enabling the production of novel bioactive compounds with enhanced therapeutic potential. As peptide-based therapeutics continue to gain prominence in addressing antimicrobial resistance, cancer, and metabolic disorders, these optimization strategies will play an increasingly vital role in accelerating drug discovery and development pipelines.
Within the field of peptide engineering, ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a promising source of bioactive compounds due to their vast structural diversity and high target specificity [9] [10]. However, the structural characterization of modified peptides, particularly those obtained in low yields from biosynthetic pathways, presents significant analytical challenges. Determining the precise chemical structure, including the identity and location of post-translational modifications (PTMs), is essential for understanding structure-activity relationships and guiding the rational engineering of new analogs [9]. This application note details the synergistic use of Mass Spectrometry (MS) and Nuclear Magnetic Resonance (NMR) spectroscopy to overcome these hurdles, providing detailed protocols tailored for researchers in RiPP engineering and drug development.
The analysis of modified peptides from RiPP pathways is fraught with difficulties, which are compounded when sample quantities are limited. The table below summarizes the core challenges and their implications for structural biology.
Table 1: Key Challenges in the Structural Characterization of Modified RiPPs
| Challenge | Impact on Structural Characterization |
|---|---|
| Low Abundance & Yield | Limits application of high-resolution techniques; necessitates highly sensitive methods [76]. |
| Complex Post-Translational Modifications (PTMs) | Difficult to identify and locate using standard proteomics; requires targeted MS approaches and high-resolution NMR [77] [9]. |
| Structural Dynamics & Compact Folds | Solution-state techniques like NMR are needed to capture dynamics, but suffer from low sensitivity with scarce samples [78]. |
| Sample Purity & Integrity | Low yields increase the risk of sample loss during purification, potentially compromising final sample quality for NMR [79]. |
An integrated approach that leverages the complementary strengths of MS and NMR is the most effective strategy for characterizing challenging peptide targets. The following workflow is designed to maximize information gain from precious, low-yield samples.
Figure 1: An integrated MS and NMR workflow for determining the structure of modified peptides from low-yield sources. The pathway shows how the techniques inform one another sequentially.
Mass spectrometry provides unparalleled sensitivity for analyzing peptide mass, sequence, and PTMs, making it ideal for low-yield samples [77].
4.1.1 Protocol: Targeted Peptide Quantitation using Multiple-Reaction Monitoring (MRM)
This protocol is adapted from targeted proteomics approaches for precise and sensitive peptide measurement [77].
4.1.2 Protocol: Peptide Sequencing and PTM Identification using MS/MS
NMR spectroscopy provides atomic-level information on peptide structure, dynamics, and PTMs in solution, and is considered a gold standard for such characterizations [80]. The following strategies are critical for low-yield scenarios.
4.2.1 Protocol: Optimizing NMR for Low-Concentration Peptides
4.2.2 Protocol: NMRseq for Sequence Tag Determination
This protocol allows for the non-destructive determination of sequence tags, which can be used for database searching to identify the full peptide sequence [78].
The following table lists key reagents and materials essential for successfully executing the protocols described above.
Table 2: Essential Research Reagents and Materials for Characterizing Modified Peptides
| Item | Function/Application | Specifications & Notes |
|---|---|---|
| Stable Isotope-Labeled Amino Acids (e.g., ¹âµN-Ala, ¹³Câ/¹âµN-Val) [79] | Selective isotopic labeling for NMR; simplifies spectra and facilitates assignment in sparse labeling strategies. | > 98% isotopic purity; controls metabolic scrambling in expression hosts. |
| NMR CryoProbe [80] | Enhances NMR sensitivity for low-yield samples. | 1.7 mm or 3 mm configuration for small volume samples; provides significant signal-to-noise improvement. |
| Specialized MS Matrices (e.g., 1,5-Diaminonaphthalene) [78] | Facilitates sequencing of native, disulfide-rich peptides by MALDI-ISD MS. | Enables reduction in the laser plume and promotes in-source decay fragmentation. |
| Fmoc-protected PTM Amino Acids [81] | Chemical synthesis of phospho-, acetyl-, or methyl-peptides for use as analytical standards. | Enables orthogonal protection strategies during Solid-Phase Peptide Synthesis (SPPS). |
| Deuterated NMR Solvents (e.g., DâO) [79] | Solvent for NMR spectroscopy; provides lock signal. | 99.9% D or higher; essential for all NMR experiments. |
The structural elucidation of modified peptides from RiPP pathways, especially those produced in low yields, demands a carefully orchestrated, multi-technique approach. By integrating the high sensitivity and PTM-screening capabilities of mass spectrometry with the atomic-resolution and solution-state structural details provided by advanced NMR, researchers can overcome these significant analytical challenges. The detailed protocols and workflows outlined here provide a actionable roadmap for RiPP engineers and drug development scientists to confidently characterize and validate complex peptide structures, thereby accelerating the development of next-generation peptide therapeutics.
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a promising class of natural products for drug development due to their structural diversity and potent bioactivities. Among these, the two-component sactibiotic thuricin CD has attracted significant research attention for its narrow-spectrum activity against clinically relevant pathogens like Clostridioides difficile. Thuricin CD consists of two peptides, Trnα and Trnβ, which undergo extensive post-translational modification to form characteristic sulfur-to-α-carbon thioether crosslinks. For over a decade, the biosynthetic mechanism underlying these modifications remained elusive, with the prevailing assumption that the two radical S-adenosylmethionine (rSAM) enzymes in the cluster, TrnC and TrnD, independently modified separate precursor peptides. Recent groundbreaking research has overturned this paradigm, revealing a complex synergistic relationship between TrnC and TrnD that challenges conventional understanding of RiPP biosynthetic pathways. This application note examines the unconventional enzyme cooperation in thuricin CD biosynthesis and its implications for peptide engineering research.
The biosynthetic gene cluster for thuricin CD encodes two radical SAM enzymes (TrnC and TrnD) and two precursor peptides (TrnA and TrnB). Previous models assumed independent functionality, where each enzyme would specifically modify one precursor peptide. However, recent investigations demonstrate that neither TrnC nor TrnD can effectively install thioether crosslinks on the precursor peptides individually [82]. Instead, these enzymes form a tight complex and collaboratively catalyze thioether crosslinking on both TrnA and TrnB [82] [83].
This cooperative mechanism represents a significant departure from the biosynthetic strategies observed in related two-component lanthipeptides, where modification enzymes typically work independently on their respective substrate peptides. Even more surprisingly, despite both TrnC and TrnD being active rSAM enzymes, only the rSAM activity of TrnC is strictly essential for thioether crosslinking [82]. This finding indicates an asymmetric functional relationship within the complex, suggesting TrnD may play a structural or regulatory role that enhances or enables TrnC's catalytic function.
Table 1: Key Experimental Findings in Thuricin CD Biosynthesis
| Experimental Approach | Key Finding | Implication |
|---|---|---|
| Individual coexpression of TrnA with TrnC or TrnD | No thioether crosslinks formed | Neither enzyme functions independently on precursor peptides |
| Coexpression of TrnA with both TrnC and TrnD | â6 Da modification (three thioether crosslinks) | Collaborative enzyme action required for modification |
| Coexpression of TrnB with both TrnC and TrnD | â6 Da modification (three thioether crosslinks) | Complex modifies both precursor peptides |
| In vitro reconstitution | TrnC and TrnD form tight complex | Physical interaction enables functional cooperation |
| Activity assessment | Only TrnC's rSAM activity essential | Functional asymmetry within enzyme complex |
The collaborative model emerged from systematic coexpression studies in E. coli. When TrnA was coexpressed individually with TrnC or TrnD, high-resolution LC-MS analysis detected no modifications on the precursor peptide [82]. However, when TrnA was coexpressed with both TrnC and TrnD, the resulting peptide exhibited a â6 Da mass shift corresponding to the formation of three thioether crosslinks [82]. Parallel experiments with TrnB yielded identical results, confirming that both precursor peptides require the collaborative action of both enzymes for proper modification.
The functional product was confirmed through bioactivity testing against Bacillus cereus. The modified peptides (trnαⲠand trnβâ²) showed potent antibacterial activity only when combined, with a minimal inhibitory concentration of 3.1 μM, while individually exhibiting no inhibition even at 50 μM [82]. This demonstrates that the synergistically modified peptides retain the characteristic bioactivity of natural thuricin CD.
Table 2: Enzyme Characteristics and Metallocluster Composition
| Enzyme | Iron Content (mol/mol protein) | Sulfur Content (mol/mol protein) | Essential for Catalysis | Proposed Role |
|---|---|---|---|---|
| TrnC | 11.3-11.6 | 10.5-10.9 | Yes | Primary catalytic enzyme |
| TrnD | 9.1-9.4 | 8.2-8.6 | No (structural) | Complex stabilization, substrate positioning |
Table 3: Essential Research Reagents for Thuricin CD Biosynthesis Studies
| Reagent | Function/Application | Key Features |
|---|---|---|
| N-terminal TF-fused TrnA/TrnB | Precursor peptide production | Enhances solubility and expression in E. coli; removable by HRV-3C protease |
| 6Ã His-tagged TrnC/TrnD | Enzyme purification | Enables anaerobic purification via Ni-NTA chromatography |
| Recombinant HRV-3C protease | Leader peptide removal | Cleaves trigger factor fusion to yield mature precursor peptides |
| TCEP (tris(2-carboxyethyl)phosphine) | Disulfide bond reduction | Reduces disulfide bonds in precursor peptides without interfering with thioether bonds |
| N-ethylmaleimide (NEM) | Cysteine derivatization | Confirms free cysteine residues in unmodified peptides |
| GluC endoprotease | Leader peptide removal | Generates mature-like peptides (trnαⲠand trnβâ²) for activity assays |
| S-adenosylmethionine (SAM) | Cofactor for rSAM enzymes | Essential for radical generation and thioether bond formation |
| Dithionite | Chemical reductant | Provides electrons for rSAM cluster reduction in in vitro assays |
Objective: Recombinant production of unmodified precursor peptides TrnA and TrnB.
Methodology:
Key Considerations:
Objective: Assess TrnC and TrnD activity on precursor peptides in vivo.
Methodology:
Expected Results:
Objective: Purify active TrnC and TrnD and reconstitute activity in vitro.
Methodology:
Key Considerations:
The collaborative mechanism of TrnC and TrnD represents a significant paradigm shift in RiPP engineering with far-reaching implications:
The thuricin CD system demonstrates that bioinformatic predictions based on gene organization can be misleading. The presence of two modification enzymes in a RiPP cluster does not necessarily indicate division of labor, but may instead indicate requirement for complex formation [82] [84]. This finding calls for more comprehensive characterization of RiPP biosynthetic pathways before engineering attempts.
The functional asymmetry in the TrnC-TrnD complex, where both enzymes are catalytically capable but only one is essential, suggests sophisticated regulatory mechanisms in RiPP biosynthesis [82]. This knowledge informs strategic engineering of similar systems:
From a pharmaceutical perspective, the narrow-spectrum activity of thuricin CD against C. difficile presents attractive therapeutic characteristics. The biosynthetic insights enable production of novel analogs with potentially optimized properties [85]. The membrane-targeting mechanism, involving collapse of membrane potential and subsequent cell death, offers a distinct mode of action from conventional antibiotics [85].
The unconventional enzyme cooperation in thuricin CD biosynthesis represents a significant advancement in our understanding of RiPP biosynthetic pathways. The collaborative model, featuring a tight complex between TrnC and TrnD with functional asymmetry, challenges previous assumptions about two-component antibiotic biosynthesis and provides valuable insights for future engineering efforts. These findings underscore the importance of detailed biochemical characterization before attempting pathway engineering or combinatorial biosynthesis. The experimental protocols and reagents outlined here provide a framework for investigating similar complex systems in other RiPP families. As our understanding of these sophisticated biosynthetic systems grows, so too does our ability to harness them for development of novel therapeutic agents with activity against clinically challenging pathogens.
The engineering of ribosomally synthesized and post-translationally modified peptides (RiPPs) demands advanced screening methodologies capable of interrogating vast sequence spaces to identify variants with optimized therapeutic properties. Key among these methodologies are mRNA display, yeast surface display, and two-hybrid systems, which establish critical genotype-phenotype linkages essential for directed evolution campaigns. While RiPP pathway engineering aims to overcome inherent limitations in clinical translatability, including suboptimal bioavailability and proteolytic stability, the selection of appropriate screening technologies fundamentally dictates success in identifying functional peptides with desired characteristics [10]. Each platform offers distinct advantages in library size, selection stringency, and compatibility with diverse experimental conditions, enabling researchers to tailor their approach based on specific protein engineering objectives.
The selection of an appropriate high-throughput screening method represents a critical decision point in any peptide engineering pipeline. mRNA display enables in vitro selection from exceptionally diverse libraries (>10^13 variants) through covalent mRNA-peptide fusions, permitting stringent selection conditions that would be incompatible with cellular systems [86] [87]. Yeast surface display leverages eukaryotic processing capabilities to express complex proteins while providing quantitative screening via flow cytometry, though with more limited library diversity (10^6-10^9 variants) [88] [89]. Two-hybrid systems, available in both yeast and mammalian configurations, enable direct detection of protein-protein interactions within cellular environments, making them particularly valuable for identifying binding partners in biologically relevant contexts [90] [91]. Understanding the capabilities and limitations of each system is essential for designing effective screening strategies for RiPP engineering.
Table 1: Comparison of High-Throughput Screening Platforms
| Platform | Theoretical Library Diversity | Genotype-Phenotype Linkage | Key Advantages | Primary Limitations |
|---|---|---|---|---|
| mRNA Display | 10^12-10^14 [86] [87] | Covalent (puromycin-mediated) [86] | Largest library size; flexible selection conditions [86] | Technically demanding; no native post-translational modifications [92] |
| Yeast Surface Display | 10^6-10^9 [88] | Non-covalent (cell surface tethering) [89] | Eukaryotic processing; flow cytometric quantification [88] | Limited library size; cellular transformation bottleneck [86] |
| Mammalian Two-Hybrid | Limited by transfection efficiency | Protein-protein interaction reconstitution [90] | Native cellular environment; detects weak interactions | Low throughput; cellular constraints on selection conditions [90] |
| Yeast Two-Hybrid | ~10^6 [91] | Protein-protein interaction reconstitution | Simplicity; genome-wide interaction mapping | False positives; limited to nuclear proteins [91] |
mRNA display represents a powerful acellular selection platform that overcomes the library size limitations inherent to cellular display methods by establishing a covalent linkage between a peptide and its encoding mRNA. This technology enables the in vitro selection of functional polypeptides from libraries exceeding 10^13 different variants, far surpassing the diversity accessible through cellular transformation [86] [87]. The fundamental innovation involves the formation of a covalent bond between the C-terminus of a translated protein and the mRNA that encoded it, achieved through the incorporation of puromycinâan antibiotic that mimics aminoacyl-tRNAâinto the mRNA template [86]. This stable mRNA-protein fusion allows application of stringent selection conditions, including extreme temperatures, denaturants, and proteases, that would disrupt biological systems or non-covalent complexes used in other display technologies.
The applications of mRNA display span multiple domains of peptide and protein engineering. The technology has been successfully employed for selecting high-affinity binding peptides against diverse targets including cell surface receptors, intracellular proteins, and small molecules [86] [93]. More recently, mRNA display has proven particularly valuable for generating constrained peptide architectures, with demonstrations showing that bicyclic peptides selected through mRNA display exhibit superior affinity and stability compared to their linear or monocyclic counterparts [93]. Additionally, the platform has been adapted for the directed evolution of enzymes, including the development of novel RNA ligases and other biocatalysts, highlighting its versatility beyond simple binding selections [87]. This flexibility makes mRNA display especially suitable for RiPP engineering, where introduction of constrained topologies can enhance proteolytic stability and bioavailability.
Traditional mRNA display protocols required 4-7 days per selection round, but significant streamlining has reduced this timeframe to just two days while maintaining the critical advantages of the technology [92]. The optimized protocol incorporates several key modifications that enhance efficiency and practicality while preserving the ability to generate highly diverse libraries.
Day 1: Library Preparation and Fusion Formation
Day 2: Selection and Amplification
This streamlined protocol maintains the exceptional diversity of traditional mRNA display (>10^13 variants) while dramatically improving accessibility and throughput. The modifications collectively address major practical limitations that previously hindered widespread adoption of mRNA display, making it a more viable option for routine protein engineering applications.
Recent methodological innovations have expanded the capabilities of mRNA display for complex peptide engineering applications. A particularly significant advancement involves the development of efficient cyclization strategies that enable generation of monocyclic and bicyclic peptide libraries directly within the mRNA display format [93]. These constrained architectures demonstrate remarkable advantages over linear peptides, including enhanced target affinity, improved proteolytic stability, and greater specificity. In one compelling demonstration, mRNA display was used to select bicyclic peptide inhibitors against fibroblast growth factor receptor 3c (FGFR3c) that significantly outperformed both linear and monocyclic formats in both binding affinity and stability assays [93]. This capability aligns exceptionally well with the needs of RiPP engineering, where introduction of structural constraints often enhances therapeutic potential.
The application of high-throughput sequencing (HTS) to mRNA display selections represents another technological frontier with particular relevance for RiPP pathway engineering. Deep sequencing of selection pools across multiple rounds enables quantitative analysis of sequence-function relationships and evolutionary trajectories [86]. This approach provides unprecedented resolution into the molecular fitness landscape, revealing consensus motifs, functional trade-offs, and structural constraints that govern peptide function. For RiPP engineering, which often involves navigating complex sequence spaces to optimize bioactivity and expressibility, this rich dataset can guide rational design decisions and library design strategies. Furthermore, the combination of HTS with mRNA display allows researchers to move beyond simple affinity selections to more complex screening objectives, such as balancing affinity with developability parametersâa critical consideration for therapeutic application of engineered RiPPs.
Yeast surface display provides a eukaryotic platform for directed evolution by tethering proteins to the cell surface of Saccharomyces cerevisiae, typically through fusion with agglutinin adhesion receptors (Aga1p and Aga2p) [88] [89]. This system leverages the eukaryotic secretory pathway to facilitate proper protein folding, disulfide bond formation, and post-translational modificationsâcapabilities particularly advantageous for expressing complex mammalian proteins and antibodies. The display valency can be controlled through regulation of fusion expression, with typical systems exhibiting 10^4-10^5 copies per cell [88]. Library diversity in yeast surface display is constrained by transformation efficiency, typically reaching 10^6-10^9 variants, which, while substantially smaller than mRNA display libraries, remains sufficient for many protein engineering applications [86] [88].
The applications of yeast surface display are particularly well-established in the antibody engineering domain, where it has been extensively used for affinity maturation of scFv and Fab fragments [88]. The platform enables quantitative screening via flow cytometry, allowing multiparameter sorting based on expression level, stability, and binding affinity. This capability facilitates direct evolution of protein stability in addition to binding function, as properly folded proteins typically display higher expression levels on the yeast surface [88]. Furthermore, the eukaryotic quality control mechanisms inherent to yeast surface display provide inherent selection for well-behaved, expressible proteinsâa valuable feature for engineering therapeutic candidates. While less frequently applied to RiPP engineering specifically, the technology offers relevant capabilities for engineering peptide binders, particularly those requiring disulfide stabilization or eukaryotic processing.
The standard yeast surface display protocol involves iterative cycles of library transformation, magnetic-activated cell sorting (MACS) for initial enrichment, and fluorescence-activated cell sorting (FACS) for high-stringency selection based on quantitative binding measurements [88].
Library Construction and Transformation
Selection and Screening
Characterization and Analysis
The entire process from library construction to identification of affinity-matured variants typically requires 4-6 weeks, with the primary bottleneck being the eukaryotic growth requirements and sorting timeline [88]. Despite this moderate throughput, the quantitative screening capabilities and eukaryotic processing make yeast surface display a valuable platform for engineering complex proteins, including those relevant to RiPP pathways.
Table 2: Key Research Reagent Solutions for High-Throughput Screening
| Reagent/System | Manufacturer/Reference | Function in Screening | Application Notes |
|---|---|---|---|
| PURExpress In Vitro Translation System | New England Biolabs [92] | Reconstituted translation machinery for mRNA display | Nuclease/protease-free; high ribosome concentration [92] |
| CheckMate Mammalian Two-Hybrid System | Promega [90] | Detects protein-protein interactions in mammalian cells | Uses GAL4 DNA-BD and VP16 AD; Dual-Luciferase readout [90] |
| Yeast Surface Display Vectors (pCTCON) | Academic sources [88] | Display scaffold for yeast surface engineering | Inducible expression; epitope tags for quantification [88] |
| Matchmaker Yeast Two-Hybrid System | Clontech [91] | Detects protein-protein interactions in yeast | GAL4-based; nutritional and colorimetric reporters [91] |
| Microfluidic Drop-Making Devices | Custom fabrication [94] | Encapsulation for drop-based screening | Enables IVT2H screening in picoliter volumes [94] |
Two-hybrid systems represent a fundamentally different approach to screening, focusing on detecting protein-protein interactions within cellular environments rather than displaying libraries for in vitro selection. The core principle involves reconstituting a transcription factor through interaction between two test proteins, thereby activating reporter gene expression [90] [91]. The original yeast two-hybrid (Y2H) system utilizes the modular GAL4 transcription factor, with the DNA-binding domain (DBD) fused to a "bait" protein and the activation domain (AD) fused to a "prey" protein [91]. Interaction brings the DBD and AD into proximity, activating transcription of reporter genes that enable growth selection or colorimetric screening. Mammalian two-hybrid systems operate on similar principles but function in mammalian cells, providing a more native physiological context with appropriate post-translational modifications and cofactor availability [90].
The applications of two-hybrid systems extend beyond simple interaction detection to include identification of novel binding partners from cDNA libraries, mapping interaction domains, and characterizing interaction specificity [90] [91]. While traditionally used for interactome mapping, these systems have been adapted for engineering applications, including the selection of affinity-matured variants and the identification of peptide inhibitors of protein-protein interactions. The recent development of drop-based in vitro two-hybrid (IVT2H) methods has further expanded these capabilities by combining the sensitivity of two-hybrid detection with the throughput of microfluidic screening [94]. This innovation enables screening of peptide libraries up to 10^6 variants for disruption or stabilization of specific protein-protein interactionsâa capability with direct relevance to RiPP engineering, where modulation of biosynthetic enzyme interactions could enable pathway optimization.
The CheckMate Mammalian Two-Hybrid System provides a standardized protocol for detecting protein-protein interactions in mammalian cells, offering the advantage of proper eukaryotic processing in a biologically relevant environment [90].
Vector Construction
Cell Transfection and Reporter Assay
Data Interpretation
While the throughput of traditional mammalian two-hybrid systems is limited by transfection efficiency and luciferase assay scalability, recent innovations including microfluidic encapsulation have begun to address these limitations, opening new possibilities for higher-throughput interaction screening in physiologically relevant contexts.
The integration of high-throughput screening technologies into RiPP engineering pipelines requires careful experimental design and understanding of workflow logistics. The following diagrams illustrate key procedural elements and decision points for implementing these technologies effectively.
Diagram 1: mRNA display screening workflow
Diagram 2: Yeast surface display screening workflow
Selection of an appropriate high-throughput screening platform depends on multiple factors, including target characteristics, desired peptide properties, and available resources. For RiPP engineering applications specifically, several considerations should guide this decision:
For RiPP pathway engineering specifically, mRNA display offers compelling advantages for initial discovery of novel bioactive sequences, particularly when coupled with cyclization strategies to enhance stability [93]. Yeast surface display provides complementary capabilities for optimizing expression and stability of engineered peptides, while two-hybrid systems enable screening for specific interactors within biosynthetic pathways. Integrating these technologies across different stages of the engineering pipeline can maximize the probability of success in developing clinically viable RiPP-based therapeutics.
The continuing evolution of high-throughput screening technologies provides peptide engineers with an expanding toolkit for addressing the unique challenges of RiPP-based therapeutic development. mRNA display stands out for its unparalleled library diversity and selection flexibility, enabling discovery of highly stable, constrained peptides directly relevant to improving RiPP pharmacokinetic properties [86] [93]. Yeast surface display offers eukaryotic processing and quantitative screening capabilities ideal for optimizing expression and stability of engineered peptides [88] [89]. Two-hybrid systems, particularly emerging microfluidic implementations, provide powerful approaches for interrogating specific protein interactions critical to RiPP biosynthesis and function [90] [94].
The integration of these technologies with high-throughput sequencing and structural biology promises to accelerate the design-build-test-learn cycle for RiPP engineering [86]. As these methodologies become more accessible and streamlined, their application to RiPP pathway engineering will undoubtedly yield novel therapeutic candidates with enhanced clinical potential. Researchers should select screening platforms based on specific project needs, considering library diversity requirements, selection stringency, and compatibility with post-translational modifications essential for RiPP bioactivity. Through strategic implementation of these powerful screening technologies, the immense potential of RiPPs as therapeutic agents can be fully realized.
Within the field of peptide engineering, the evaluation of bioactivity is a critical step in transitioning from designed sequences to viable therapeutic candidates. For research focused on Ribosomally synthesized and post-translationally modified peptides (RiPPs), this process involves a triad of essential assessments: determining antimicrobial potency, establishing the spectrum of activity, and evaluating toxicity to eukaryotic cells [9]. RiPPs are a promising class of natural products, and their engineered analogues offer significant potential for addressing multidrug-resistant bacterial infections [58] [95]. This document outlines standardized protocols and application notes for these core evaluations, providing a framework for researchers and drug development professionals to characterize novel RiPP-based antimicrobial peptides (AMPs) reliably and consistently.
The tables below summarize key quantitative metrics and standards essential for the bioactivity evaluation of engineered peptides.
Table 1. Key Quantitative Metrics for AMP Bioactivity Evaluation
| Metric | Typical Value/Range for Promising Candidates | Significance and Interpretation |
|---|---|---|
| Minimum Inhibitory Concentration (MIC) | ⤠2 µg/mL against target pathogens [96] | Measures direct antimicrobial potency; lower values indicate greater efficacy. |
| Hemolysis (HC(_{50})) | Significantly higher than MIC (e.g., >100 µg/mL) [96] | Indicates selectivity for bacterial over mammalian cells; high HC(_{50}) is critical for low toxicity. |
| Cytotoxicity (CC(_{50})) | Significantly higher than MIC [97] | Reflects safety profile against eukaryotic cell lines; high CC(_{50}) is required for therapeutic use. |
| Therapeutic Index (TI) | As large as possible (HC({50}) or CC({50}) / MIC) | Overall indicator of selectivity and safety window; a higher TI is preferred. |
Table 2. Standardized Media for Antimicrobial Susceptibility Testing
| Medium Type | Composition | Applicable Organisms |
|---|---|---|
| Cation-Adjusted Mueller-Hinton Broth (CA-MHB) | Standardized broth for broth microdilution [98] | Non-fastidious bacteria (e.g., E. coli, P. aeruginosa, S. aureus) |
| Mueller-Hinton Agar (MHA) | Standardized agar for disk diffusion [98] | Non-fastidious bacteria |
| Mueller-Hinton Fastidious (MH-F) Broth/Agar | MHB/Agar + 5% defibrinated horse blood + 20 mg/L β-NAD [98] | Fastidious organisms (e.g., Streptococci, Campylobacter) |
This protocol determines the lowest concentration of a peptide that visually inhibits bacterial growth (MIC) and establishes its spectrum of activity against a panel of clinically relevant bacteria [98] [99].
Peptide Preparation:
Inoculum Preparation:
Inoculation and Incubation:
Result Interpretation and Spectrum Determination:
This method provides a qualitative and rapid assessment of antimicrobial activity and spectrum [99].
Agar Plate Preparation:
Inoculation and Disk Application:
Incubation and Measurement:
This protocol assesses the safety profile of AMPs by measuring their damaging effects on red blood cells (hemolysis) and mammalian cell lines (cytotoxicity) [97] [96].
Part A: Hemolysis Assay
Erythrocyte Preparation:
Peptide Incubation:
Quantification:
(Abs_sample - Abs_negative_control) / (Abs_positive_control - Abs_negative_control) * 100. The HC(_{50}) is the peptide concentration causing 50% hemolysis.Part B: Mammalian Cell Cytotoxicity Assay (e.g., MTT Assay)
Cell Seeding:
Peptide Treatment:
Viability Measurement:
(Abs_sample - Abs_blank) / (Abs_negative_control - Abs_blank) * 100. The CC(_{50}) is the peptide concentration that reduces cell viability by 50%.
Diagram 1: Bioactivity evaluation workflow for RiPPs and AMPs.
Diagram 2: Key mechanisms of antimicrobial peptide action.
Table 3. Essential Reagents and Materials for Bioactivity Evaluation
| Item | Function/Application | Example/Specification |
|---|---|---|
| Cation-Adjusted Mueller-Hinton Broth (CA-MHB) | Standardized medium for MIC determination; ensures reproducible cation concentrations critical for peptide activity [98]. | Commercially available, prepared according to EUCAST/CLSI standards. |
| Mueller-Hinton Agar (MHA) | Standardized medium for disk diffusion assays [98]. | Can be supplemented with horse blood and β-NAD (MH-F) for fastidious bacteria. |
| Sensititre Broth Microdilution Plates | Custom-designed plates for high-throughput, standardized MIC testing [99]. | 96-well plate format, pre-loaded with antibiotics or custom compounds. |
| Sterile Blank Disks (6 mm) | Used as a carrier for antimicrobial agents in diffusion assays [99]. | Typically made of cellulose or filter paper. |
| Cell Culture Media & Reagents | Maintenance and toxicity testing of eukaryotic cell lines. | DMEM/RPMI-1640, Fetal Bovine Serum (FBS), Penicillin-Streptomycin. |
| MTT Reagent | A tetrazolium salt used to quantitatively measure mammalian cell viability and proliferation. | (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium Bromide). |
| Phosphate-Buffered Saline (PBS) | Washing and resuspending red blood cells for hemolysis assays. | Isotonic, pH-buffered saline. |
| Triton X-100 | A non-ionic detergent used as a positive control (100% lysis) in hemolysis assays. | Typically used at a 1% (v/v) concentration. |
Within the field of peptide therapeutics, ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a promising class of natural products with diverse bioactivities [9] [100]. However, the successful development of peptide-based drugs, including RiPPs, is often critically compromised by poor pharmacokinetics, primarily due to rapid proteolytic degradation in the body and consequent short systemic circulation times [101] [102]. A deep understanding of the factors governing peptide stability and the implementation of robust profiling protocols are therefore essential for optimizing RiPP candidates and advancing them through the drug development pipeline. This Application Note provides a detailed framework for assessing the resistance to proteolysis and serum half-life of RiPPs, presenting standardized experimental protocols and data analysis techniques tailored for research scientists and drug development professionals.
The stability of a peptide in a biological fluid is an intrinsic property governed by its physicochemical characteristics. A meta-analysis of peptide stability data has identified several key properties with significant predictive power for proteolytic resistance [101].
It is critical to note that the stability of a therapeutic peptide is not an absolute value but is highly dependent on the in vitro system used for testing. Studies have demonstrated considerable differences in degradation rates for the same peptide when incubated in fresh blood, plasma, or serum, as these matrices have varying compositions of active proteases and inhibitors [102]. Consequently, stability data must be interpreted within the context of the assay system.
This protocol describes a standard method for determining the terminal half-life of a peptide in human or animal serum/plasma, a core parameter for pharmacokinetic profiling [101] [102].
3.1.1 Principle The peptide of interest is incubated in serum or plasma at physiological temperature (37°C). Aliquots are removed over time, and the reaction is quenched. The concentration of intact peptide remaining is quantified, allowing for the calculation of its degradation rate constant and half-life.
3.1.2 Materials and Reagents
3.1.3 Procedure
3.1.4 Data Analysis and Calculation
The workflow for this protocol is summarized in the diagram below:
This protocol is designed to investigate the differential stability of peptides in various hematological matrices, a critical consideration for translating in vitro results to in vivo performance [102].
3.2.1 Principle The same peptide is incubated in three different matricesâfresh blood (stabilized with an anticoagulant like KâEDTA), plasma, and serumâobtained from the same subject. The degradation profiles are then directly compared to evaluate the impact of the coagulation cascade and other matrix-specific factors on peptide stability.
3.2.2 Key Materials
3.2.3 Procedure
3.2.4 Data Interpretation Compare the calculated half-lives and the metabolite profiles (identified via MS) across the three matrices. Peptides containing basic residues (Lys, Arg) may be degraded significantly faster in serum due to the activation of trypsin-like serine proteases (e.g., thrombin) during the clotting process [102].
The following table summarizes key physicochemical properties and experimental half-life data, providing a template for the systematic comparison of peptide candidates. The data is curated from published studies [101] [102].
Table 1: Physicochemical Properties and Experimental Half-Life of Model Peptides
| Peptide Name | Molecular Weight (Da) | Net Charge (pH 7.0) | Isoelectric Point (pI) | Hydro- phobicity (H) | Experimental Half-Life (tâ/â) | Experimental Matrix |
|---|---|---|---|---|---|---|
| Api88 | ~2,300 | +6 | >10 | High | ~1 min | Mouse Serum [102] |
| Chex1 | ~1,800 | +4 | ~10.5 | Medium | ~10 min | Mouse Serum [102] |
| Model Peptide A | 1,542 | +2 | 9.8 | Low | 45 min | Human Serum [101] |
| Model Peptide B | 2,099 | -1 | 5.5 | High | >120 min | Human Serum [101] |
| DRP-1 | ~1,600 | +3 | ~9.5 | Medium | ~30 min | Mouse Blood [102] |
| DRP-2 | ~1,600 | +3 | ~9.5 | Medium | ~5 min | Commercial Serum [102] |
Table 2: Key Reagent Solutions for Stability and Half-Life Assays
| Research Reagent | Function & Application in Protocol |
|---|---|
| KâEDTA Anticoagulant Tubes | Chelates calcium to prevent coagulation; used for collecting plasma and for fresh blood stability studies [102]. |
| Serum Separator Tubes | Contains a gel barrier and clot activators for clean serum preparation after centrifugation [102]. |
| Trichloroacetic Acid (TCA) 10% (v/v) | A potent protein denaturant used to quench proteolytic reactions immediately after sampling [102]. |
| Protease Inhibitor Cocktail | A mixture of inhibitors targeting various protease classes; an alternative to TCA for quenching, especially if subsequent activity assays are planned. |
| Phosphate-Buffered Saline (PBS) | An isotonic, physiologically compatible buffer for dissolving and diluting peptide stocks without inducing matrix stress [102]. |
| Fibrinopeptide A | Serves as an internal control to monitor the onset and progression of coagulation in serum-based stability assays [102]. |
Robust experimental profiling of proteolytic stability and serum half-life is a cornerstone of RiPP engineering and therapeutic development. The protocols detailed herein provide a standardized approach to generate reproducible and pharmacologically relevant data. By understanding the key physicochemical drivers of stability and acknowledging the critical differences between hematological matrices, researchers can make more informed decisions to guide the rational design of RiPP-based therapeutics with optimized pharmacokinetic properties. Integrating these stability profiles with other efficacy and safety parameters will ultimately accelerate the translation of promising RiPP candidates from the laboratory to the clinic.
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a rapidly expanding superfamily of natural products, unified by a common biosynthetic logic: a ribosomally synthesized precursor peptide is transformed into a structurally complex, bioactive molecule through a series of post-translational modifications (PTMs) [8] [103]. Their genetic encodability, combined with the remarkable promiscuity of their biosynthetic enzymes, makes RiPPs exceptionally attractive for bioengineering [8] [104]. This application note provides a comparative analysis of engineered RiPPs against their natural counterparts and conventional therapeutics, framed within the context of peptide engineering research. It includes structured data, detailed protocols for key experiments, and essential resource guides to support researchers in leveraging RiPP pathways for drug discovery.
The following tables summarize key comparative data on bioactivities, therapeutic properties, and biophysical characteristics of RiPPs.
Table 1: Antimicrobial Activity of Selected Natural and Engineered RiPPs
| RiPP Compound | Class | Bioactivity | Target / Mode of Action | Potency (MIC) | Source |
|---|---|---|---|---|---|
| Microvionin [8] | Lipolanthine | Anti-Gram-positive | Bacterial cell membrane | 0.1 - 0.46 μg mLâ»Â¹ (vs. S. aureus) | Natural |
| Darobactin [8] [103] | Darobactin | Anti-Gram-negative | BamA complex, outer membrane protein folding | N/A | Natural |
| Thuricin CD [104] | Sactipeptide | Anti-Gram-positive | Two-component system; pore formation? | N/A | Natural |
| Goadvionin B2 [8] | Polyketide/RiPP hybrid | Anti-Gram-positive | N/A | 6.4 μg mLâ»Â¹ (vs. S. aureus) | Heterologous Expression |
| Engineered Lanthipeptides [103] | Lanthipeptide | Anti-Gram-positive | Cytoplasmic membrane disruption | Varies by mutation | Bioengineering |
Table 2: Comparison of Therapeutic Modalities
| Property | Natural RiPPs | Engineered RiPPs | Conventional Therapeutics |
|---|---|---|---|
| Structural Diversity [8] [103] | High, but limited to natural scaffolds | Extremely high via enzyme promiscuity & directed evolution | Low (Small Molecules); High (Biologics) |
| Target Specificity [105] [103] | High for specific microbial targets | Can be optimized for potency & specificity | Variable; small molecules often have off-target effects |
| Stability [106] [103] | Good (e.g., protease resistance from PTMs) | Can be further enhanced through engineering | Poor (Natural Peptides); Good (Small Molecules, mAbs) |
| Manufacturability [106] | Challenging (low yield, complex purification) | Improved via heterologous expression & optimized BGCs | Established & low-cost (Small Molecules); High-cost (mAbs) |
| Membrane Permeability [105] | Generally poor for intracellular targets | Can be improved with PTM engineering & sequence design | Good (Small Molecules); Poor (mAbs) |
| Half-life [105] [106] | Short (natural peptides) | Can be extended (e.g., lipid conjugation, macrocyclization) | Long (mAbs); Short (natural peptides) |
Table 3: Key RiPP Classes, Modifications, and Bioengineering Potential
| RiPP Class | Characteristic PTMs | Example Bioactivities | Engineering Approach |
|---|---|---|---|
| Lanthipeptides [103] | Lanthionine (Lan)/Methyllanthionine (MeLan) thioether bridges | Antibacterial (e.g., Nisin) | Leader peptide engineering, mutasynthesis, fusion of modification enzymes |
| Sactipeptides [104] [103] | Sulfur-to-α-carbon thioether linkages by rSAM enzymes | Antibacterial (e.g., Thuricin CD) | Exploiting enzyme complexes (e.g., TrnC/TrnD) for heterologous production |
| Lipolanthines [8] | Labionin/Solabionin cross-links & N-terminal lipidation | Antibacterial (e.g., Microvionin, Solabiomycin) | Hybrid BGC engineering combining RiPP and PKS elements |
| Lasso Peptides [103] [107] | Lariat knot-like topology | Antibacterial, Antiviral, Anticancer | Machine learning (e.g., LassoESM) to predict enzyme-substrate pairs for cyclization |
| Thiopeptides [8] [103] | Central pyridine, thiazole/oxazole rings, macrocyclization | Antibacterial, inhibits protein synthesis | Precursor peptide gene mutagenesis to alter core peptide sequence |
Objective: To identify novel RiPP Biosynthetic Gene Clusters (BGCs) and express them in a heterologous host (e.g., Streptomyces lividans) for compound production and characterization [8] [31].
Workflow Overview:
Materials:
Procedure:
Objective: To characterize the function and specificity of individual RiPP maturase enzymes using purified components [104].
Workflow Overview:
Materials:
Procedure:
trnC and trnD for thuricin CD [104]) into a protein expression vector (e.g., pET series) with an N- or C-terminal His-tag.Table 4: Essential Reagents and Tools for RiPP Engineering Research
| Item | Function / Application | Example / Specification |
|---|---|---|
| Bioinformatics Suites [31] | RiPP BGC identification from genomic/metagenomic data | antiSMASH (enzyme-centric), DeepRiPP/TrRiPP (precursor-centric) |
| Specialized AI Models [107] | Predicting lasso peptide properties and enzyme compatibility | LassoESM (a large language model tailored for lasso peptides) |
| Heterologous Host Systems [8] | Expression of cryptic or silent RiPP BGCs | Streptomyces lividans TK23, E. coli BL21(DE3) |
| Expression Vectors [8] | Cloning and transfer of large RiPP BGCs | pIJ10257 (for Streptomyces), BAC vectors (for large clusters) |
| Peptide Synthesis Service [108] | Providing high-quality precursor peptides & analogs for PTM studies | GenScript PepPower platform (custom synthesis, diverse modifications) |
| Radical SAM Enzyme Reagents [104] | In vitro activity assays for sactipeptides/ranthipeptides | S-adenosylmethionine (SAM), sodium dithionite (reducing agent) |
| Analytical Standards | Validation and quantification of RiPP structures | Commercially available lantibiotics (e.g., Nisin), custom-synthesized RiPPs |
Ribosomally synthesized and post-translationally modified peptides (RiPPs) represent a rapidly expanding superfamily of natural products unified by a common biosynthetic logic: a genetically encoded precursor peptide is transformed into a structurally complex bioactive molecule through the action of maturase enzymes [39]. This biosynthetic pathway offers significant advantages for drug discovery and engineering. RiPPs are encoded by relatively compact biosynthetic gene clusters (BGCs), and their maturase enzymes, though diverse and often promiscuous, provide tremendous biotechnological potential for generating novel structural variants [109] [39]. The RiPP landscape has been extensively explored in bacteria, leading to several clinical agents, whereas the investigation of fungal RiPPs is a more recent development [109].
Therapeutically, RiPPs have garnered immense interest for their exceptional capabilities in addressing key challenges in modern medicine, including the modulation of "undruggable" protein-protein interactions and combating antibiotic resistance [109]. Their inherent structural diversity and target specificity make them ideal candidates for next-generation therapeutics. Among the most clinically advanced RiPP classes are the lanthipeptides, which contain (methyl)lanthionine rings that confer remarkable stability under a broad range of biological and physical conditions [110]. This review details the clinical pipeline of RiPP-derived therapeutics, from established candidates like nisin and duramycin to investigational agents such as LFF571 and NVB302, and provides detailed experimental frameworks for their research and development.
The progression of RiPPs from natural products to clinical agents showcases the potential of this diverse family. The table below summarizes key RiPP-based therapeutics and their development status.
Table 1: Clinical Pipeline of RiPP-Based Therapeutics
| Agent | RiPP Class | Therapeutic Indication | Mechanism of Action | Development Status |
|---|---|---|---|---|
| Duramycin [109] | Lanthipeptide | Cystic Fibrosis | Increases mucus hydration | Approved and in clinical use |
| Nisin [111] [110] | Lanthipeptide | Food preservative; Antimicrobial resistance research | Binds to lipid II, inhibiting cell wall synthesis and forming pores | Preclinical research & tool compound |
| Fidaxomicin [112] [113] | Macrolide (Non-RiPP) | Clostridium difficile Infection (CDI) | Inhibits RNA polymerase | Approved in 2011 |
| LFF571 [114] [113] [115] | Thiopeptide | Clostridium difficile Infection (CDI) | Inhibits elongation factor Tu (EF-Tu) | Investigational Agent (Phase II completed) |
| NVB302 [110] | Lanthipeptide | Clostridium difficile Infection (CDI) | Not fully detailed in search results; presumed similar to other lanthipeptides | Undergone clinical trials |
The search results highlight LFF571 as a prominent investigational RiPP-derived agent. It is a semi-synthetic derivative of the natural product GE2270 A, optimized to improve aqueous solubility and antibacterial activity [114] [115]. Its mechanism of action involves inhibition of the clinically unprecedented target elongation factor Tu (EF-Tu), which is essential for bacterial protein synthesis [113]. A proof-of-concept Phase II clinical trial demonstrated that LFF571 was safe and effective for treating patients with mild to moderate C. difficile infection [113].
Application Note: This protocol outlines a bioinformatic workflow for identifying novel RiPP BGCs in fungal genomes, as applied to lichen-forming fungi [109]. It is essential for expanding the known diversity of RiPPs and discovering new structural scaffolds.
Materials and Reagents:
Methodology:
Application Note: This protocol describes the generation of novel macrocyclic lanthipeptides from disulfide-bond-containing antimicrobial peptide (AMP) templates, enhancing their stability and enabling further chemical modification [110].
Materials and Reagents:
Methodology:
Diagram: Workflow for Engineering Hybrid Lanthipeptides
Application Note: This standard protocol is used to determine the minimum inhibitory concentration (MIC) of therapeutic candidates like LFF571 against Clostridium difficile and to assess the potential for resistance development [113].
Materials and Reagents:
Methodology:
Table 2: Key Reagents for RiPP Research and Development
| Reagent / Tool | Function / Application | Example Use Case |
|---|---|---|
| antiSMASH Software [109] | Predicts biosynthetic gene clusters (BGCs) from genomic data. | Initial identification of putative RiPP BGCs in lichenized fungi [109]. |
| BiG-SCAPE & MIBiG [109] | Compares and classifies BGCs into families (GCFs); dereplication against known clusters. | Grouping 987 lichen RiPP BGCs into families and identifying novel clusters [109]. |
| CinM Synthetase [110] | Class-II lanthipeptide synthetase; installs (methyl)lanthionine rings. | Engineering stable lanthipeptide analogues from disulfide-bonded AMPs [110]. |
| NisP Protease [110] | Cleaves leader peptides from modified precursor peptides. | Releasing the core lanthipeptide after maturation by CinM [110]. |
| MALDI-TOF MS [110] | High-sensitivity mass spectrometry for peptide analysis. | Verifying successful dehydration and cyclization of engineered lanthipeptides [110]. |
The clinical pipeline from nisin and duramycin to LFF571 and NVB302 underscores the tremendous potential of RiPP biosynthetic pathways as a platform for peptide engineering. The integration of advanced genome mining techniques with robust experimental protocols for engineering and evaluation is accelerating the discovery and development of this promising class of therapeutics. As our understanding of RiPP biosynthesis deepens, particularly with the ongoing exploration of understudied producers like lichen-forming fungi and the discovery of hybrid RiPP-derived molecules like lipopeptides, the pipeline is poised for further expansion, offering new hope for addressing pressing challenges in infectious diseases and beyond.
RiPP biosynthetic pathways represent a powerful and genetically encodable platform for peptide engineering, combining the diversity of natural product biosynthesis with the precision of synthetic biology. The integration of foundational knowledge with advanced genome mining, enzyme engineering, and optimization strategies has dramatically expanded our ability to create novel RiPPs with tailored properties. Future directions will focus on leveraging structural biology and computational predictions to overcome remaining specificity challenges, further expanding the chemical space of accessible modifications, and accelerating the clinical translation of engineered RiPPs to address urgent needs in antimicrobial therapy, oncology, and other therapeutic areas. The continued unraveling of Nature's synthetic potential through RiPP research promises a new era of programmable peptide therapeutics.