Tuesday, June 24, 2025

The Reign of 10x Genomics: Unveiling the Top Products in Single-Cell RNA Sequencing for Plant and Animal Research


The single-cell RNA sequencing (scRNA-seq) market is dominated by a few key players, with 10x Genomics' Chromium platform standing out as the undisputed leader in animal research. While the plant science field is more fragmented, many of the same technologies are adapted for botanical studies, supplemented by specialized kits and services. Here’s a breakdown of the top 10 selling product categories and the companies behind them that are driving discoveries at the cellular level.

The scRNA-seq landscape in animal research is characterized by a high-throughput, droplet-based approach, with a clear market leader. For plant research, the journey to single-cell analysis is often more arduous, requiring robust protoplasting techniques before leveraging similar sequencing technologies.

Powering Animal Research: A Market Led by Throughput and Precision

In the well-established animal scRNA-seq market, the top-selling products are often integrated systems encompassing instruments, consumables, and software.

1. 10x Genomics Chromium Controllers and Kits: The undisputed champion, the Chromium Controller and the newer, higher-throughput Chromium X Series, are fixtures in many research labs. Their accompanying Chromium Next GEM Single Cell 3' and 5' reagent kits are the top-selling consumables, enabling the transcriptional profiling of tens of thousands to millions of cells. The platform's ease of use, robust workflow, and extensive community support have solidified its market dominance.

2. Illumina Sequencing Systems: While not a direct scRNA-seq kit provider, Illumina's sequencing platforms, particularly the NovaSeq and NextSeq series, are the downstream workhorses for the vast majority of scRNA-seq experiments. Their deep sequencing capabilities are essential for analyzing the libraries generated by platforms like the 10x Chromium, making them an indispensable part of the ecosystem.

3. Bio-Rad SureCell and ddSEQ Systems: Bio-Rad offers a competitive droplet-based system with its ddSEQ Single-Cell Isolator and the SureCell WTA 3' Library Prep Kit. These products are recognized for their reliability and are a significant player in the market, appealing to researchers looking for alternatives to the dominant platform.

4. Parse Biosciences Evercode Whole Transcriptome Kits: A rapidly emerging player, Parse Biosciences has gained significant traction with its Evercode Whole Transcriptome kits. Their combinatorial barcoding technology offers a high-throughput, instrument-free approach that is particularly attractive for large-scale studies and screening applications, challenging the status quo with its scalability and cost-effectiveness.

5. Singleron Biotechnologies GEXSCOPE and FocuSCOPE Kits: Singleron is another key innovator, particularly in the Asian market, with its GEXSCOPE Single Cell RNA Library Kits and the versatile FocuSCOPE platform. Their focus on providing complete solutions, from instrumentation to data analysis, has positioned them as a strong competitor.


The Emerging Landscape of Plant Single-Cell RNA Sequencing

The application of scRNA-seq in plant biology is a burgeoning field. The primary challenge lies in the initial step of isolating viable single cells (protoplasts) from tissues with rigid cell walls. Consequently, the "top-selling" products are a mix of adapted animal-focused platforms and specialized plant-centric solutions.

6. Adapted 10x Genomics and Bio-Rad Workflows: Many plant researchers adapt the leading droplet-based systems from 10x Genomics and Bio-Rad for their studies. This often involves customized protoplast isolation protocols developed in-house or through collaborations, followed by the use of standard single-cell library preparation kits.

7. QIAGEN Plant-Specific RNA Extraction and Library Prep Kits: QIAGEN offers a range of products that are crucial for the upstream and library preparation stages of plant scRNA-seq. Their RNeasy Plant Mini Kits for RNA extraction and various QIAseq library preparation kits are widely used and trusted for their performance with challenging plant samples.

8. Plate-Based Sorting and Library Preparation (e.g., Takara Bio): For studies focusing on specific or rare cell types, plate-based methods remain popular. This often involves fluorescence-activated cell sorting (FACS) to isolate individual protoplasts into 96- or 384-well plates, followed by library preparation using kits like the Takara Bio SMART-Seq series, which are known for their high sensitivity with low-input RNA.

9. Lifeasible Custom Plant scRNA-seq Services: Recognizing the technical hurdles in plant scRNA-seq, companies like Lifeasible have carved a niche by offering comprehensive services. They provide expertise in protoplast isolation from various plant species and tissues, followed by sequencing and bioinformatic analysis, making this powerful technology accessible to a broader range of plant scientists.

10. Thermo Fisher Scientific RNA Extraction and Quantification Reagents: As a major supplier of molecular biology reagents, Thermo Fisher Scientific's products are integral to many scRNA-seq workflows. Their TRIzol reagent for RNA extraction and Qubit fluorometers for nucleic acid quantification are staples in labs conducting both plant and animal single-cell studies.

In conclusion, while the animal scRNA-seq market is clearly led by high-throughput, droplet-based platforms from companies like 10x Genomics, the plant research domain is more varied, relying on a combination of adapted technologies and specialized service providers to overcome its unique challenges. The trend across both fields is a move towards higher cell numbers, multi-omic analyses, and the integration of spatial context, promising even deeper insights into the intricate workings of life at its most fundamental level.


Sunday, June 22, 2025

Plants' Secret Internet: How Tiny RNAs Are Hacking Nature for Bigger, Better Crops!

 

Long-distance transport of RNA Molecules in Plants

Long-Distance RNA Transport in Plants: Analysis Methods and Agricultural Applications


Plants, unlike mobile organisms, rely on intricate internal communication to manage their growth, development, and responses to environmental changes. A key part of this communication is the long-distance transport of various RNA molecules, which challenges the traditional idea that gene expression is confined to a single cell. This report explores the basic ways RNA moves within plants, the advanced tools used to study this movement, and its significant potential for improving agriculture.

The phloem and plasmodesmata, parts of the plant's vascular system, are the main routes for moving large molecules, including diverse RNA types like messenger RNAs (mRNAs), small RNAs (sRNAs such as miRNAs and siRNAs), and long non-coding RNAs (lncRNAs). This isn't just passive movement; it's a highly regulated and selective process. Specific RNA motifs, structural features, and modifications after transcription, all managed by RNA-binding proteins (RBPs), guide this transport.

Thanks to breakthroughs in analytical methods—from sophisticated grafting experiments combined with high-throughput sequencing to real-time imaging and proteomics—we're constantly learning more about RNA trafficking. While these methods have revealed the crucial roles of mobile RNAs in processes like flowering, leaf development, and stress responses, they've also highlighted challenges, especially in telling apart genuine mobile signals from experimental noise.

The agricultural implications are huge, mainly through RNA interference (RNAi) technologies. RNAi offers a precise and environmentally friendly way to improve crops, leading to better disease and pest resistance, increased yields, and desirable quality traits. Looking ahead, we need to overcome current research limitations, fully understand the complex rules governing RNA transport, and use synthetic biology and nanotechnology to create new mobile RNAs for specific trait modifications and sustainable farming practices.


1. Introduction: The Importance of Long-Distance RNA Signaling in Plants

Plants, unlike animals, can't move to escape unfavorable conditions. Instead, they must adapt by orchestrating complex biological processes across their spatially separated organs. This requires sophisticated internal communication networks, which involve a diverse array of signaling molecules. Beyond well-known signals like hormones and peptides, large molecules, including proteins and various forms of RNA, are crucial for this systemic communication.

The idea of RNA as a mobile signaling molecule has fundamentally changed how we view gene expression, moving it beyond a strictly local event. A growing body of evidence shows that specific RNA molecules can travel long distances within a plant, acting as non-cell autonomous carriers of information. This mobility allows plants to integrate various environmental cues—such as light, nutrient availability, or pathogen attack—and coordinate physiological responses throughout the entire organism. For example, if roots detect a nutrient shortage, signals can be sent to the shoots. Similarly, stress signals from leaves can be relayed to other parts of the plant, enabling a unified adaptive strategy. This capacity for rapid, coordinated, and precise adjustments highlights RNA's role as a central integrator of plant plasticity, allowing plants remarkable flexibility and resilience in changing conditions. Understanding these intricate RNA-mediated communication networks is vital for developing strategies to manipulate plant responses for agricultural benefit.


2. Mechanisms and Pathways of Long-Distance RNA Transport

The long-distance transport of RNA molecules in plants is a complex, multi-step process, mainly facilitated by the plant's vascular system. This system efficiently relays genetic information and regulatory signals between distant tissues.

The Vascular System: Phloem and Plasmodesmata as Conduits

The primary pathway for long-distance RNA transport is the phloem, a specialized vascular tissue that distributes sugars, amino acids, hormones, and macromolecules from "source" tissues (like mature leaves) to "sink" tissues (such as roots, developing fruits, and young leaves). This extensive network enables system-wide delivery of various signals. The phloem's role in integrating a wide range of signaling pathways to regulate plant development and stress responses has led to its description as a "plant internet." This analogy emphasizes the complexity and far-reaching nature of phloem-mediated communication, where RNA "information" is sent and received by distant cellular "nodes," enabling coordinated responses to local stimuli across the entire plant. This sophisticated system suggests the phloem plays a more active and intelligent role than just passive delivery.

RNA molecules move into and out of the phloem sieve elements (SEs) from neighboring companion cells (CCs)—where many mobile RNAs are synthesized—via plasmodesmata (PD). Plasmodesmata are nanochannels embedded within the plant cell wall, forming a continuous cytoplasmic and membrane system (symplasm) that directly connects adjacent cells. These channels allow small molecules to diffuse passively, but more importantly, they facilitate the selective transport of larger macromolecules, including RNA and proteins.

Cellular Factors Facilitating Selective Transport

The transport of macromolecules, particularly endogenous RNAs, isn't a simple passive process; it's actively regulated and highly selective. While small non-native proteins might diffuse without selection, specific endogenous RNAs and proteins are actively chosen for transport. This active selection is critical for maintaining cellular order and energy efficiency. If transport were purely passive, all RNAs would move indiscriminately, leading to cellular chaos and significant energy waste. Instead, this regulated process ensures that only specific, necessary information is transported to precise destinations at the right times, providing fine-tuned spatial and temporal control over gene expression in distant cells. The regulation of cell-to-cell transport through plasmodesmata can involve mechanisms like callose deposition, which dynamically controls the size-exclusion limit of these pores, thereby modulating what can pass through. Identifying the intricate mechanisms of this "smart" transport system is fundamental for engineering targeted RNA delivery in agriculture.

Key RNA Motifs and Modifications Governing Mobility

The selectivity of RNA transport largely comes from specific sequence and structural motifs within the RNA molecules themselves, acting as molecular "zip codes" or recognition elements for the transport machinery. These include:

  • Polypyrimidine (poly-CU) sequences: Often found in the 3' untranslated region (UTR) of mobile mRNAs, these motifs can bind to specific RNA-binding proteins (RBPs) to form ribonucleoprotein (RNP) complexes. This interaction is thought to aid RNA mobility and stability during transport.
  • Transfer RNA (tRNA)-related sequences: These sequences, including tRNA-like structures (TLS), are notably rich in the 3' UTRs of mobile mRNAs. Some studies suggest these sequences are necessary and sufficient for long-distance RNA transport.
  • Single Nucleotide Mutations: Changes at specific single nucleotide positions can affect RNA mobility, possibly by impacting RNP complex formation or overall RNA structure.
  • Untranslated Regions (UTRs): Both the 3' and 5' UTRs are crucial for regulating mRNA expression, stability, localization, and, importantly, their mobility within the phloem.
  • Stem-loop structures: These distinct secondary structures, common in precursor microRNAs (pre-miRNAs), also contribute to RNA mobility.

Beyond these motifs, post-transcriptional RNA modifications are another critical layer of regulatory control for transport. Recent research emphasizes the importance of methylated 5′ cytosine (m5C) for RNA transport and function. This modification can increase mRNA stability, which is beneficial for long-distance transport, and may interact with specific methyltransferases for selective transport. The observation that high m5C content in Arabidopsis mRNA is negatively correlated with mRNA translation activity suggests a mechanism to prevent premature protein synthesis during transport, ensuring the RNA arrives intact at its destination before being translated. This discovery expands the "RNA zip code" hypothesis, indicating that the epitranscriptome—the landscape of RNA modifications—is as vital as the sequence itself in determining an RNA's fate, including its long-distance mobility. This opens new avenues for engineering RNA mobility by targeting these specific modifications.

The Crucial Role of RNA-Binding Proteins (RBPs) in Transport and Protection

RNA-binding proteins (RBPs) are essential for various aspects of RNA biology, including RNA metabolism, transport, and a plant's ability to adapt to diverse environmental conditions. A critical function of RBPs in long-distance RNA transport is their role in protecting RNA molecules from degradation, a necessary safeguard as RNAs travel through the plant's vascular system.

RBPs form dynamic ribonucleoprotein (RNP) complexes with mobile RNAs. These complexes are the functional units of transport, guiding RNA molecules to specific subcellular locations and mediating their delivery through plasmodesmata and into the phloem. The interaction between RBPs and RNA is not just for protection; it's fundamental to the selectivity and directionality of transport. The RBP effectively "licenses" the RNA for long-distance travel, ensuring it reaches the correct destination and potentially influencing its translation or stability upon arrival.

Several phloem-mobile RBPs have been identified and characterized, including:

  • CmPP16 (16-kD Cucurbita maxima phloem protein): Its cross-reactivity with viral movement proteins suggests a shared mechanism for systemic transport.
  • Phloem Lectins (CsPP2 from cucumber and CmmLec17 from melon): These abundant proteins in phloem sap can interact with viroid RNAs and a broad spectrum of mRNAs, facilitating their movement.
  • CmPSRP1 (Cucurbita maxima Phloem Small RNA Binding Protein1): Preferentially binds to small single-stranded RNAs, potentially involved in si/miRNA transport.
  • Pumpkin Eukaryotic Translation Initiation Factor 5A (eIF5A): Binds mRNA, particularly the 3′UTR of mobile StBEL5 mRNA, suggesting its role in RNP complexes that regulate RNA transport or metabolism.
  • RBP50: A polypyrimidine tract-binding (PTB) protein, forming the core of RNP complexes that transport specific sets of mRNAs, including those encoding transcription factors.
  • AtRRP44a: In Arabidopsis thaliana, this protein acts as an "escort protein" essential for the normal movement of RNA messages between cells, and its absence leads to improper plant development.

The pervasive involvement of RBPs emphasizes that the RNP complex, rather than naked RNA, is the functional unit of long-distance transport. Engineering mobile RNAs for agricultural applications will thus likely require a comprehensive understanding of, and potentially co-engineering with, the endogenous RBP machinery to ensure efficient and targeted delivery.


3. Analytical Methods for Studying Mobile RNAs in Plants

Investigating the complex dynamics of long-distance RNA transport in plants requires a diverse array of advanced analytical methods. These techniques enable the identification, tracking, quantification, and functional characterization of mobile RNA molecules.

Grafting Experiments: A Cornerstone for Identifying Mobile RNAs

Grafting experiments remain a foundational technique for studying long-distance transport. This method involves physically joining two different plant genotypes—a scion (shoot) and a rootstock—and then tracking the movement of RNA molecules across the graft junction. This approach provides direct in vivo evidence of RNA mobility.

For grafts within the same species or closely related ecotypes, Single Nucleotide Polymorphisms (SNPs) serve as genetic markers to distinguish between RNAs from the scion versus the rootstock. By sequencing RNA from tissues on both sides of the graft, researchers can identify transcripts that have moved from their original genotype into the grafted partner. In cases of heterografts (grafts between different species), the greater sequence divergence simplifies tracking, as RNA sequencing reads can be unambiguously mapped to the respective genomes of the donor and recipient species. Natural grafts, such as those formed between parasitic dodder plants and their hosts, also serve as valuable models for studying cross-species RNA transfer due to their distinct genetic backgrounds.

While grafting is an indispensable tool for demonstrating long-distance RNA transport, recent critical re-evaluations of RNA sequencing datasets from grafted plants have highlighted methodological nuances. Meta-analyses suggest that a significant portion of previously identified mobile mRNAs might be artifacts resulting from technical noise, genome mis-mapping, or contamination. This calls for a more cautious and stringent approach to data interpretation. The scientific community now emphasizes the need for rigorous experimental design, advanced bioinformatics tools, and integrative methodologies to distinguish true mobile signals from background noise, ensuring that conclusions about RNA mobility are robustly validated beyond mere detection.

Molecular Profiling: RNA Sequencing of Phloem Sap and Single-Cell Transcriptomics

Molecular profiling techniques are crucial for identifying and characterizing the diverse populations of RNA molecules involved in long-distance signaling. cDNA library and omics profiling have been instrumental in identifying a wide range of RNA signals across various plant species.

A more direct approach involves RNA Sequencing (RNA-seq) of phloem exudates. This technique directly identifies the RNA populations within the phloem sap, including mRNAs, small RNAs (siRNAs, miRNAs), and even tRNA-derived fragments (tRFs). Specialized methods, such as Ethylenediaminetetraacetic Acid (EDTA) collection, are used to minimize cellular damage and obtain relatively pure phloem contents, despite the presence of substances like P protein that can complicate RNA extraction.

However, traditional short-read RNA-seq has limitations, especially with complex transcripts, alternative splicing isoforms, and fusion genes, often leading to splicing errors and hindering comprehensive analysis of transcript structure and function. To overcome these challenges, Long-read RNA Sequencing (DRS), particularly Nanopore-based Direct RNA Sequencing, has emerged as a powerful tool. DRS captures full-length transcripts, allowing the identification of novel lncRNAs, analysis of poly(A) tail length changes (which affect RNA stability and translation efficiency), and direct detection of various RNA modifications like m6A and m5C. This technological progression provides a more accurate and comprehensive understanding of the mobile RNA landscape, moving beyond mere presence to detailed structural and modification-dependent functions.

Furthermore, single-cell transcriptomics (scRNAseq) is an emerging technique that allows for the study of RNA transport at a cell-type specific level. This provides unprecedented resolution for establishing cell-type specific RNA transport patterns and identifying associated motifs. The evolution of these omics technologies continuously deepens our understanding of RNA mobility, which is critical for targeted manipulation in agricultural contexts.

Advanced Imaging Techniques

While molecular profiling identifies the presence and types of mobile RNAs, advanced imaging techniques are essential for visualizing their dynamic movements and localizations in vivo. Fluorescence Microscopy is widely used, often employing systems where RNA molecules are tagged with specific stem-loop motifs that bind to fluorescently-labeled bacterial proteins (e.g., BglG, MS2, λN). The BglG system, for instance, has effectively tracked mRNA granules and their intercellular transport through plasmodesmata.

A significant advancement in this area is the RNA-Triggered Fluorescence (RTF) reporter system. This engineered platform enables dynamic, real-time tracking of RNA expression at both cellular and whole-plant scales, using programmable RNA switches for precise control. This allows researchers to observe the trajectories, speeds, and interactions of RNA molecules as they move through living plant tissues.

Another promising technology is the application of Aggregation-Induced Emission Luminogens (AIEgens) for plant RNA bioimaging. AIEgens show high fluorescence intensity, good photostability, and low cellular toxicity. Their unique property of increasing fluorescence upon aggregation helps overcome the common issue of aggregation-caused quenching seen with conventional fluorophores. This technology holds promise for more effective RNA visualization in plants, especially given interference from plants' naturally fluorescent substances.

Fluorescent In Situ Hybridization (FISH) is a histological technique that uses nucleic-acid based probes to localize specific RNA sequences within cells or tissues. FISH provides valuable spatial and temporal information regarding gene expression in situ at single-cell resolution, allowing direct visualization and quantification of individual RNA molecules. This technique can visualize mRNA, small RNAs (siRNA, ASOs), microRNAs, and lncRNAs, and has been successfully used in various crop species.

The development of these real-time visualization methods is critical for understanding the dynamic processes of RNA movement. They go beyond static snapshots from sequencing data to provide direct evidence of RNA trajectories and interactions in vivo, which is essential for understanding transport mechanisms rather than just the presence of mobile RNAs. Continued development and integration of these imaging tools will be crucial for unraveling the intricacies of RNA trafficking and validating findings from omics approaches.

Proteomics Approaches for Identifying RNA-Binding Proteins

Given the crucial role of RNA-binding proteins (RBPs) in RNA transport and protection, proteomics approaches are vital for identifying the protein components of the phloem sap and the ribonucleoprotein (RNP) complexes that facilitate RNA mobility. Shotgun proteomics, for example, has been used to extract a "core proteome" of proteins ubiquitously present in various plant tissues, including phloem sap.

Studies have revealed that this core proteome includes numerous RBPs and other proteins involved in long-distance signaling and stress responses. The presence of a significant "core stress responsive proteome" (CSRP) in the phloem suggests that the phloem functions not merely as a transport conduit but as an active signaling hub where proteins and RNAs interact to coordinate systemic stress responses across the entire plant. This highlights the importance of the dynamic interplay between RNA and protein in mediating plant communication and adaptation. Future research should focus on the dynamic interactions within these RNP complexes, how their composition changes under different environmental conditions, and how these changes influence RNA mobility and ultimate function.


 Author: KuriousK. | Subscribe: Don’t miss updates—follow this blog!

 

Relevant Bibliography: 

1. RNA Motifs and Modification Involve in RNA Long-Distance Transport in Plants - Frontiers, https://www.frontiersin.org/journals/cell-and-developmental-biology/articles/10.3389/fcell.2021.651278/full

2.    Long-Distance Signaling via Mobile RNAs - ResearchGate, https://www.researchgate.net/publication/290254417_Long-Distance_Signaling_via_Mobile_RNAs

3.    Phloem RNA-binding proteins as potential components of the long-distance RNA transport system - Frontiers, https://www.frontiersin.org/journals/plant-science/articles/10.3389/fpls.2013.00130/full

4.    Phloem RNA-binding proteins as potential components of the long-distance RNA transport system - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC3650515/

5.    Relevance of Translational Regulation on Plant Growth and Environmental Responses | Frontiers Research Topic, https://www.frontiersin.org/research-topics/4647/relevance-of-translational-regulation-on-plant-growth-and-environmental-responses/magazine

6.    RNA Motifs and Modification Involve in RNA Long-Distance Transport in Plants - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC8047152/

8.    Plants: RNA notes to self | Cold Spring Harbor Laboratory, https://www.cshl.edu/plants-rna-notes-to-self/

9.    Phloem-mobile messenger RNAs and root development - Frontiers, https://www.frontiersin.org/journals/plant-science/articles/10.3389/fpls.2013.00257/full

10.  Phloem-mobile messenger RNAs and root development - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC3713340/

11.  PLAMORF: Long-distance RNA signalling in plants, https://plamorf.eu/

12.  RNA trafficking in parasitic plant systems - Frontiers, https://www.frontiersin.org/journals/plant-science/articles/10.3389/fpls.2012.00203/full

13.  Long-Distance Movement of Solanum tuberosum Translationally Controlled Tumor Protein (StTCTP) mRNA - MDPI, https://www.mdpi.com/2223-7747/12/15/2839

14.  plantae.org, https://plantae.org/mechanisms-of-long-distance-mrna-movement/#:~:text=The%20long%2Ddistance%20trafficking%20of,translocation%20through%20phloem%2C%20and%20unloading

15.  Innovations, Challenges and Future Directions of T7RNA Polymerase in Microbial Cell Factories | ACS Synthetic Biology, https://pubs.acs.org/doi/10.1021/acssynbio.5c00139

16.  The Mobile Small RNAs: Important Messengers for Long-Distance Communication in Plants, https://www.frontiersin.org/journals/plant-science/articles/10.3389/fpls.2022.928729/full

17.  Long-read RNA Sequencing: A Powerful Tool for Analyzing Plant and Animal Transcriptomes - CD Genomics, https://www.cd-genomics.com/longseq/long-read-rna-sequencing-a-powerful-tool-for-analyzing-plant-and-animal-transcriptomes.html

18.  RNA methylation in plants: An overview - Frontiers, https://www.frontiersin.org/journals/plant-science/articles/10.3389/fpls.2023.1132959/full

19.  Mechanisms Underlying Graft Union Formation and Rootstock Scion Interaction in Horticultural Plants - Frontiers, https://www.frontiersin.org/journals/plant-science/articles/10.3389/fpls.2020.590847/full

20.  Transfer of endogenous small RNAs between branches of scions and rootstocks in grafted sweet cherry trees - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC7386610/

21.  Re-analyzing Mobile mRNA: Limits of Long-Distance Communication - Bioengineer.org, https://bioengineer.org/re-analyzing-mobile-mrna-limits-of-long-distance-communication/

22.  The Small RNA Component of Arabidopsis thaliana Phloem Sap and Its Response to Iron Deficiency - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC10421156/

23.  Aggregation-Induced Emission Luminogens: A New Possibility for Efficient Visualization of RNA in Plants - MDPI, https://www.mdpi.com/2223-7747/13/5/743

24.  How to visualize mRNA in vivo - IBMP - CNRS, https://www.ibmp.cnrs.fr/how-to-visualize-mrna-in-vivo/?lang=en

25.  RNA-triggered fluorescence controlled by RNA switches for real-time RNA expression tracking in living plants | bioRxiv, https://www.biorxiv.org/content/10.1101/2025.03.03.641157v1.full-text

26.  Plant RNA Fluorescent In Situ Hybridization (FISH) Service - Creative Biogene, https://www.creative-biogene.com/services/plant-rna-fluorescent-in-situ-hybridization-fish-service.html

27.  RNA FISH in Plant | Creative Bioarray, https://www.creative-bioarray.com/services/rna-fish-in-plant.htm

28.  Phloem Sap Proteins Are Part of a Core Stress Responsive Proteome Involved in Drought Stress Adjustment - Frontiers, https://www.frontiersin.org/journals/plant-science/articles/10.3389/fpls.2021.625224/full

29.  Different types of RNAs and their functions - FutureLearn, https://www.futurelearn.com/info/courses/translational-research/0/steps/14201

30.  Unlocking Plant Genetics with mRNA - Number Analytics, https://www.numberanalytics.com/blog/ultimate-guide-messenger-rna-plant-genetics

31.  Regulatory Small RNAs for a Sustained Eco-Agriculture - PMC - PubMed Central, https://pmc.ncbi.nlm.nih.gov/articles/PMC9863784/

32.  The plant noncoding transcriptome: a versatile environmental sensor | The EMBO Journal, https://www.embopress.org/doi/10.15252/embj.2023114400

33.  Long Noncoding RNAs in Plants - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC6689229/

34.  Plant long non-coding RNAs: identification and analysis to unveil their physiological functions - Frontiers, https://www.frontiersin.org/journals/plant-science/articles/10.3389/fpls.2023.1275399/full

35.  Expression Profiles and Characteristics of Apple lncRNAs in Roots, Phloem, Leaves, Flowers, and Fruit - MDPI, https://www.mdpi.com/1422-0067/23/11/5931

36.  Plant long non-coding RNAs: identification and analysis to unveil their physiological functions, https://pmc.ncbi.nlm.nih.gov/articles/PMC10644886/

37.  Phloem-mobile signals affecting flowers: Applications for crop breeding - ResearchGate, https://www.researchgate.net/publication/235440758_Phloem-mobile_signals_affecting_flowers_Applications_for_crop_breeding

38.  Texas A&M AgriLife researcher discusses RNAi use in crops ..., https://agrilifetoday.tamu.edu/2024/03/06/texas-am-agrilife-researcher-helps-outline-rnai-alternative-to-knock-out-technology-in-thought-piece/

39.  Application of Exogenous dsRNAs-induced RNAi in Agriculture: Challenges and Triumphs, https://www.frontiersin.org/journals/plant-science/articles/10.3389/fpls.2020.00946/full

40.  RNA Interference in Agriculture: Methods, Applications, and ..., https://www.researchgate.net/publication/377716998_RNA_Interference_in_Agriculture_Methods_Applications_and_Governance

41.  RNA Interference and CRISPR/Cas Gene Editing for Crop Improvement: Paradigm Shift towards Sustainable Agriculture - MDPI, https://www.mdpi.com/2223-7747/10/9/1914

42.  RNAs - A New Frontier in Crop Protection - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC8957476/

43. Cross-Kingdom RNA Transport Based on Extracellular Vesicles Provides Innovative Tools for Plant Protection - ResearchGate, https://www.researchgate.net/publication/384491571_Cross-Kingdom_RNA_Transport_Based_on_Extracellular_Vesicles_Provides_Innovative_Tools_for_Plant_Protection

44.  De novo RNA base editing in plant organelles with engineered synthetic P-type PPR editing factors | Nucleic Acids Research | Oxford Academic, https://academic.oup.com/nar/article/53/7/gkaf279/8109927

45.  Identification of phloem-associated translatome alterations during leaf development in Prunus domestica L - PMC, https://pmc.ncbi.nlm.nih.gov/articles/PMC6355854/

46.  RNA velocity—current challenges and future perspectives | Molecular Systems Biology, https://www.embopress.org/doi/10.15252/msb.202110282

Friday, June 20, 2025

A Glimmer of Hope: Personalized RNA Vaccines Teach the Immune System to Fight Pancreatic Cancer

Source: Rojas et al.,  Nature, 2023

 

 Author: KuriousK. | Subscribe: Don’t miss updates—follow this blog! 

For decades, a diagnosis of pancreatic cancer has been one of the most feared in medicine. With a grim survival rate that has barely budged in over 60 years, it remains one of the deadliest cancers. Standard treatments like surgery and chemotherapy can help, but for the vast majority of patients, the cancer relentlessly returns.

But what if we could teach our own bodies to hunt and destroy this formidable enemy?

A groundbreaking study published in Nature has offered a powerful glimpse into this very possibility. Researchers from Memorial Sloan Kettering Cancer Center, in collaboration with BioNTech and Genentech, have demonstrated that a personalized mRNA vaccine can awaken a patient's immune system, sending a powerful army of T-cells to attack pancreatic cancer cells and significantly delay the disease's return.

The Challenge: A Cancer That Hides in Plain Sight

Our immune system’s T-cells are expert soldiers, constantly patrolling our bodies for foreign invaders like viruses and bacteria. They can also recognize and eliminate cancer cells, but notoriously "cold" tumors like pancreatic cancer are masters of disguise. They build fortress-like environments and have very few unique markers, or "neoantigens," on their surface, allowing them to hide from the immune system.

The researchers behind this study decided to turn this weakness into a weapon. They hypothesized that even a few neoantigens—which are unique to each patient's tumor—could be enough to act as a "most wanted poster" for the immune system.

The Breakthrough: A Custom-Made Weapon for Every Patient

In a revolutionary Phase I clinical trial, scientists developed a truly personalized treatment protocol. Here’s how it worked:

    1. Surgery: First, a patient's pancreatic tumor was surgically removed.
    2. Genetic Analysis: The tumor was immediately sent to a lab where scientists sequenced its DNA to identify its unique mutational fingerprint—the neoantigens.
    3. Custom Vaccine Creation: Using this genetic blueprint, a personalized mRNA vaccine (named autogene cevumeran) was created for each patient. This vaccine contained instructions to teach the immune system to recognize up to 20 of that specific patient's neoantigens.
    4. A Three-Pronged Attack: Patients first received a dose of immunotherapy (atezolizumab) to "take the brakes off" their immune system. Then, they received their personalized vaccine to direct the T-cells to their target. Finally, they underwent a standard course of chemotherapy.

The Stunning Results

The results were remarkable. The complex, time-sensitive process of creating and delivering a personalized vaccine was successful and safe. But more importantly, it was effective.

In 8 out of 16 patients, the vaccine triggered a massive and powerful T-cell response. These newly activated T-cells specifically targeted the neoantigens from the patient's own cancer.

The clinical impact was even more striking. The study measured recurrence-free survival—the length of time patients lived before their cancer returned.

    • For the 8 patients who did not respond to the vaccine, the cancer returned after a median of 13.4 months.
    • For the 8 patients who did respond, their median recurrence-free survival had not yet been reached at the 18-month follow-up mark.

This indicates a dramatic and meaningful delay in cancer recurrence for those whose immune systems were successfully activated by the vaccine.

In one incredible case, the researchers witnessed the vaccine in action. A patient developed a small lesion in their liver, suspected to be a metastasis. A biopsy revealed it was not a full-blown tumor, but a dense cluster of the very same T-cells that the vaccine had trained. On subsequent scans, the lesion had vanished, suggesting the vaccine-activated T-cells had traveled to the site and eliminated the microscopic spread of cancer.

What's Next?

This was an early-stage trial with a small number of patients, and it's not a cure. However, its findings are incredibly promising. It provides powerful evidence that personalized mRNA vaccines can turn "cold" tumors "hot," making them vulnerable to an immune attack.

The success of this trial has paved the way for a larger, global randomized trial to confirm these findings. For a disease that has seen so little progress for so long, this research represents a beacon of hope and a monumental step forward in the fight against pancreatic cancer. It signals that the era of personalized immunotherapy is not just coming—for some, it has already begun.

 

Reference:  Rojas, L.A., Sethna, Z., Soares, K.C. et al. Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature 618, 144–150 (2023). https://doi.org/10.1038/s41586-023-06063-y 


Author: KuriousK. | Subscribe: Don’t miss updates—follow this blog!

Tuesday, June 17, 2025

Get Flawless RNA-Seq Results: This Enzyme Comparison Will Blow Your Mind!

 

A Comparative Analysis of Reverse Transcriptase Enzymes in Minimizing Bias and Enhancing Accuracy in RNA-Sequencing

 


RNA sequencing (RNA-Seq) has revolutionized our understanding of the transcriptome, offering unprecedented insights into gene expression, novel transcript discovery, and alternative splicing. However, a critical step in this process, reverse transcription (RT), often acts as a "black box," masking inherent biases that can profoundly impact the accuracy of results. This blog delves into the intricacies of RT bias, its contributing factors, and the strategies for its mitigation, ensuring more reliable and interpretable RNA-Seq data.

The Unseen Hurdles: Understanding Reverse Transcription Bias in RNA-Seq

At its core, RNA-Seq relies on converting unstable RNA into more stable complementary DNA (cDNA) using reverse transcriptase enzymes. While seemingly straightforward, this conversion is a major source of bias, broadly categorized into intrasample bias (uneven representation within a single sample) and intersample bias (inconsistencies between different samples).

Several key factors contribute to these quantitative discrepancies:

    • RNA Secondary Structure: RNA molecules can fold into complex 3D structures, forming stable impediments (e.g., hairpin loops, stem-loops). These structures can block primer binding or stall the RTase, leading to an underrepresentation of highly structured transcripts. Some RTases can be over 100-fold more efficient at navigating these structures than others.
    • Primary RNA Sequence Characteristics (e.g., GC content): High Guanine-cytosine (GC) content often correlates with increased RNA secondary structure stability, making these regions challenging for RTases. Studies in microbial communities have shown that temperature-induced RT bias can be partially explained by the G-C content of bacterial groups.
    • Primer-RNA Interactions and Priming Efficiency: The choice of priming strategy (oligo(dT), gene-specific, or random) significantly impacts bias. Complex RNA structures can obstruct primer annealing, leading to inefficient priming. While random primers can offer higher cDNA yield, they may also introduce their own biases and decrease reproducibility.
    • RNase H Activity and Template Switching: Most retroviral RTases possess an RNase H domain, which degrades the RNA strand in an RNA:cDNA hybrid. While crucial for viral replication, in RNA-Seq, this activity can cause premature degradation of the RNA template, leading to a negative bias against longer transcripts. More insidiously, RNase H can facilitate template switching, where the RTase jumps to another RNA molecule or a different region, creating "falsitrons" (large intramolecular deletions) or fused cDNA molecules, confounding analysis.
    • Consequences on Gene Expression and Transcript Discovery: The cumulative effect of these biases is substantial. They can create artificial impressions of differential abundance among transcripts, lead to non-uniform coverage across transcripts, and compromise accurate reconstruction and quantification of transcript isoforms. For example, poly-A selection often introduces a significant 3' bias, overrepresenting the 3' ends of transcripts.


It's crucial to understand that these factors are interconnected. A high-GC transcript is more likely to form stable secondary structures, presenting a compounded challenge to the RTase. Addressing bias requires a holistic approach, considering the synergistic interplay of RNA characteristics, primer design, and enzyme properties.

The RT Arsenal: Key Biochemical Properties of Reverse Transcriptase Enzymes

The judicious selection of a reverse transcriptase enzyme is paramount for minimizing bias. Modern RTases are engineered versions of their retroviral ancestors, optimized for in vitro applications. Key properties to consider include:

    • Thermostability: The ability to maintain activity at higher temperatures (e.g., 50-65°C) is critical. Elevated temperatures help denature stable RNA secondary structures, making the template more accessible and reducing premature stops. For example, SuperScript IV and Luna RT are highly thermostable.
    • Processivity: This refers to the number of nucleotides an enzyme can synthesize without dissociating from its template. High processivity is essential for generating long, full-length cDNA strands, reducing 3'-end bias and improving representation of longer transcripts. SuperScript IV, Induro® RT, and MarathonRT (MRT) are recognized for their high processivity, with Induro® RT exceeding 20kb maximum product length.
    • RNase H Activity: Most modern RTases are engineered with reduced or inactive RNase H domains (e.g., SuperScript IV, Luna RT, ProtoScript II RT, Induro® RT). This minimizes premature template degradation and template switching artifacts.
    • Fidelity: The accuracy of DNA synthesis from an RNA template (its error rate). While retroviral RTs are inherently error-prone, improvements in workflows like Unique Molecular Identifiers (UMIs) can help assess and correct for RT-induced errors.
    • Sensitivity: The ability to efficiently convert RNA to cDNA at very low input concentrations is critical for single-cell RNA-Seq. SuperScript IV is noted for its high sensitivity, capable of generating cDNA from as little as 10 pg of RNA.
    • Inhibitor Resistance: The ability to perform effectively in the presence of contaminants from biological samples (e.g., FFPE tissue, RNA extraction carryover). SuperScript IV has significantly improved resistance to various inhibitors.

 


Choosing an RTase is not about picking the "newest" enzyme, but rather selecting one that strategically balances these engineered characteristics to best suit the specific experimental design.

        Table 1: Key Biochemical Properties of Common Reverse Transcriptase Enzymes 

*Engineered to have an inactive RNase H domain, but still possesses some RNase H activity. ** The RT does possess terminal transferase activity, but the added nontemplated nucleotides are not suitable for efficient adaptor ligation by template switching. *** 3kb using random hexamers and poly-d(T) primers; up to 12kb with gene-specific primers; one-step RT-qPCR Luna® mixes produce cDNA <1kb.15

The Field of Play: Comparative Performance of Commercial and Specialized RTases

The landscape of commercial RTases is diverse. While MMLV-derived RTases, particularly the SuperScript series (II, III, IV), are frequently cited for robust performance, others like Maxima H-, ProtoScript, Luna, WarmStart RTx, Induro, AMV, and M-MuLV also play significant roles.

Performance Comparison Highlights:

    • Yield and Reproducibility: Maxima H- and SuperScript IV consistently demonstrate superior efficiency in converting RNA to cDNA, yielding higher positive reaction rates and expression levels. Absolute reaction yields can vary widely (7.3% to 137.9%) across different RTases.
    • Sensitivity to Low RNA Input: For single-cell RNA-Seq, Maxima H- and SuperScript IV are top performers, exhibiting a higher ability to capture rare transcripts and improving resolution in clustering analysis.
    • Handling Challenging RNA Templates:
      • Highly Structured RNA: MarathonRT (MRT) is exceptionally insensitive to RNA secondary structures, demonstrating consistent speed even with complex RNAs. TGIRT (Thermostable Group II Intron RT) also performs well, though slower than MRT. In contrast, SuperScript IV can be significantly hindered by stable structures, with one study showing 86% of reactions stopping at a specific GC stem loop, compared to only 8% for MRT.
      • Long Transcripts: Induro® RT (>20kb) and SuperScript IV (>12kb) are designed for long RNA molecules, while MRT is ultraprocessive, completing synthesis in a single pass.
      • Varying GC Content: Performing RT at higher temperatures (e.g., 55°C) can mitigate GC-content related biases, particularly for extreme GC content templates.
    • Specific Bias Mitigation Capabilities:
      • TGIRT-III: Engineered for enhanced thermostability, processivity, and fidelity, TGIRT-III can read through RNA modifications that stall conventional RTases, enabling precise mapping of these modifications. It's also less biased by specific modifications like m1A and effective at capturing full-length tRNAs.
      • Modified Retroelement RTs (e.g., BoMoC in OTTR): The Ordered Two-Template Relay (OTTR) method uses a modified Bombyx mori R2 protein (BoMoC) to capture obligatorily end-to-end sequences and simultaneously append sequencing adapters. This significantly minimizes biases and information loss, especially for low-input microRNA samples.

Table 2: Comparative Performance of Selected RTases in Minimizing Bias Across Diverse Transcripts


The emergence of specialized RTases signifies a growing recognition that a "one-size-fits-all" approach is not optimal. Researchers must consider the unique biochemical characteristics of their target RNA populations and the tailored capabilities of specialized RTases.

 


The Path Forward: Strategies and Best Practices for Minimizing RT-Induced Bias

Minimizing RT bias requires a multi-faceted approach, integrating careful wet-lab optimization with sophisticated bioinformatic corrections:

Optimizing RT Reaction Conditions:
      • Higher reaction temperatures (55°C or above) are strongly recommended for thermostable RTases to denature stable RNA secondary structures and resolve GC-content impediments.
      • Empirically determine optimal incubation time for certain RNA templates.
  1. Informed Enzyme Selection:

      • Low-input/single-cell RNA-Seq: Prioritize Maxima H- or SuperScript IV for their high sensitivity and reproducibility.
      • Highly structured/long RNA transcripts: Opt for highly thermostable and processive enzymes with minimal RNase H activity, such as MarathonRT (MRT), TGIRT-III, Induro® RT, or SuperScript IV. For precise end-to-end capture of structured small RNAs, consider specialized methods like OTTR.
      • Samples with inhibitors/degraded quality: SuperScript IV is a robust choice due to its enhanced inhibitor resistance.
      • GC-content bias concerns: Perform RT at higher temperatures (e.g., 55°C).
  2. High-Quality RNA Input: Always begin with high-quality, intact RNA (e.g., RNA Integrity Number (RIN) > 6). Degraded RNA can significantly introduce biases like uneven gene coverage and 3'–5' transcript bias.
    Reference RNA Samples and ERCC Spike-ins: Include well-characterized reference RNA samples and External RNA Control Consortium (ERCC) spike-ins to assess RNA-Seq performance and quantify RT bias. Deviations from expected values indicate sequence-dependent or protocol-dependent biases.
    Bioinformatic Approaches: While wet-lab strategies are essential, bioinformatic tools can play a complementary role. Computational models can help remove biases related to primary RNA sequence characteristics. Tools like Salmon attempt to correct for local sequence biases, GC content biases, and positional biases. However, these corrections have limitations and cannot fully compensate for fundamental issues introduced during wet-lab steps.

    The most robust approach involves a synergistic interplay between meticulous wet-lab optimization and sophisticated dry-lab correction. Bioinformatic correction should not be seen as a substitute for sound experimental practices.

     


    Conclusion and Recommendations for Accurate RNA-Seq

    The reverse transcription step is a critical, yet often underestimated, source of technical bias in RNA-Seq. These biases, stemming from complex interactions between RNA characteristics, primer design, and RTase properties, can significantly distort quantitative accuracy and transcriptomic representation.

    Modern RTases, with enhanced thermostability, processivity, reduced RNase H activity, high sensitivity, and inhibitor resistance, are pivotal in achieving unbiased cDNA synthesis. Newer generation enzymes like SuperScript IV and Maxima H- consistently outperform older versions, especially in low RNA input scenarios. Specialized RTases such as TGIRT-III and those in the OTTR method offer unique advantages for profiling difficult-to-capture RNA populations.

    To maximize RNA-Seq accuracy, it's recommended to:

    • For low-input/single-cell RNA-Seq, prioritize Maxima H- or SuperScript IV.
    • For highly structured/long RNA, opt for MarathonRT (MRT), TGIRT-III, Induro® RT, or SuperScript IV. Consider OTTR for precise end-to-end capture of structured small RNAs.
    • For inhibitor-prone/degraded samples, choose SuperScript IV.
    • For GC-content bias, perform RT at higher temperatures (e.g., 55°C) and consistently use the same RT enzyme across comparative studies.
    • Always use high-quality RNA (RIN > 6-7.5).
    • Integrate ERCC spike-ins for robust quality control and bias assessment.

Future RTase engineering will likely focus on even greater fidelity, processivity, and resistance to RNA modifications. Coupled with novel library preparation chemistries (e.g., direct RNA sequencing), these advancements will continue to drive the field toward ever more precise and reliable transcriptome analyses.

Featured Story

How RNA Regulates Metabolic Stress