Single-cell RNA sequencing (sc-RNA-seq) has fundamentally reshaped the field of transcriptomics by offering an unparalleled resolution into gene expression at the individual cell level. The global market of sc-RNA-seq, valued at $2.45 billion in 2024, is projected to reach $9.91 billion by 2034.
Recent advances have made scRNA-Seq more accessible and cost-effective. With platforms now capable of analyzing tens of thousands of cells per run at just a few cents per cell, the technology is becoming a staple in biomedical research.
These improvements are driving significant progress in fields like cancer, where scRNA-Seq helps pinpoint tumor heterogeneity and treatment-resistant cells, leading to more targeted therapies.
In this blog, we will explore the role of scRNA-seq in biomarker discovery and its implications for targeted therapies.
- scRNA-seq reveals gene expression at the single-cell level, enabling the discovery of rare cell types and subtle biomarkers.
- From identifying drug resistance in cancer to uncovering immune cell profiles in neurological and autoimmune diseases, scRNA-seq supports more accurate diagnosis, prognosis, and personalized therapy.
- scRNA-seq detects low-abundance transcripts and integrates with other modalities (e.g., scATAC-seq), offering a comprehensive view of disease biology and strengthening biomarker validation.
- scRNA-seq needs careful handling due to data complexity, high costs, and technical steps like normalization and cell typing.
- Affordable, AI-powered solutions that streamline every step of scRNA-seq projects, making advanced biomarker discovery more accessible.
What is scRNA-Seq?
scRNA-seq is an advanced technique that provides a high-resolution analysis of gene expression at the individual cell level, unlike traditional RNA-seq, which gives an average gene expression profile from a mixed cell population.
Core Principles of scRNA-seq
- Cell Isolation: Individual cells are first isolated from a biological sample (such as tissue or cell culture). This is done through techniques like fluorescence-activated cell sorting (FACS), micromanipulation, laser capture microdissection (LCM), or droplet-based microfluidic systems.
- RNA Extraction and Conversion: Once isolated, cells undergo lysis to release their RNA. The messenger RNA (mRNA) is then reverse transcribed into complementary DNA (cDNA).
- Amplification: The cDNA is amplified to create enough material for sequencing. This step is essential because the RNA content in a single cell is very small.
- Library Preparation: The amplified cDNA is then processed into a sequencing library, which is required for the high-throughput sequencing stage.
- High-Throughput Sequencing: The sequencing libraries are subjected to high-throughput sequencing, which generates the raw data on gene expression.
- Data Analysis: This stage involves several steps:
- Preprocessing to clean and organize the data.
- Quality Control to filter out low-quality data.
- Cell Clustering to identify distinct cell types or subpopulations.
- Differential Gene Expression Analysis to identify genes with altered activity across different cells or conditions.
- Pathway Analysis to understand the functional implications of the observed gene expression changes.
Now, let’s find out how scRNA-seq is beneficial for biomarker discovery.
Benefits of scRNA-seq in Biomarker Discovery

scRNA-seq accelerates the discovery of actionable biomarkers by enabling more granular insights into cellular behaviors and interactions. Here are the benefits of scRNA-seq in biomarker discovery:
- Resolution of Cellular Heterogeneity: scRNA-seq uncovers the diversity of cell types and rare subpopulations that bulk RNA-seq masks, revealing hidden cellular states crucial for disease understanding.
- Identification of Novel Cell Types and States: The technique can detect previously unrecognized cell subtypes and transient states, particularly in complex conditions like cancer and autoimmune disorders.
- Precise Biomarker Discovery: scRNA-seq enables the identification of biomarkers directly involved in disease mechanisms, improving diagnostic and prognostic accuracy over traditional methods.
- High-Resolution Transcriptomic Insights: Provides detailed gene expression profiles at the single-cell level, capturing subtle expression changes that bulk RNA-seq would miss, even with minimal starting material.
- Sensitivity to Low Abundance Transcripts: Capable of detecting rare and low-copy transcripts, offering greater sensitivity for finding biomarkers in small populations of cells.
- Multi-Omics Integration: By combining scRNA-seq with other single-cell modalities (e.g., scATAC-seq, scBCR-seq, scTCR-seq), scRNA-seq enables a more holistic view of cellular function, enhancing biomarker discovery.
Multi-omics integration provides a deeper understanding of genotype-phenotype associations, increasing confidence in biomarker validation and clinical applicability. So, let’s see applications of scRNA-seq in biomarker discovery.
Role of scRNA-seq in Biomarker Discovery

scRNA-seq, powered by advancements in high-throughput sequencing and microfluidics, enables the detection of subtle genetic and protein expression variations at the individual cell level. This technology is essential for exploring cellular heterogeneity, uncovering novel genetic traits linked to clinical outcomes, and providing insights into disease progression and prognosis in clinical trials.
1. In Cancer Research
scRNA-seq has been extensively applied across numerous cancer types, with studies integrating data from over 41,900 single cancer cells to illuminate the intricate landscape of tumor heterogeneity and identify promising therapeutic targets.
It has uncovered poor prognosis and medication resistance in various cancers, including lung, breast, ovarian, and gastric cancers. For example,
- Immune Checkpoint Inhibitor (ICI) Prognosis & irAE Severity: A study used scRNA-seq on peripheral blood mononuclear cells (PBMC) from patients before immune checkpoint inhibitor (ICI) treatment to map immune cell diversity. This helped identify biomarkers that predict ICI resistance and the severity of immune-related adverse events (irAEs), allowing for better patient selection and treatment planning.
- Tumor Microenvironment (TME) Characterization: scRNA-seq is key to studying the TME, which includes cancer, immune, and stromal cells. It provides single-cell insights into how these cells interact, helping to understand treatment resistance, track tumor growth, and find new therapeutic targets.
2. In Autoimmune Diseases
scRNA-seq is a powerful tool for analyzing gene expression in autoimmune conditions, offering high throughput and resolution to explore cell development, differentiation, and function across time and space.
- Systemic Lupus Erythematosus (SLE): scRNA-seq, combined with scBCR-seq, revealed a bias in V(D)J gene usage for B-cell receptors (BCR) in SLE patients, specifically a preference for IGHV3-23, aiding in the development of new diagnostic and therapeutic approaches. Nickerson et al. (2023) further uncovered the high heterogeneity of age-associated B cells (ABCs) in SLE, highlighting their dependence on Toll-like receptor (TLR) signaling and reactivity to self-antigens.
- Kawasaki Disease (KD): scRNA-seq revealed an increase in plasma cells in KD patients, indicating enhanced B-cell activation and antibody responses following IVIG treatment.
- Psoriasis: scRNA-seq found lower diversity in IGHA1 and IGHG1 and increased usage of IGHV in psoriasis patients, alongside increased CDR silent mutations in IGHA and rearrangements in IGHG, and higher BCR selection pressure.
- Membranous Nephropathy (MN): Substantial alterations in gene expression patterns and clonal proliferation are observed within memory and naive B cells in NEG pMN patients. An expanded CD38+ naive B cell population demonstrated activation-related functional properties and increased IgM/IgD to IgG1 class switching, leading to heightened autoantibody production.
- Inflammatory Bowel Disease (IBD) / Ulcerative Colitis (UC): Boland et al. (2020) revealed clonal relationships and heterogeneity of immune cells in UC patients, noting a significant increase in IgG1+ plasma cells in colonic tissue with unique transcriptional features and BCR clonotypes.
- Rheumatoid Arthritis (RA): Higher somatic mutation rates were observed in non-naive B cells from RA patients, particularly in the synovial tissue.
3. In Neurological Disorders
scRNA-seq effectively reveals how specific cell types contribute to disease progression in neurological conditions. For example,
- Cerebrospinal Fluid (CSF) Analysis: A comprehensive study utilized scRNA-seq on CSF and peripheral blood mononuclear cells (PBMCs) from individuals across various neurological diseases. This research identified unique CSF immune cell populations and associated biomarkers:
- Myeloid Cells: Unique CSF-exclusive populations were identified, such as ACY3+ DCs (characterized by ACY3, S100B, KIT) and AREG+ cDC2s (marked by AREG, RGS1), which were found to be more abundant in MS subjects. Microglia-like cells (
- TREM2, SLC2A5, CH25H) were also uniquely populated in the CSF. Notably, FN1+ MG (microglia) showed a higher frequency in neurodegenerative diseases and expressed key AD risk genes (ABI3, CD33, PTK2B).
- B Cells and Plasmablasts: The CSF exhibited a higher frequency of switched memory and activated B cells, along with plasmablasts. Atypical memory B cells were more frequent in MS subjects, characterized by elevated TBX21, ITGAX, and FCRL5 expression.
- CD4+ T Cells: Th CD4+ T cells (TNFRSF4, NSG1, EGR1) and CCR5hi effector memory cells (CCR5, PDCD1, CXCR6) were found more frequently within the CSF compartment.
- CD8+ T Cells: Naive CD8+ T cells and Tem GZMK+ CD8+ T cells were observed with heightened representation in the CSF.
- NK Cells: CD56bright NK cells, proliferating NK cells, and Tissue-resident-like NK cells (TR-NK) were significantly elevated in the CSF. A unique population of
- Group 3 innate lymphoid cells (ILC3s) were also found to be abundant in the CSF.
- Alzheimer’s Disease (AD): scRNA-seq has identified disease-associated microglia (DAM), characterized by upregulated expression of Trem2, ApoE, and inflammatory cytokines. These microglial subtypes are linked to amyloid progression and synaptic loss, correlating with clinical markers of cognitive decline.
- Parkinson’s Disease (PD): scRNA-seq has revealed unique transcriptional changes in specific subtypes of dopaminergic neurons, related to oxidative stress, mitochondrial dysfunction, and impaired proteostasis. Genetic markers like GBA and LRRK2 mutations are also linked to more aggressive or variable disease phenotypes.
- Huntington’s Disease (HD): scRNA-seq has enabled detailed analysis of striatal medium spiny neurons, revealing transcriptional dysregulation driven by mutant huntingtin protein (HTT). Genetic modifiers such as FAN1 and MLH1 have been identified to influence disease onset and severity.
Apart from these, emerging as non-invasive biomarkers, DNA methylation patterns (e.g., hypermethylation of the APP promoter in AD, SNCA promoter in PD) and histone modifications are being investigated for their potential to predict disease progression.
4. In Cardiovascular Diseases
scRNA-seq helps identify stem cell differentiation processes and pinpoint targets for disease treatment in the cardiovascular system.
- Cardiovascular Calcification: A 2024 study identified two key biomarkers, ITGAX and MYD88, as diagnostic indicators of cardiovascular calcification. Both were found to be significantly upregulated in calcified samples and strongly associated with immune processes. scRNA-seq confirmed their high expression in multiple immune cell types, suggesting potential diagnostic and therapeutic targets.
- Adult Heart Disease: scRNA-seq offers insights into cardiac cellular heterogeneity in pressure overload-induced heart failure models, elucidating genetic programs underlying cardiac hypertrophy and failure, and tracking cell fate transitions in myocardial infarction.
- Vascular Lesions (Atherosclerosis and Abdominal Aortic Aneurysm): scRNA-seq has advanced the understanding of atherosclerosis as a multifactorial disease and revealed specific types of smooth muscle cells and macrophages involved in AAA development.
5. In Infectious Diseases
scRNA-seq, alongside scTCR-seq and scBCR-seq, is becoming an increasingly vital tool for studying cerebrospinal fluid (CSF) and other tissues to understand various aspects of neurological diseases, including those with infectious etiologies.
- Fibrostenotic Crohn’s Disease (CD): A study utilized scRNA-seq to characterize genes specific for strictures at a cellular level in fibrostenotic Crohn’s disease.
- It identified GREM1 as exclusively expressed in fibroblasts from strictures, positioning it as a potent gene associated with stricture development.
- EHD2 and FGF2 were predominantly expressed in fibroblasts and endothelial cells, with EHD2 showing higher expression in endothelial cells from strictures.
- LY96 and SRM were found to be upregulated in immune cells within intestinal strictures, underscoring the intricate interaction between stromal and immune cells in the disease.
These identified genes and pathways hold promise as biomarkers for the early detection of strictures and can guide the development of targeted therapies, potentially improving outcomes for patients with Crohn’s disease.
Across various disease categories, scRNA-seq highlights biomarkers found within specific cell types or states, rather than broad tissue averages. This specificity enhances diagnostic and prognostic accuracy, leading to better patient stratification and targeted therapies with fewer off-target effects.
Moreover, scRNA-seq reveals biomarkers that change with disease activity or treatment response. Examples include the increase of IL-4R−IFN-β+ B cells correlating with SLE activity, B cell shifts after IVIG treatment in Kawasaki Disease, and clonal B cell expansion before treatment in Myasthenia Gravis.
Predictive biomarkers for immune checkpoint inhibitor resistance and immune-related adverse events further demonstrate scRNA-seq’s ability to track disease progression and therapy responses.
While scRNA-seq offers transformative insights, it also presents significant technical and computational challenges that researchers must navigate.
Technical Challenges in scRNA-seq in Biomarker Discovery

Researchers employing scRNA-seq for biomarker discovery frequently encounter a range of experimental and computational hurdles that necessitate careful consideration and specialized solutions. These are:
- Low RNA Input: Single cells contain limited RNA, leading to incomplete reverse transcription and amplification. Optimizing cell lysis, RNA extraction, and using pre-amplification methods can improve RNA yield and sequencing quality.
- Data Volume and Sparsity: scRNA-seq generates large, high-dimensional datasets, often with sparse data due to dropout events. This sparsity complicates statistical analysis and modeling.
- Batch Effects: Technical and biological variations across sequencing runs, instruments, or sample collections can create batch effects, obscuring biological signals. Correcting for batch effects with tools like Scarf and Seurat can be computationally intensive.
- Data Normalization: Normalizing scRNA-seq data to account for sequencing depth and library size is critical but can introduce biases if not done carefully, distorting biological variations.
- Cell Type Annotation: Accurately annotating cell types in complex datasets is challenging. Traditional marker gene methods can be biased or inaccurate. Machine learning and deep neural networks (DNN) offer improved annotation by handling noise and biological variability without relying on predefined markers.
- Computational Demands: Advanced analytical methods for scRNA-seq require significant computational resources. Tools like scaLR efficiently process large datasets in low-resource environments by partitioning data into manageable batches, offering faster execution times and lower memory usage.
The sheer scale and inherent complexity of scRNA-seq data, characterized by its vast volume, high dimensionality, and sparsity, mean that traditional bioinformatics approaches are often insufficient for extracting meaningful insights. That’s where Biostate AI comes in.
How Biostate AI Will Simplify RNA-Seq Analysis for Your Research
Biostate AI addresses these challenges by offering an affordable, streamlined, and AI-driven RNA sequencing service. We reduce the technical complexity and computational load typically associated with RNA-seq, making it accessible for researchers of all sizes.
Here is what we offer:
- Affordable Pricing: High-quality RNA sequencing starting at just $80 per sample, making advanced analysis accessible for all research teams.
- Flexible Sample Requirements: Accommodates challenging samples such as FFPE tissue and as little as 10µL blood.
- Low RIN Compatibility: Works with samples having as low as RIN 2, unlike typical industry standards of RIN ≥5.
- Comprehensive RNA Coverage: Includes mRNA, lncRNA, miRNA, and piRNA, providing broad insights into gene expression and regulatory elements.
- AI-Powered Analysis: Utilizes OmicsWeb AI and Prognosis AI, transforming raw RNA data into accurate disease predictions and therapy selection.
- Rapid Turnaround: Delivers results within 1-3 weeks, ensuring quick data-driven insights for research progression.
Biostate AI’s combination of affordability, flexibility, and AI-driven analytics is transforming the accessibility and efficiency of RNA sequencing, accelerating biomarker discovery, and supporting the move from research to clinical applications.
Final Words!
Recent studies demonstrate how scRNA-seq in biomarker discovery is advancing clinical applications, from predicting cancer treatment responses to mapping immune dysfunction in autoimmune diseases and pinpointing cellular vulnerabilities in neurological disorders.
This precision makes scRNA-seq in biomarker discovery a powerful tool for developing more accurate and actionable biomarkers for clinical use. However, scRNA-seq presents technical and computational challenges that require advanced experimental protocols to work on.
Biostate AI provides an affordable solution, offering high-quality RNA sequencing with advanced AI-driven analysis, making scRNA-seq more accessible for researchers.
Starting at just $80 per sample and with a rapid turnaround time of 1-3 weeks, Biostate AI makes RNA-seq analysis affordable and accessible to all. Get your quote today!
FAQs
1. How does single-cell RNA sequencing improve upon traditional bulk RNA sequencing for biomarker discovery?
Single-cell RNA sequencing (scRNA-seq) offers a significant improvement by analyzing gene expression at the individual cell level, unlike bulk RNA-seq’s population averages. This high resolution reveals cellular heterogeneity, uncovers rare cell types, and detects subtle transcriptional changes, enabling precise identification of cell-type-specific and disease-associated biomarkers that traditional methods often miss. The result is more accurate, actionable biomarkers for diagnosis, prognosis, and therapy selection.
2. Can scRNA-seq help identify biomarkers in rare or previously uncharacterized cell types?
Yes. Unlike bulk RNA-seq, scRNA-seq can uncover rare cell populations and their unique gene expression profiles, making it possible to discover biomarkers specific to these rare or novel cell types that would be missed in population-averaged approaches.
3. Is it possible to discover non-coding RNA biomarkers (like lncRNAs or circRNAs) using scRNA-seq?
Absolutely. Some advanced scRNA-seq protocols can capture both polyA+ and polyA- RNAs, enabling the detection of long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) as potential biomarkers, which are increasingly recognized for their roles in disease.
4. How does scRNA-seq contribute to biomarker discovery in liquid biopsies or non-invasive samples?
scRNA-seq has been adapted for use with circulating tumor cells (CTCs) and other cells in liquid biopsies, allowing for the identification of disease-associated markers from blood samples, which can be less invasive than traditional tissue biopsies.
5. Can scRNA-seq data be used to repurpose existing drugs by identifying new cellular targets or drug response biomarkers?
Yes. By comparing gene expression profiles of cells treated with various drugs, scRNA-seq can reveal unexpected similarities or differences in cellular responses, suggesting new uses for existing drugs or uncovering markers of drug sensitivity or resistance.
