Collections

Single-cell and Spatial Omics
Sort by Default Latest Most read  
Please wait a minute...
  • Select all
    |
  • Reviews
    WANG Na,ZHAO Li-Nan, HAN Ze-Guang
    Chinese Journal of Biochemistry and Molecular Biol. 2020, 36(5): 488-493. https://doi.org/10.13865/j.cnki.cjbmb.2020.03.1367
    Traditional bulk tumor analysis with high-throughput DNA sequencing yields an average of gene expression in a population of cells, ignoring the heterogeneity between tumor cells. Hepatocellular carcinoma is one of the most common malignant tumors of human beings, with an obvious intratumor heterogeneity. The analysis of cell population could not accurately determine the clonal structures of tumor cells and identity the cell types, status and subpopulation distribution of immune microenvironment. The analysis at single-cell resolution is necessary to reveal the subpopulation of multiple malignant cells within tumors, as well as the cell types and their status in tumor microenvironment, promoting to find new therapeutic targets and effective biomarkers in clinical diagnosis, subtyping and treatment. This paper reviews the application of single-cell DNA and RNA sequencing technology in the study of hepatocellular carcinoma immune microenvironment, heterogeneity, clonal evolution, metastasis mechanism and biomarkers discovery. In addition, we also summarize the advantages of single cell multiomic sequencing in finding new tumor subsets, accurately identifying the heterogeneity of tumor cells and understanding the composition of tumor microenvironment.
  • Reviews
    LI Bo, MA Yuan-Wu
    Chinese Journal of Biochemistry and Molecular Biol. 2020, 36(9): 1024-1032. https://doi.org/10.13865/j.cnki.cjbmb.2020.06.1135

    Hematopoietic stem cells (HSCs) play a crucial role in the hematopoietic system, including maintaining self-renewal and differentiating into all kinds of functional blood cells. If there is functional impairment in HSCs, it will lead to many blood diseases, for instance, myeloid leukemia, lymphocytic leukemia, myeloproliferative disorders, etc. Presently, the most important thing in this field is to understand the mechanisms and regulation networks of HSC senescence and leukemic transformation. As a heterogeneous population, HSC is regulated by all kinds of factors in a complex and hierarchical manner. Those factors include cytokines, growth factors, extracellular matrix proteins and so on. With the development of the single cell technology, we can learn the HSCs’ cell fate regulation network through genomics, transcriptomics, epigenetics and proteomics. With the genome editing tools, we can generate genetically modified animal models to study the gene function and test the safety and efficacy of HSC-related product. In addition, genome editing tools combined with the single cell technology can achieve the lineage tracing and cell fate regulation of HSCs. Therefore, single-cell sequencing and gene editing technologies are revolutionizing the development of this field. In this review, we summarize the advanced applications and problems of those new tools to promote their usage in the future studies.

  • Techniques and Methods
    SHI Lei, SHEN Xiang-Yu, SHEN Yuan
    Chinese Journal of Biochemistry and Molecular Biol. 2020, 36(12): 1508-1513. https://doi.org/10.13865/j.cnki.cjbmb.2020.10.1324
    We developed a microinject method in the unicellular Paramecium model to establish a proper transgene methodology for protists for genetic research,. In this study, intraflagellar transport (IFT) genes including IFT46 and IFT43 with its own promoter were cloned into Pzz02-GFP plasmids, then recombinant plasmids were transformed into nutrient macronuclear of Paramecium tetraurelia by microinjection. The result of Western blotting shows that IFT46-GFP or IFT43-GFP fusion proteins were produced in Paramecium cells. By fluorescence or confocal microscopy, we record the bi-directional movements of IFT proteins along cilia, which confirms the efficiency of the microinjection technology in transforming exogenous genes to unicellular models like Paramecium.
  • Research Papers
    CI Bai-Quan, XIAO Yao, TAN Tao, WANG Feng
    Chinese Journal of Biochemistry and Molecular Biol. 2021, 37(12): 1621-1637. https://doi.org/10.13865/j.cnki.cjbmb.2021.09.1202
    In mammals, by functioning as the progenitor of matured gametes (sperms or oocytes), primordial germ cells (PGCs) are of remarkable importance in life reproduction and handing down genetic information from generation to generation. However, in the case of primates, very little is known on their gene modules and molecular networks underlying the specification of PGCs. In this study, firstly we carried out the single-cell RNA-seq (scRNA-seq) in PGC differentiation (at three time points of day 0, day 2, day 4) of Macaca fascicularis, and further identified the gene modules related to PGC development based on weighted gene co-expression network analysis (WGCNA). In total, we obtained 91, 55 and 66 single cells at day 0, day 2 and day 4, respectively. Based on 212 scRNA-seq data at three different time points, we identified 17 different gene modules. Among which, the MEsalmon module was highly positively associated with undifferentiated day 0 with a Pearson correlation coefficient (PCC) of 0.89 and a P-value of 2E-72. While the MEblue module was positively associated with both day 2 and day 4, and interestingly, its PCC remarkably increased from 0.24 (P-value=5E-04) to 0.66 (P-value=4E-28) with the PGC differentiation from day 2 to day 4. Enriched terms for the MEblue module were observed to be BMP and Wnt signaling pathways, and the biological processes of epithelial cell differentiation, male gonad development, etc. Thus, it is very likely that the co-expressed genes in this module are involved in driving the initiation of PGC differentiation in Macaca fascicularis. Furthermore, we identified the differentially expressed genes at each time points and performed GO/KEGG analysis. Finally, combined with the STRING database, we constructed the protein-protein interaction (PPI) networks for the development of PGCs in Macaca fascicularis using Cytoscape and MCODE. In total, we obtained 8 densely-connected core subnetworks closely related to PGC differentiation, identified the hub regulators such as BMP4, WNT3, TFAP2C and SOX17 in the PPI networks, and performed comparative analysis among human, Macaca fascicularis and mouse. In summary, our findings provide the basis for deep understanding of the cell biology and gene regulation underlying the fate of PGCs at the single cell level and shed light on embryonic developmental characteristics at the level of systems biology in a highly evolved primate.
  • Reviews
    LIU Yi-Chen, CHENG Ming, CAI Rui
    Chinese Journal of Biochemistry and Molecular Biology. 2025, 41(2): 201-209. https://doi.org/10.13865/j.cnki.cjbmb.2024.12.1177
    Adipose tissue consists of multiple types of cells with different proliferative capacities, different directions of differentiation, and different executive functions, so parsing the subcellular types of adipose tissue is of great significance in elucidating the molecular mechanisms of the formation of important traits in the organism. In recent years, with the substantial improvement of the sensitivity of scientific instruments, the continuous improvement of the automation level and precision from sample preparation to data analysis, and the need to improve the resolution of scientific research, scientists have come up with a variety of studies on single cells, and the single-cell multi-omics technology has developed rapidly. Single-cell multi-omics technology mainly includes single-cell genomics, single-cell transcriptomics, single-cell proteomics and single-cell metabolomics and the joint application of each of them. This technology realizes the ability to reveal the heterogeneity of adipocytes, helps discovering new adipocyte isoforms and exploring the trajectory of adipocyte differentiation at the level of single-cell resolution at different levels of biological processes, and has achieved great breakthrough progress in the field of biology and medicine. Single-cell multi-omics technology has been serving as an entry point for research on the occurrence and development of many diseases and accelerating the development of key emerging technologies such as gene editing and stem cell breeding. In this paper, we summarize the types of single-cell multi-omics technology and its characteristics, review the progress of the application of this technology in the deposition of intramuscular, intermuscular, subcutaneous and visceral adipose tissues, and discuss its prospects and challenges. This review aims to demonstrate the important role of this technology in the study of adipose deposition, and provide theoretical basis for the analysis of the trait formation mechanism and the treatment of metabolic disorders, such as obesity, in human beings.
  • CJBMB: 40 Years of Biochemistry and Molecular Biology in China Special Column of Single-cell and Spatial Omics
    LIN Guan-Chuan, PAN Xing-Hua
    Chinese Journal of Biochemistry and Molecular Biology. 2025, 41(11): 1559-1565. https://doi.org/10.13865/j.cnki.cjbmb.2025.10.0001
    Single-cell and spatial omics technologies are spearheading a profound paradigm shift in the life sciences, moving beyond ‘population averages’ to ‘single-cell resolution’ and reintegrating ‘cellular constitution’ with ‘tissue spatial architecture’, thereby dramatically advancing our understanding of biological complexity. This review provides a brief comprehensive overview of recent advancements in the field. Technologically, the evolution has progressed from single-cell transcriptomics to integrated approaches capturing multiple molecular layers simultaneously, while the emergence of transcriptome-badsed spatial omics has successfully preserved the spatial positioning of cells or microscosystem within native tissues, and further enabling spatial epigenomics and spatial-multiomics. Computationally, artificial intelligence and machine learning have become central engines, powering tools for data integration, spatial deconvolution, cellular communication and other novel foundation models, which not only tackle the challenges of massive datasets but also serve as instruments for novel biological discovery. These technological leaps have fostered significant theoretical innovations. In clinical translation, these technologies, particularly in precision oncology, demonstrate transformative potential by dissecting tumor heterogeneity, mapping the spatial architecture of the tumor immune microenvironment, and enabling disease modeling through single-cell-guided deconvolution of bulk data, offering new avenues for diagnosis, prognosis, and personalized therapy. Despite ongoing challenges in technological throughput, computational scalability, and clinical integration, the continued convergence of single-cell and spatial omics with AI promises to propel basic research towards a more mechanistic and predictive era, ultimately reshaping the future of precision medicine.
  • Research Paper
    ZHANG Xin-Tong, ZHU Jian-Jun, WU Jin, WU Hao, LU Fan, ZHANG Wen-Tao, CHANG Jing-Jia, TANG Ting, OU Zhi-Gao, JIA Feng-Feng, LI Li, YU Peng-Fei, LIU Ming
    Chinese Journal of Biochemistry and Molecular Biology. 2025, 41(10): 1511-1528. https://doi.org/10.13865/j.cnki.cjbmb.2025.08.1145
    Hepatocellular carcinoma (HCC), which is essentially primary liver cancer, is closely related to CD8 T cell immune infiltration and immune suppression. We constructed a CD8+ T cells related risk score model to predict the prognosis of HCC patients and provided therapeutic guidance based on the risk score. Using integrated bulk RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq) datasets, we identified stable CD8+ T cell signatures. Based on these signatures, a 3-gene risk score model, comprised of KLRB1, RGS2, and TNFRSF1B was constructed. The risk score model was well validated through an independent external validation cohort. We divided patients into high-risk and low-risk groups according to the risk score and compared the differences in immune microenvironment between these two groups. Compared with low-risk patients, high-risk patients have higher M2-type macrophage content (P<0.0001) and lower CD8+ T cells infiltration (P<0.0001). High-risk patients predict worse response to immunotherapy treatment than low-risk patients (P<0.01). Drug sensitivity analysis shows that PI3K-β inhibitor AZD6482 and TGFβRII inhibitor SB505124 may be suitable therapies for high-risk patients, while the IGF-1R inhibitor BMS-754807 or the novel pyrimidine-based anti-tumor metabolic drug Gemcitabine could be potential therapeutic choices for low-risk patients. Moreover, expression of these 3-gene model was verified by immunohistochemistry. In summary, the establishment and validation of a CD8+ T cell-derived risk model can more accurately predict the prognosis of HCC patients and guide the construction of personalized treatment plans.
  • CJBMB: 40 Years of Biochemistry and Molecular Biology in China Special Column of Single-cell and Spatial Omics
    XIONG Ming-Fu, KONG Si-Yuan, ZHANG Yong-Sheng
    Chinese Journal of Biochemistry and Molecular Biology. 2025, 41(11): 1566-1578. https://doi.org/10.13865/j.cnki.cjbmb.2025.09.1276
    As an important tissue of the body (accounting for approximately 40% of body weight), skeletal muscle is composed of various cell types such as muscle fibers, muscle stem cells, and endothelial cells. It participates in physiological processes including movement, energy metabolism, and internal environment homeostasis through temporal and spatial specific regulation. Its development is divided into two critical stages: embryonic and postnatal periods. Abnormal development can lead to diseases such as muscular dystrophy and directly affect the yield and quality of livestock meat. In recent years, the combination of single-cell transcriptomics (scRNA-seq) and spatial omics (single-cell spatial omics technology) has provided a high-resolution research tool for analyzing the spatiotemporal dynamic regulatory network and intercellular interactions in skeletal muscle development. This article reviews the molecular mechanisms of skeletal muscle development and its application value in animal husbandry breeding, and systematically combs the research progress, analysis processes, data resources, and future directions of single-cell omics, spatial omics, and single-cell spatial omics technology in skeletal muscle development. Among them, single-cell omics can reveal the heterogeneity of skeletal muscle cells, myofiber differentiation trajectories in different livestock and poultry (such as cattle, pigs, and Tibetan chickens) through methods like pseudotime analysis and RNA velocity analysis. Furthermore, single-cell omics can identify key transcription factors (e.g., MYF5, MYOD1) and cell communication pathways (e.g., FGF7-FGFR2), and simultaneously clarify the molecular differences in myoblast differentiation timing and cell composition ratio among different breeds. Relying on technologies such as Visium and Seq-Scope, spatial omics realizes the spatial localization of gene expression in pathological models of mice, Atlantic salmon, and broiler chickens. Spatial omics also clarifies the spatial distribution laws of neuromuscular junction region-specific genes and inflammation-fibrosis cascade reactions, and makes up for the defect of losing spatial context in single-cell technology. Although there are limited direct application cases of single-cell spatial omics technology, it has already analyzed the abnormal fate of myoblasts in facioscapulohumeral muscular dystrophy through MERFISH technology. In terms of technology selection, it is necessary to consider research objectives, molecular modalities, and resolution requirements. At the same time, data analysis needs to address challenges such as data sparsity through methods like DCA denoising and RCTD cell type mapping. In addition, this article summarizes 16 muscle development-related databases including HCA and PanglaoDB. This review further discusses the potential applications of these three types of technologies in the directional regulation of myoblast fate, precise intervention in the growth cycle, improvement of microenvironment interactions, and the development of multi-omics genetic breeding models. This paper is providing a more comprehensive and detailed theoretical reference and technical support for basic research on skeletal muscle development and practical applications in the animal husbandry industry.
  • Research Paper
    YANG Yang, MA Yi-Xuan, FAN Xin-Yue, ZHAO Wen-Xue, QI Yi-Ming, GAO Ning, ZHAO Ju-Mei, DU Juan
    Chinese Journal of Biochemistry and Molecular Biology. 2025, 41(10): 1529-1540. https://doi.org/10.13865/j.cnki.cjbmb.2025.09.1203
    Inflammatory response, immunosuppression, and drug sensitivity have been reported to have a significant correlation with the disulfidptosis levels in cancer patients. However, the value of disulfidptosis in colorectal cancer therapy remains unclear. Therefore, we classified the CRC cells into different cell types using single-cell sequencing data and cell-specific markers and analyzed their relationship with the cell disulfidptosis level. We found that the high disulfidptosis regions were concentrated in epithelial-like CRC cells. Further exploration using the disulfidptosis and programmed cell death 1 inhibitor therapy treated differential expression genes indicated that CRC patients with high disulfidptosis levels exhibited a lower risk profile and increased sensitivity to immunotherapy. By using the spatial transcriptomic analysis, we found that ubiquinol-cytochrome c reductase core protein 1 (UQCRC1), a disulfidptosis-related gene, is highly expressed in epithelial-like CRC cells and co-localized with immune-infiltrated tumor regions. Additional bioinformatic analyses and experimental validation further confirmed that UQCRC1 was downregulated in CRC tissues. Overexpression of UQCRC1 suppressed CRC cell proliferation and migration.These findings indicate that UQCRC1 is a potential target for CRC diagnosis and treatment.
  • CJBMB: 40 Years of Biochemistry and Molecular Biology in China Special Column of Single-cell and Spatial Omics
    LI Liu-Jia-Yu, WANG Cheng, YU Li-Mei
    Chinese Journal of Biochemistry and Molecular Biology. 2025, 41(11): 1610-1621. https://doi.org/10.13865/j.cnki.cjbmb.2025.10.1268
    Mesenchymal stem cells (MSCs) hold great promise in regenerative medicine due to their multi-lineage differentiation potential and immunomodulatory properties. However, their functional heterogeneity and strong dependency on the microenvironment remain major challenges for clinical application. In recent years, the combination of single-cell transcriptome sequencing (scRNA-seq) and spatial transcriptomics sequencing (ST-seq) has provided revolutionary tools for systematically deciphering the heterogeneity, functional diversity, and microenvironmental interactions of MSCs. Using scRNA-seq, researchers have successfully resolved the functional heterogeneity of MSCs and identified key functional subpopulations, such as pro-angiogenic, immunoregulatory, and matrix-remodeling subsets. Meanwhile, ST-seq has revealed the distinct spatial distribution of MSCs within tissues and their dynamic interaction networks with the microenvironment. The integration of these two technologies has not only enabled the construction of a three-dimensional “identity-location-function” atlas of MSCs, but also uncovered spatiotemporal dynamic regulatory mechanisms of specific subpopulations during tissue repair. Looking forward, the combination of ultra-high-resolution ST-seq platforms such as Xenium, multi-omics integration, and artificial intelligence-driven analysis will shift MSCs research from descriptive studies toward precise intervention, offering new strategies for functional subpopulation screening and microenvironment reprogramming therapy. This review systematically summarizes the latest advances in scRNA-seq and ST-seq technologies in MSC research, discusses their applications in elucidating cellular heterogeneity, spatial microenvironment, and clinical translation, and provides a theoretical basis and technical guidance for precision treatment in regenerative medicine.
  • CJBMB: 40 Years of Biochemistry and Molecular Biology in China Special Column of Single-cell and Spatial Omics
    ZHU Xiao-Xi, WANG Cheng, YU Li-Mei
    Chinese Journal of Biochemistry and Molecular Biology. 2025, 41(11): 1600-1609. https://doi.org/10.13865/j.cnki.cjbmb.2025.10.1273
    Ischemic stroke (IS) research has faced bottlenecks due to the limitations of conventional technologies in resolving cellular heterogeneity and spatiotemporal dynamics. The development of single-cell and spatial omics technologies provides revolutionary tools to break through these constraints. Single-cell omics technologies, by performing high-throughput sequencing on thousands of cells, reveal the high heterogeneity and dynamic state transitions of neurons, glial cells, immune cells, and others post-IS. For instance, microglia contain pro-inflammatory and anti-inflammatory functional subsets, while astrocytes exhibit distinct activation state spectra. Pseudotime analysis further reconstructs the fate trajectories of cells during the damage and repair processes. Spatial omics technologies, conversely, reconstruct spatial maps of gene expression through in situ capture, elucidating molecular gradients between the ischemic core, penumbra, and healthy brain regions, and enabling the analysis of critical cell-cell interaction networks. Integrating the deep phenotyping capability of single-cell sequencing with the in situ localization information from spatial omics constitutes the current core strategy. This multimodal analysis allows for precise anchoring of cell subtypes to their spatial microenvironments, revealing their distribution patterns and functions, and constructing a more accurate atlas of cell-cell communication. This significantly advances the refined dissection of IS mechanisms. This strategy has already accelerated the discovery of potential biomarkers and spatiotemporally specific therapeutic targets. Although challenges remain in sample preparation, data integration, and technical noise, future interdisciplinary collaboration, multi-omics integration, and in-depth mining with artificial intelligence promise to comprehensively transform our understanding of IS. Ultimately, it holds the potential to promote advances in its early diagnosis, precise subtyping, and the development of individualized treatment strategies.
  • CJBMB: 40 Years of Biochemistry and Molecular Biology in China Special Column of Single-cell and Spatial Omics
    LUO Yu-Yan, LUO Xiao-Min, HUANG Jie-Ru, XU Si-Wen
    Chinese Journal of Biochemistry and Molecular Biology. 2025, 41(11): 1579-1589. https://doi.org/10.13865/j.cnki.cjbmb.2025.09.1270
    Single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) is a powerful technique for studying cellular heterogeneity and gene regulatory networks, widely applied in epigenetic research. However, the complexity of data analysis workflows and high programming requirements have limited its broader adoption among non-programmer researchers. To address this issue, we developed Signac.UIO, a modular and visual scATAC-seq analysis platform based on the R Shiny framework, integrating mainstream tools such as Signac and Seurat. The platform includes ten key modules covering quality control, cell filtering, dimensionality reduction, clustering, differential analysis, cell annotation, pathway enrichment, motif analysis, and transcription factor footprinting. Through a graphical user interface, users can perform full analyses and obtain interactive visualization results. The platform’s stability and utility have been validated using a public PBMC dataset and it is currently deployed online (https://xulabgdpu.org.cn/Signac.UIO), providing an efficient and user-friendly tool for single-cell epigenomics research.
  • CJBMB: 40 Years of Biochemistry and Molecular Biology in China Special Column of Single-cell and Spatial Omics
    ZHU Xian-Pei, NIU Bin, YANG Jie-Lin
    Chinese Journal of Biochemistry and Molecular Biology. 2025, 41(11): 1590-1599. https://doi.org/10.13865/j.cnki.cjbmb.2025.07.1052
    This review aims to summarize the progress of multi-omics technologies in the study of antibiotic resistance in Cronobacter, with the goal of gaining a deep understanding of its resistance mechanisms and providing a scientific basis for the development of new treatment methods and prevention strategies. By integrating genomics, transcriptomics, proteomics, and metabolomics, the study analyzes gene variations, expression patterns, protein function changes, and metabolic pathway adjustments in Cronobacter. This includes the use of whole-genome sequencing to reveal gene variations related to antibiotic resistance, RNA-seq technology to monitor changes in gene expression patterns, proteomics to study protein expression and function, and metabolomics to analyze dynamic changes in metabolites. The research has found that factors such as biofilm formation and outer membrane proteins significantly affect the antibiotic resistance of Cronobacter. In addition, new potential influencing factors have been identified, including the expression changes of multidrug efflux pump genes, which may play a key role in enhancing antibiotic efflux and reducing intracellular antibiotic concentrations. Multi-omics technologies provide a comprehensive and in-depth perspective for the study of antibiotic resistance in Cronobacter, revealing multiple factors and potential mechanisms that affect resistance. Although some new influencing factors have been identified, their specific molecular mechanisms still require further investigation. The application prospects of multi-omics technologies are broad, and they are expected to provide important support for the development of new treatment methods and prevention strategies.