Clinical application of single-cell RNA sequencing in disease and therapy

Aisha Shigna Nadukkandy , Sowmiya Kalaiselvan , Lin Lin , Yonglun Luo

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (11) : e70512

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (11) : e70512 DOI: 10.1002/ctm2.70512
REVIEW

Clinical application of single-cell RNA sequencing in disease and therapy

Author information +
History +
PDF

Abstract

Background: The emergence of single-cell RNA sequencing (scRNA-seq) technology has revolutionized our capacity to study cell functions in complex tissue microenvironments. Traditional transcriptomic approaches, such as microarrays and bulk RNA sequencing, lacked the resolution to distinguish signals from heterogeneous cell populations or rare cell types, limiting their clinical utility. Since 2009, scRNA-seq has evolved as a new and powerful tool for revisiting somatic evolution and functions under physiological or pathological conditions.

Main Topics Covered: This review focus on elaborating on the clinical applications of scRNA-seq technology, with a particular emphasis on the application of scRNA-seq methods in revisiting the somatic cell evolution in human diseases. We further provide a snapshot of the scRNA-seq applications in biomarker discovery and drug development, current challenges associated with the technology, and future directions.

Conclusions: With the recent progresses in single cell and spatial transcriptome technologies, scRNA-seq enables a deeper understanding of the complexity of human diseases. The integration of AI and machine learning algorithms into big data analysis offers hope for overcoming these hurdles, potentially allowing scRNA-seq and multi-omics approaches to bridge the gap in our understanding of complex biological systems and advances the development of precision medicine.

Keywords

single-cell RNA sequencing / the dawn of a new genome medicine era

Cite this article

Download citation ▾
Aisha Shigna Nadukkandy, Sowmiya Kalaiselvan, Lin Lin, Yonglun Luo. Clinical application of single-cell RNA sequencing in disease and therapy. Clinical and Translational Medicine, 2025, 15(11): e70512 DOI:10.1002/ctm2.70512

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Tang F, Barbacioru C, Wang Y, et al. mRNA-seq whole-transcriptome analysis of a single cell. Nat Methods. 2009; 6: 377-382.

[2]

Ziegenhain C, Vieth B, Parekh S, et al. Comparative analysis of single-cell RNA sequencing methods. Mol Cell. 2017; 65: 631-643. e634.

[3]

Hwang B, Lee JH, Bang D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp Mol Med. 2018; 50: 1-14.

[4]

Jovic D, Liang X, Zeng H, Lin L, Xu F, Luo Y. Single-cell RNA sequencing technologies and applications: a brief overview. Clin Transl Med. 2022; 12: e694.

[5]

Haque A, Engel J, Teichmann SA, Lönnberg T. A practical guide to single-cell RNA-sequencing for biomedical research and clinical applications. Genome Med. 2017; 9: 75.

[6]

Santoro F, Chien KR, Sahara M. Isolation of human ESC-derived cardiac derivatives and embryonic heart cells for population and single-cell RNA-seq analysis. STAR Protoc. 2021; 2: 100339.

[7]

Gladka MM, Molenaar B, de Ruiter H, et al. Single-cell sequencing of the healthy and diseased heart reveals cytoskeleton-associated protein 4 as a new modulator of fibroblasts activation. Circulation. 2018; 138: 166-180.

[8]

Lacar B, Linker SB, Jaeger BN, et al. Nuclear RNA-seq of single neurons reveals molecular signatures of activation. Nat Commun. 2016; 7: 11022.

[9]

Xin Y, Kim J, Ni M, et al. Use of the Fluidigm C1 platform for RNA sequencing of single mouse pancreatic islet cells. Proc Natl Acad Sci U S A. 2016; 113: 3293-3298.

[10]

Nayak R, Hasija Y. A hitchhiker's guide to single-cell transcriptomics and data analysis pipelines. Genomics. 2021; 113: 606-619.

[11]

Picelli S. Single-cell RNA-sequencing: the future of genome biology is now. RNA Biol. 2017; 14: 637-650.

[12]

Hedlund E, Deng Q. Single-cell RNA sequencing: technical advancements and biological applications. Mol Aspects Med. 2018; 59: 36-46.

[13]

Zhu X, Wolfgruber TK, Tasato A, Arisdakessian C, Garmire DG, Garmire LX. Granatum: a graphical single-cell RNA-Seq analysis pipeline for genomics scientists. Genome Med. 2017; 9: 108.

[14]

Lun AT, McCarthy DJ, Marioni JC. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with bioconductor. F1000Res. 2016; 5: 2122.

[15]

Guo M, Wang H, Potter SS, Whitsett JA, Xu Y. SINCERA: a pipeline for single-cell RNA-seq profiling analysis. PLoS Comput Biol. 2015; 11: e1004575.

[16]

Xu C, Su Z. Identification of cell types from single-cell transcriptomes using a novel clustering method. Bioinformatics. 2015; 31: 1974-1980.

[17]

Setty M, Tadmor MD, Reich-Zeliger S, et al. Wishbone identifies bifurcating developmental trajectories from single-cell data. Nat Biotechnol. 2016; 34: 637-645.

[18]

Marco E, Karp RL, Guo G, et al. Bifurcation analysis of single-cell gene expression data reveals epigenetic landscape. Proc Natl Acad Sci U S A. 2014; 111: E5643-5650.

[19]

Vitak SA, Torkenczy KA, Rosenkrantz JL, et al. Sequencing thousands of single-cell genomes with combinatorial indexing. Nat Methods. 2017; 14: 302-308.

[20]

Chen C, Xing D, Tan L, et al. Single-cell whole-genome analyses by linear amplification via transposon insertion (LIANTI). Science. 2017; 356: 189-194.

[21]

Guo F, Li L, Li J, et al. Single-cell multi-omics sequencing of mouse early embryos and embryonic stem cells. Cell Res. 2017; 27: 967-988.

[22]

Casasent AK, Schalck A, Gao R, et al. Multiclonal invasion in breast tumors identified by topographic single cell sequencing. Cell. 2018; 172: 205-217. e212.

[23]

Demaree B, Weisgerber D, Lan F, Abate AR. An ultrahigh-throughput microfluidic platform for single-cell genome sequencing. J Vis Exp. 2018; 135: 57598.

[24]

Han X, Wang R, Zhou Y, et al. Mapping the mouse cell atlas by Microwell-seq. Cell. 2018; 172: 1091-1107 e1017.

[25]

Rosenberg AB, Roco CM, Muscat RA, et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science. 2018; 360: 176-182.

[26]

Van de Sande B, Lee JS, Mutasa-Gottgens E, et al. Applications of single-cell RNA sequencing in drug discovery and development. Nat Rev Drug Discov. 2023; 22: 496-520.

[27]

Osmani Z, Brouwer WP, Grashof DG, et al. Metabolic dysfunction-associated steatohepatitis reduces interferon and macrophage liver gene signatures in patients with chronic hepatitis B. J Hepatol. 2025; 82: 594-603.

[28]

Sin DD. What single cell RNA sequencing has taught us about chronic obstructive pulmonary disease. Tuberc Respir Dis (Seoul). 2024; 87: 252-260.

[29]

Alexander MJ, Budinger GRS, Reyfman PA. Breathing fresh air into respiratory research with single-cell RNA sequencing. Eur Respir Rev. 2020; 29: 200060.

[30]

Plasschaert LW, Žilionis R, Choo-Wing R, et al. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature. 2018; 560: 377-381.

[31]

Reyfman PA, Walter JM, Joshi N, et al. Single-cell transcriptomic analysis of human lung provides insights into the pathobiology of pulmonary fibrosis. Am J Respir Crit Care Med. 2019; 199: 1517-1536.

[32]

Treutlein B, Brownfield DG, Wu AR, et al. Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature. 2014; 509: 371-375.

[33]

Vieira Braga FA, Kar G, Berg M, et al. A cellular census of human lungs identifies novel cell states in health and in asthma. Nat Med. 2019; 25: 1153-1163.

[34]

Levy ML, Bacharier LB, Bateman E, et al. Key recommendations for primary care from the 2022 Global Initiative for Asthma (GINA) update. NPJ Prim Care Respir Med. 2023; 33: 7.

[35]

Kuruvilla ME, Lee FE, Lee GB. Understanding asthma phenotypes, endotypes, and mechanisms of disease. Clin Rev Allergy Immunol. 2019; 56: 219-233.

[36]

Chen A, Diaz-Soto MP, Sanmamed MF, et al. Single-cell characterization of a model of poly I:C-stimulated peripheral blood mononuclear cells in severe asthma. Respir Res. 2021; 22: 122.

[37]

Li H, Wang H, Sokulsky L, et al. Single-cell transcriptomic analysis reveals key immune cell phenotypes in the lungs of patients with asthma exacerbation. J Allergy Clin Immunol. 2021; 147: 941-954.

[38]

Farzan N, Vijverberg SJ, Kabesch M, Sterk PJ, Maitland-van der Zee AH. The use of pharmacogenomics, epigenomics, and transcriptomics to improve childhood asthma management: where do we stand?. Pediatr Pulmonol. 2018; 53: 836-845.

[39]

Modena BD, Bleecker ER, Busse WW, et al. Gene expression correlated with severe asthma characteristics reveals heterogeneous mechanisms of severe disease. Am J Respir Crit Care Med. 2017; 195: 1449-1463.

[40]

Agustí A, Celli BR, Criner GJ, et al. Global Initiative for Chronic Obstructive Lung Disease 2023 report: GOLD executive summary. Eur Respir J. 2023; 61: 2300239.

[41]

Lee Y, Song J, Jeong Y, Choi E, Ahn C, Jang W. Meta-analysis of single-cell RNA-sequencing data for depicting the transcriptomic landscape of chronic obstructive pulmonary disease. Comput Biol Med. 2023; 167: 107685.

[42]

Bahr TM, Hughes GJ, Armstrong M, et al. Peripheral blood mononuclear cell gene expression in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2013; 49: 316-323.

[43]

Tiew PY, Dicker AJ, Keir HR, et al. A high-risk airway mycobiome is associated with frequent exacerbation and mortality in COPD. Eur Respir J. 2021; 57: 2002050.

[44]

Tiew PY, Ko FWS, Narayana JK, et al. “High-risk” clinical and inflammatory clusters in COPD of Chinese descent. Chest. 2020; 158: 145-156.

[45]

Dicker AJ, Huang JT, Lonergan M, et al. The sputum microbiome, airway inflammation, and mortality in chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2021; 147: 158-167.

[46]

Ramsheh MY, Haldar K, Esteve-Codina A, et al. Lung microbiome composition and bronchial epithelial gene expression in patients with COPD versus healthy individuals: a bacterial 16S rRNA gene sequencing and host transcriptomic analysis. Lancet Microbe. 2021; 2: e300-e310.

[47]

Kusko RL, Brothers JF, Tedrow J, et al. Integrated genomics reveals convergent transcriptomic networks underlying chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2016; 194: 948-960.

[48]

Karampitsakos T, Juan-Guardela BM, Tzouvelekis A, Herazo-Maya JD. Precision medicine advances in idiopathic pulmonary fibrosis. EBioMedicine. 2023; 95: 104766.

[49]

Herazo-Maya JD, Noth I, Duncan SR, et al. Peripheral blood mononuclear cell gene expression profiles predict poor outcome in idiopathic pulmonary fibrosis. Sci Transl Med. 2013; 5: 205ra136.

[50]

Kraven LM, Taylor AR, Molyneaux PL, et al. Cluster analysis of transcriptomic datasets to identify endotypes of idiopathic pulmonary fibrosis. Thorax. 2023; 78: 551-558.

[51]

Ruan P, Todd JL, Zhao H, et al. Integrative multi-omics analysis reveals novel idiopathic pulmonary fibrosis endotypes associated with disease progression. Respir Res. 2023; 24: 141.

[52]

Du Y, Zhang J, Guo K, Yin Y. Identification of potential biomarkers for idiopathic pulmonary arterial hypertension using single-cell and bulk RNA sequencing analysis. Front Genet. 2024; 15: 1328234.

[53]

Rajkumar R, Konishi K, Richards TJ, et al. Genomewide RNA expression profiling in lung identifies distinct signatures in idiopathic pulmonary arterial hypertension and secondary pulmonary hypertension. Am J Physiol Heart Circ Physiol. 2010; 298: H1235-H1248.

[54]

Geraci MW, Moore M, Gesell T, et al. Gene expression patterns in the lungs of patients with primary pulmonary hypertension: a gene microarray analysis. Circ Res. 2001; 88: 555-562.

[55]

Hoffmann J, Wilhelm J, Olschewski A, Kwapiszewska G. Microarray analysis in pulmonary hypertension. Eur Respir J. 2016; 48: 229-241.

[56]

Kan M, Shumyatcher M, Himes BE. Using omics approaches to understand pulmonary diseases. Respir Res. 2017; 18: 149.

[57]

Cottam MA, Caslin HL, Winn NC, Hasty AH. Multiomics reveals persistence of obesity-associated immune cell phenotypes in adipose tissue during weight loss and weight regain in mice. Nat Commun. 2022; 13: 2950.

[58]

Schwalie PC, Dong H, Zachara M, et al. A stromal cell population that inhibits adipogenesis in mammalian fat depots. Nature. 2018; 559: 103-108.

[59]

Hepler C, Shan B, Zhang Q, et al. Identification of functionally distinct fibro-inflammatory and adipogenic stromal subpopulations in visceral adipose tissue of adult mice. Elife. 2018; 7: e39636.

[60]

Merrick D, Sakers A, Irgebay Z, et al. Identification of a mesenchymal progenitor cell hierarchy in adipose tissue. Science. 2019; 364: eaav2501.

[61]

Vijay J, Gauthier M, Biswell RL, et al. Single-cell analysis of human adipose tissue identifies depot and disease specific cell types. Nat Metab. 2020; 2: 97-109.

[62]

Sun W, Dong H, Balaz M, et al. snRNA-seq reveals a subpopulation of adipocytes that regulates thermogenesis. Nature. 2020; 587: 98-102.

[63]

Wang Z, Keogh A, Waldt A, et al. Single-cell and bulk transcriptomics of the liver reveals potential targets of NASH with fibrosis. Sci Rep. 2021; 11: 19396.

[64]

Su Q, Kim SY, Adewale F, et al. Single-cell RNA transcriptome landscape of hepatocytes and non-parenchymal cells in healthy and NAFLD mouse liver. iScience. 2021; 24: 103233.

[65]

Park SR, Cho CS, Xi J, Kang HM, Lee JH. Holistic characterization of single-hepatocyte transcriptome responses to high-fat diet. Am J Physiol Endocrinol Metab. 2021; 320: E244-E258.

[66]

Watson BR, Paul B, Rahman RU, et al. Spatial transcriptomics of healthy and fibrotic human liver at single-cell resolution. Nat Commun. 2025; 16: 319.

[67]

Andrews TS, Nakib D, Perciani CT, et al. Single-cell, single-nucleus, and spatial transcriptomics characterization of the immunological landscape in the healthy and PSC human liver. J Hepatol. 2024; 80: 730-743.

[68]

Andrews TS, Atif J, Liu JC, et al. Single-cell, single-nucleus, and spatial RNA sequencing of the human liver identifies cholangiocyte and mesenchymal heterogeneity. Hepatol Commun. 2022; 6: 821-840.

[69]

Lawlor N, George J, Bolisetty M, et al. Single-cell transcriptomes identify human islet cell signatures and reveal cell-type-specific expression changes in type 2 diabetes. Genome Res. 2017; 27: 208-222.

[70]

Li J, Klughammer J, Farlik M, et al. Single-cell transcriptomes reveal characteristic features of human pancreatic islet cell types. EMBO Rep. 2016; 17: 178-187.

[71]

Wang YJ, Schug J, Won K, et al. Single-cell transcriptomics of the human endocrine pancreas. Diabetes. 2016; 65: 3028-3038.

[72]

Segerstolpe Å, Palasantza A, Eliasson P, et al. Single-cell transcriptome profiling of human pancreatic islets in health and type 2 diabetes. Cell Metab. 2016; 24: 593-607.

[73]

Dorajoo R, Ali Y, Tay VSY, et al. Single-cell transcriptomics of East-Asian pancreatic islets cells. Sci Rep. 2017; 7: 5024.

[74]

Wang D, Wang J, Bai L, et al. Long-term expansion of pancreatic islet organoids from resident Procr(+) progenitors. Cell. 2020; 180: 1198-1211. e1119.

[75]

Sona C, Yeh Y, Patsalos A, et al. Evidence of islet CADM1-mediated immune cell interactions during human type 1 diabetes. JCI Insight. 2022; 7: e153136.

[76]

Zakharov PN, Hu H, Wan X, Unanue ER. Single-cell RNA sequencing of murine islets shows high cellular complexity at all stages of autoimmune diabetes. J Exp Med. 2020; 217: e20192362.

[77]

Fukaishi T, Nakagawa Y, Fukunaka A, et al. Characterisation of Ppy-lineage cells clarifies the functional heterogeneity of pancreatic beta cells in mice. Diabetologia. 2021; 64: 2803-2816.

[78]

Muraro M, Dharmadhikari G, Grün D, et al. A single-cell transcriptome atlas of the human pancreas. Cell Syst. 2016; 3: 385-394. e383.

[79]

Zhu AX, Abbas AR, de Galarreta MR, et al. Molecular correlates of clinical response and resistance to atezolizumab in combination with bevacizumab in advanced hepatocellular carcinoma. Nat Med. 2022; 28: 1599-1611.

[80]

Duvall E, Benitez CM, Tellez K, et al. Single-cell transcriptome and accessible chromatin dynamics during endocrine pancreas development. Proc Natl Acad Sci U S A. 2022; 119: e2201267119.

[81]

Khan SU, Huang Y, Ali H, et al. Single-cell RNA Sequencing (scRNA-seq): advances and challenges for cardiovascular diseases (CVDs). Curr Probl Cardiol. 2024; 49: 102202.

[82]

Chen Y, Liu Y, Gao X. The application of single-cell technologies in cardiovascular research. Front Cell Dev Biol. 2021; 9: 751371.

[83]

Miao R, Dong X, Gong J, et al. Cell landscape atlas for patients with chronic thromboembolic pulmonary hypertension after pulmonary endarterectomy constructed using single-cell RNA sequencing. Aging (Albany NY). 2021; 13: 16485-16499.

[84]

Zaragoza MV, Bui T-A, Widyastuti HP, et al. LMNA-related dilated cardiomyopathy: single-cell transcriptomics during patient-derived iPSC differentiation support cell type and lineage-specific dysregulation of gene expression and development for cardiomyocytes and epicardium-derived cells with lamin A/C haploinsufficiency. Cells. 2024; 13: 1479.

[85]

Chaffin M, Papangeli I, Simonson B, et al. Single-nucleus profiling of human dilated and hypertrophic cardiomyopathy. Nature. 2022; 608: 174-180.

[86]

Simonson B, Chaffin M, Hill MC, et al. Single-nucleus RNA sequencing in ischemic cardiomyopathy reveals common transcriptional profile underlying end-stage heart failure. Cell Rep. 2023; 42: 112086.

[87]

Lu J, Ren J, Liu J, et al. High-resolution single-cell transcriptomic survey of cardiomyocytes from patients with hypertrophic cardiomyopathy. Cell Prolif. 2024; 57: e13557.

[88]

Hua X, Hu G, Hu Q, et al. Single-cell RNA sequencing to dissect the immunological network of autoimmune myocarditis. Circulation. 2020; 142: 384-400.

[89]

Hou P, Fang J, Liu Z, et al. Macrophage polarization and metabolism in atherosclerosis. Cell Death Dis. 2023; 14: 691.

[90]

Hu Z, Liu W, Hua X, et al. Single-cell transcriptomic atlas of different human cardiac arteries identifies cell types associated with vascular physiology. Arterioscler Thromb Vasc Biol. 2021; 41: 1408-1427.

[91]

Depuydt MA, Prange KH, Slenders L, et al. Microanatomy of the human atherosclerotic plaque by single-cell transcriptomics. Circ Res. 2020; 127: 1437-1455.

[92]

Roberts ME, Barvalia M, Silva JA, et al. Deep phenotyping by mass cytometry and single-cell RNA-sequencing reveals LYN-regulated signaling profiles underlying monocyte subset heterogeneity and lifespan. Circ Res. 2020; 126: e61-e79.

[93]

Fan X, Zhou Y, Guo X, Xu M. Utilizing single-cell RNA sequencing for analyzing the characteristics of PBMC in patients with Kawasaki disease. BMC Pediatr. 2021; 21: 277.

[94]

Zeng L, Yang K, Zhang T, et al. Research progress of single-cell transcriptome sequencing in autoimmune diseases and autoinflammatory disease: a review. J Autoimmun. 2022; 133: 102919.

[95]

Liu J, Xu T, Jin Y, Huang B, Zhang Y. Progress and clinical application of single-cell transcriptional sequencing technology in cancer research. Front Oncol. 2020; 10: 593085.

[96]

Zhang Y, Narayanan SP, Mannan R, et al. Single-cell analyses of renal cell cancers reveal insights into tumor microenvironment, cell of origin, and therapy response. Proc Natl Acad Sci U S A. 2021; 118: e2103240118.

[97]

Nofech-Mozes I, Soave D, Awadalla P, Abelson S. Pan-cancer classification of single cells in the tumour microenvironment. Nat Commun. 2023; 14: 1615.

[98]

Tirosh I, Suva ML. Cancer cell states: lessons from ten years of single-cell RNA-sequencing of human tumors. Cancer Cell. 2024; 42: 1497-1506.

[99]

Chen S, Jiang W, Du Y, et al. Single-cell analysis technologies for cancer research: from tumor-specific single cell discovery to cancer therapy. Front Genet. 2023; 14: 1276959.

[100]

Eble JA, Niland S. The extracellular matrix in tumor progression and metastasis. Clin Exp Metastasis. 2019; 36: 171-198.

[101]

Pan X, Zhang H, Xu D, et al. Identification of a novel cancer stem cell subpopulation that promotes progression of human fatal renal cell carcinoma by single-cell RNA-seq analysis. Int J Biol Sci. 2020; 16: 3149-3162.

[102]

Davis RT, Blake K, Ma D, et al. Transcriptional diversity and bioenergetic shift in human breast cancer metastasis revealed by single-cell RNA sequencing. Nat Cell Biol. 2020; 22: 310-320.

[103]

Okamoto T, duVerle D, Yaginuma K, et al. Comparative analysis of patient-matched PDOs revealed a reduction in OLFM4-associated clusters in metastatic lesions in colorectal cancer. Stem Cell Rep. 2021; 16: 954-967.

[104]

Jie X, Zhang M, Du M, et al. Detection of circulating tumor cells and evaluation of epithelial-mesenchymal transition patterns of circulating tumor cells in ovarian cancer. Transl Cancer Res. 2022; 11: 2636-2646.

[105]

Stewart CA, Gay CM, Xi Y, et al. Single-cell analyses reveal increased intratumoral heterogeneity after the onset of therapy resistance in small-cell lung cancer. Nat Cancer. 2020; 1: 423-436.

[106]

Miyamoto DT, Zheng Y, Wittner BS, et al. RNA-Seq of single prostate CTCs implicates noncanonical Wnt signaling in antiandrogen resistance. Science. 2015; 349: 1351-1356.

[107]

Marjanovic ND, Hofree M, Chan JE, et al. Emergence of a high-plasticity cell state during lung cancer evolution. Cancer Cell. 2020; 38: 229-246. e213.

[108]

Fan Y, Li Y, Yao X, et al. Epithelial SOX9 drives progression and metastases of gastric adenocarcinoma by promoting immunosuppressive tumour microenvironment. Gut. 2023; 72: 624-637.

[109]

Zhang Z, Wang Z, Chen Y, et al. Integrated analysis of single-cell and bulk RNA sequencing data reveals a pan-cancer stemness signature predicting immunotherapy response. Genome Med. 2022; 14: 45.

[110]

Kumar V, Ramnarayanan K, Sundar R, et al. Single-cell atlas of lineage states, tumor microenvironment, and subtype-specific expression programs in gastric cancer. Cancer Discov. 2022; 12: 670-691.

[111]

Kim N, Kim HK, Lee K, et al. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat Commun. 2020; 11: 2285.

[112]

Song H, Weinstein HNW, Allegakoen P, et al. Single-cell analysis of human primary prostate cancer reveals the heterogeneity of tumor-associated epithelial cell states. Nat Commun. 2022; 13: 141.

[113]

Khodayari S, Khodayari H, Saeedi E, Mahmoodzadeh H, Sadrkhah A, Nayernia K. Single-cell transcriptomics for unlocking personalized cancer immunotherapy: toward targeting the origin of tumor development immunogenicity. Cancers (Basel). 2023; 15: 3615.

[114]

Sade-Feldman M, Yizhak K, Bjorgaard SL, et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell. 2018; 175: 998-1013. e1020.

[115]

Oh DY, Kwek SS, Raju SS, et al. Intratumoral CD4(+) T cells mediate anti-tumor cytotoxicity in human bladder cancer. Cell. 2020; 181: 1612-1625. e1613.

[116]

Yang W, Han B, Chen Y, Geng F. SAAL1, a novel oncogene, is associated with prognosis and immunotherapy in multiple types of cancer. Aging (Albany NY). 2022; 14: 6316-6337.

[117]

Strickler JH, Hanks BA, Khasraw M. Tumor mutational burden as a predictor of immunotherapy response: is more always better?. Clin Cancer Res. 2021; 27: 1236-1241.

[118]

Gromeier M, Brown MC, Zhang G, et al. Very low mutation burden is a feature of inflamed recurrent glioblastomas responsive to cancer immunotherapy. Nat Commun. 2021; 12: 352.

[119]

Lv W, Zeng Y, Li C, et al. Spatial-temporal diversity of extrachromosomal dna shapes urothelial carcinoma evolution and the tumor immune microenvironment. Cancer Discov. 2025: OF1-OF22.

[120]

Deng Q, Han G, Puebla-Osorio N, et al. Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas. Nat Med. 2020; 26: 1878-1887.

[121]

Chung DJ, Shah N, Wu J, et al. Randomized phase II trial of dendritic cell/myeloma fusion vaccine with lenalidomide maintenance after upfront autologous hematopoietic cell transplantation for multiple myeloma: BMT CTN 1401. Clin Cancer Res. 2023; 29: 4784-4796.

[122]

Qazilbash MH, Saini NY, Cha S, et al. A randomized phase 2 trial of idiotype vaccination and adoptive autologous T-cell transfer in patients with multiple myeloma. Blood. 2022; 139: 1289-1301.

[123]

Tian J, Chen JH, Chao SX, et al. Combined PD-1, BRAF and MEK inhibition in BRAF(V600E) colorectal cancer: a phase 2 trial. Nat Med. 2023; 29: 458-466.

[124]

Cascone T, Leung CH, Weissferdt A, et al. Neoadjuvant chemotherapy plus nivolumab with or without ipilimumab in operable non-small cell lung cancer: the phase 2 platform NEOSTAR trial. Nat Med. 2023; 29: 593-604.

[125]

Maron SB, Chatila W, Walch H, et al. Determinants of survival with combined HER2 and PD-1 blockade in metastatic esophagogastric cancer. Clin Cancer Res. 2023; 29: 3633-3640.

[126]

Eschweiler S, Ramírez-Suástegui C, Li Y, et al. Intermittent PI3Kdelta inhibition sustains anti-tumour immunity and curbs irAEs. Nature. 2022; 605: 741-746.

[127]

Wang L, Wang FS, Gershwin ME. Human autoimmune diseases: a comprehensive update. J Intern Med. 2015; 278: 369-395.

[128]

Lee DSW, Rojas OL, Gommerman JL. B cell depletion therapies in autoimmune disease: advances and mechanistic insights. Nat Rev Drug Discov. 2021; 20: 179-199.

[129]

Kalaiselvan S, Rasool MK. The anti-inflammatory effect of triphala in arthritic-induced rats. Pharm Biol. 2015; 53: 51-60.

[130]

Stephenson W, Donlin LT, Butler A, et al. Single-cell RNA-seq of rheumatoid arthritis synovial tissue using low-cost microfluidic instrumentation. Nat Commun. 2018; 9: 791.

[131]

Zhang F, Wei K, Slowikowski K, et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat Immunol. 2019; 20: 928-942.

[132]

Vanlandewijck M, He L, Mäe MA, et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature. 2018; 554: 475-480.

[133]

Wei K, Korsunsky I, Marshall JL, et al. Notch signalling drives synovial fibroblast identity and arthritis pathology. Nature. 2020; 582: 259-264.

[134]

Mizoguchi F, Slowikowski K, Wei K, et al. Functionally distinct disease-associated fibroblast subsets in rheumatoid arthritis. Nat Commun. 2018; 9: 789.

[135]

Kuo D, Ding J, Cohn IS, et al. HBEGF(+) macrophages in rheumatoid arthritis induce fibroblast invasiveness. Sci Transl Med. 2019; 11: eaau8587.

[136]

Alivernini S, MacDonald L, Elmesmari A, et al. Distinct synovial tissue macrophage subsets regulate inflammation and remission in rheumatoid arthritis. Nat Med. 2020; 26: 1295-1306.

[137]

Orange DE, Yao V, Sawicka K, et al. RNA Identification of PRIME cells predicting rheumatoid arthritis flares. N Engl J Med. 2020; 383: 218-228.

[138]

Zeng L, Yang T, Yang K, et al. Efficacy and safety of curcumin and curcuma longa extract in the treatment of arthritis: a systematic review and meta-analysis of randomized controlled trial. Front Immunol. 2022; 13: 891822.

[139]

Guo C, Liu Q, Zong D, et al. Single-cell transcriptome profiling and chromatin accessibility reveal an exhausted regulatory CD4+ T cell subset in systemic lupus erythematosus. Cell Rep. 2022; 41: 111606.

[140]

Der E, Ranabothu S, Suryawanshi H, et al. Single cell RNA sequencing to dissect the molecular heterogeneity in lupus nephritis. JCI Insight. 2017; 2: e93009.

[141]

Nehar-Belaid D, Hong S, Marches R, et al. Mapping systemic lupus erythematosus heterogeneity at the single-cell level. Nat Immunol. 2020; 21: 1094-1106.

[142]

Zeng L, Yang T, Yang K, et al. Curcumin and Curcuma longa extract in the treatment of 10 types of autoimmune diseases: a systematic review and meta-analysis of 31 randomized controlled trials. Front Immunol. 2022; 13: 896476.

[143]

Perez RK, Gordon MG, Subramaniam M, et al. Single-cell RNA-seq reveals cell type-specific molecular and genetic associations to lupus. Science. 2022; 376: eabf1970.

[144]

Raharja A, Mahil SK, Barker JN. Psoriasis: a brief overview. Clin Med (Lond). 2021; 21: 170-173.

[145]

Cheng JB, Sedgewick AJ, Finnegan AI, et al. Transcriptional programming of normal and inflamed human epidermis at single-cell resolution. Cell Rep. 2018; 25: 871-883.

[146]

Srivastava AK, Chand Yadav T, Khera HK, et al. Insights into interplay of immunopathophysiological events and molecular mechanistic cascades in psoriasis and its associated comorbidities. J Autoimmun. 2021; 118: 102614.

[147]

Bielecki P, Riesenfeld SJ, Hütter J, et al. Skin-resident innate lymphoid cells converge on a pathogenic effector state. Nature. 2021; 592: 128-132.

[148]

Grivas A, Grigoriou M, Malissovas N, et al. Combined—whole blood and skin fibroblasts- transcriptomic analysis in psoriatic arthritis reveals molecular signatures of activity, resistance and early response to treatment. Front Immunol. 2022; 13: 964274.

[149]

Liu J. Psoriatic arthritis: epidemiology, diagnosis, and treatment. World J Orthop. 2014; 5: 537-543.

[150]

Abji F, Rasti M, Gómez-Aristizábal A, et al. Proteinase-mediated macrophage signaling in psoriatic arthritis. Front Immunol. 2020; 11: 629726.

[151]

Martini A, Lovell DJ, Albani S, et al. Juvenile idiopathic arthritis. Nat Rev Dis Primers. 2022; 8: 5.

[152]

Julé AM, Hoyt KJ, Wei K, et al. Th1 polarization defines the synovial fluid T cell compartment in oligoarticular juvenile idiopathic arthritis. JCI Insight. 2021; 6: e149185.

[153]

Schulert GS, Pickering AV, Do T, et al. Monocyte and bone marrow macrophage transcriptional phenotypes in systemic juvenile idiopathic arthritis reveal TRIM8 as a mediator of IFN-gamma hyper-responsiveness and risk for macrophage activation syndrome. Ann Rheum Dis. 2021; 80: 617-625.

[154]

Asano Y. Systemic sclerosis. J Dermatol. 2018; 45: 128-138.

[155]

Gaydosik AM, Tabib T, Domsic R, Khanna D, Lafyatis R, Fuschiotti P. Single-cell transcriptome analysis identifies skin-specific T-cell responses in systemic sclerosis. Ann Rheum Dis. 2021; 80: 1453-1460.

[156]

Tabib T, Huang M, Morse N, et al. Myofibroblast transcriptome indicates SFRP2(hi) fibroblast progenitors in systemic sclerosis skin. Nat Commun. 2021; 12: 4384.

[157]

Apostolidis SA, Stifano G, Tabib T, et al. Single cell RNA sequencing identifies HSPG2 and APLNR as markers of endothelial cell injury in systemic sclerosis skin. Front Immunol. 2018; 9: 2191.

[158]

Garcia-Montoya L, Gul H, Emery P. Recent advances in ankylosing spondylitis: understanding the disease and management. F1000Res. 2018; 7: F1000.

[159]

Li Z, Chen S, Cui H, et al. Tenascin-C-mediated suppression of extracellular matrix adhesion force promotes entheseal new bone formation through activation of Hippo signalling in ankylosing spondylitis. Ann Rheum Dis. 2021; 80: 891-902.

[160]

Chiou J, Geusz RJ, Okino M, et al. Interpreting type 1 diabetes risk with genetics and single-cell epigenomics. Nature. 2021; 594: 398-402.

[161]

Honardoost MA, Adinatha A, Schmidt F, et al. Systematic immune cell dysregulation and molecular subtypes revealed by single-cell RNA-seq of subjects with type 1 diabetes. Genome Med. 2024; 16: 45.

[162]

Elmentaite R, Ross AD, Roberts K, et al. Single-cell sequencing of developing human gut reveals transcriptional links to childhood Crohn's disease. Dev Cell. 2020; 55: 771-783. e775.

[163]

Mitsialis V, Wall S, Liu P, et al. Single-cell analyses of colon and blood reveal distinct immune cell signatures of ulcerative colitis and Crohn's disease. Gastroenterology. 2020; 159: 591-608 e510.

[164]

Li G, Zhang B, Hao J, et al. Identification of novel population-specific cell subsets in Chinese ulcerative colitis patients using single-cell RNA sequencing. Cell Mol Gastroenterol Hepatol. 2021; 12: 99-117.

[165]

Devlin JC, Axelrad J, Hine AM, et al. Single-cell transcriptional survey of ileal-anal pouch immune cells from ulcerative colitis patients. Gastroenterology. 2021; 160: 1679-1693.

[166]

Pozojevic J, Spielmann M. Single-cell sequencing in neurodegenerative disorders. Mol Diagn Ther. 2023; 27: 553-561.

[167]

Obeso J, Stamelou M, Goetz C, et al. Past, present, and future of Parkinson's disease: a special essay on the 200th Anniversary of the Shaking Palsy. Mov Disord. 2017; 32: 1264-1310.

[168]

Smajić S, Prada-Medina CA, Landoulsi Z, et al. Single-cell sequencing of human midbrain reveals glial activation and a Parkinson-specific neuronal state. Brain. 2022; 145: 964-978.

[169]

Agarwal D, Sandor C, Volpato V, et al. A single-cell atlas of the human substantia nigra reveals cell-specific pathways associated with neurological disorders. Nat Commun. 2020; 11: 4183.

[170]

La Manno G, Gyllborg D, Codeluppi S, et al. Molecular diversity of midbrain development in mouse, human, and stem cells. Cell. 2016; 167: 566-580 e519.

[171]

Fernandes HJ, Patikas N, Foskolou S, et al. Single-cell transcriptomics of Parkinson's disease human in vitro models reveals dopamine neuron-specific stress responses. Cell Rep. 2020; 33: 108263.

[172]

Welch JD, Kozareva V, Ferreira A, Vanderburg C, Martin C, Macosko EZ. Single-cell multi-omic integration compares and contrasts features of brain cell identity. Cell. 2019; 177: 1873-1887 e1817.

[173]

Lang C, Campbell KR, Ryan BJ, et al. Single-cell sequencing of iPSC-dopamine neurons reconstructs disease progression and identifies HDAC4 as a regulator of Parkinson cell phenotypes. Cell Stem Cell. 2019; 24: 93-106 e106.

[174]

Stocchi F, Bravi D, Emmi A, Antonini A. Parkinson disease therapy: current strategies and future research priorities. Nat Rev Neurol. 2024; 20: 695-707.

[175]

Tabar V, Sarva H, Lozano AM, et al. Phase I trial of hES cell-derived dopaminergic neurons for Parkinson's disease. Nature. 2025; 641: 978-983.

[176]

Sawamoto N, Doi D, Nakanishi E, et al. Phase I/II trial of iPS-cell-derived dopaminergic cells for Parkinson's disease. Nature. 2025; 641: 971-977.

[177]

Wu Y, Beiser AS, Breteler MMB, et al. The changing prevalence and incidence of dementia over time—current evidence. Nat Rev Neurol. 2017; 13: 327-339.

[178]

Mathys H, Davila-Velderrain J, Peng Z, et al. Author correction: single-cell transcriptomic analysis of Alzheimer's disease. Nature. 2019; 571: E1.

[179]

Grubman A, Chew G, Ouyang JF, et al. A single-cell atlas of entorhinal cortex from individuals with Alzheimer's disease reveals cell-type-specific gene expression regulation. Nat Neurosci. 2019; 22: 2087-2097.

[180]

Lau SF, Cao H, Fu AKY, Ip NY. Single-nucleus transcriptome analysis reveals dysregulation of angiogenic endothelial cells and neuroprotective glia in Alzheimer's disease. Proc Natl Acad Sci U S A. 2020; 117: 25800-25809.

[181]

Leng K, Li E, Eser R, et al. Molecular characterization of selectively vulnerable neurons in Alzheimer's disease. Nat Neurosci. 2021; 24: 276-287.

[182]

Morabito S, Miyoshi E, Michael N, et al. Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer's disease. Nat Genet. 2021; 53: 1143-1155.

[183]

Xiong L, Xue L, Du R, et al. Single-cell RNA sequencing reveals B cell-related molecular biomarkers for Alzheimer's disease. Exp Mol Med. 2021; 53: 1888-1901.

[184]

Chen X, Firulyova M, Manis M, et al. Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy. Nature. 2023; 615: 668-677.

[185]

Koutsodendris N, Blumenfeld J, Agrawal A, et al. Neuronal APOE4 removal protects against tau-mediated gliosis, neurodegeneration and myelin deficits. Nat Aging. 2023; 3: 275-296.

[186]

Kukanja P, Langseth CM, Rubio Rodríguez-Kirby LA, et al. Cellular architecture of evolving neuroinflammatory lesions and multiple sclerosis pathology. Cell. 2024; 187: 1990-2009 e1919.

[187]

Cotsapas C, Mitrovic M, Hafler D. Multiple sclerosis. Handb Clin Neurol. 2018; 148: 723-730.

[188]

Pappalardo JL, Zhang L, Pecsok MK, et al. Transcriptomic and clonal characterization of T cells in the human central nervous system. Sci Immunol. 2020; 5: eabb8786.

[189]

Aoyama S, Mori M, Uzawa A, et al. Serum anti-John Cunningham virus antibody seroprevalence and index among Japanese patients with neuromyelitis optica spectrum disorders. Mult Scler. 2020; 26: 128-129.

[190]

Masuda T, Sankowski R, Staszewski O, et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature. 2019; 566: 388-392.

[191]

Clark IC, Gutiérrez-Vázquez C, Wheeler MA, et al. Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science. 2021; 372: eabf1230.

[192]

Homberger C, Hayward RJ, Barquist L, Vogel J. Improved bacterial single-cell RNA-Seq through automated MATQ-Seq and Cas9-based removal of rRNA reads. mBio. 2023; 14: e03557-03522.

[193]

Imdahl F, Vafadarnejad E, Homberger C, Saliba A, Vogel J. Single-cell RNA-sequencing reports growth-condition-specific global transcriptomes of individual bacteria. Nat Microbiol. 2020; 5: 1202-1206.

[194]

Nishimura M, Takeyama H, Hosokawa M. Enhancing the sensitivity of bacterial single-cell RNA sequencing using RamDA-seq and Cas9-based rRNA depletion. J Biosci Bioeng. 2023; 136: 152-158.

[195]

Blattman SB, Jiang W, Oikonomou P, Tavazoie S. Prokaryotic single-cell RNA sequencing by in situ combinatorial indexing. Nat Microbiol. 2020; 5: 1192-1201.

[196]

Ma P, Amemiya HM, He LL, et al. Bacterial droplet-based single-cell RNA-seq reveals antibiotic-associated heterogeneous cellular states. Cell. 2023; 186: 877-891 e814.

[197]

McNulty R, Sritharan D, Pahng SH, et al. Probe-based bacterial single-cell RNA sequencing predicts toxin regulation. Nat Microbiol. 2023; 8: 934-945.

[198]

Wang B, Lin AE, Yuan J, et al. Single-cell massively-parallel multiplexed microbial sequencing (M3-seq) identifies rare bacterial populations and profiles phage infection. Nat Microbiol. 2023; 8: 1846-1862.

[199]

Xu Z, Wang Y, Sheng K, et al. Droplet-based high-throughput single microbe RNA sequencing by smRandom-seq. Nat Commun. 2023; 14: 5130.

[200]

Imdahl F, Saliba A. Advances and challenges in single-cell RNA-seq of microbial communities. Curr Opin Microbiol. 2020; 57: 102-110.

[201]

Homberger C, Barquist L, Vogel J. Ushering in a new era of single-cell transcriptomics in bacteria. microLife. 2022; 3: uqac020.

[202]

Brennan MA, Rosenthal AZ. Single-cell RNA sequencing elucidates the structure and organization of microbial communities. Front Microbiol. 2021; 12: 713128.

[203]

Nishimura M, Takahashi K, Hosokawa M. Recent advances in single-cell RNA sequencing of bacteria: techniques, challenges, and applications. J Biosci Bioeng. 2025; 139: 341-346.

[204]

Yadav S, Mehta P, Soni J, et al. Single-cell RNA-Seq reveals intracellular microbial diversity within immune cells during SARS-CoV-2 infection and recovery. iScience. 2023; 26: 108357.

[205]

Ghaddar B, Biswas A, Harris C, et al. Tumor microbiome links cellular programs and immunity in pancreatic cancer. Cancer Cell. 2022; 40: 1240-1253 e1245.

[206]

Wu Y, Zhuang J, Song Y, Gao X, Chu J, Han S. Advances in single-cell sequencing technology in microbiome research. Genes Dis. 2024; 11: 101129.

[207]

Avital G, Avraham R, Fan A, Hashimshony T, Hung DT, Yanai I. scDual-Seq: mapping the gene regulatory program of Salmonella infection by host and pathogen single-cell RNA-sequencing. Genome Biol. 2017; 18: 200.

[208]

Cai Y, Dai Y, Wang Y, et al. Single-cell transcriptomics of blood reveals a natural killer cell subset depletion in tuberculosis. EBioMedicine. 2020; 53: 102686.

[209]

Kazer SW, Aicher TP, Muema DM, et al. Integrated single-cell analysis of multicellular immune dynamics during hyperacute HIV-1 infection. Nat Med. 2020; 26: 511-518.

[210]

Avraham R, Haseley N, Brown D, et al. Pathogen cell-to-cell variability drives heterogeneity in host immune responses. Cell. 2015; 162: 1309-1321.

[211]

Lin WN, Tay MZ, Lu R, Liu Y, Chen C, Cheow LF. The role of single-cell technology in the study and control of infectious diseases. Cells. 2020; 9: 1440.

[212]

Golumbeanu M, Cristinelli S, Rato S, et al. Single-cell RNA-seq reveals transcriptional heterogeneity in latent and reactivated HIV-infected cells. Cell Rep. 2018; 23: 942-950.

[213]

Nowakowski T, Pollen A, Di Lullo E, Sandoval-Espinosa C, Bershteyn M, Kriegstein A. Expression analysis highlights AXL as a candidate Zika virus entry receptor in neural stem cells. Cell Stem Cell. 2016; 18: 591-596.

[214]

Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020; 181: 271-280 e278.

[215]

Walls AC, Park Y, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell. 2020; 181: 281-292 e286.

[216]

Hoffmann M, Kleine-Weber H, Pöhlmann S. A multibasic cleavage site in the spike protein of SARS-CoV-2 is essential for infection of human lung cells. Mol Cell. 2020; 78: 779-784. e775.

[217]

Walls AC, Park Y, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell. 2020; 181: 281-292 e286.

[218]

Ziegler CG, Allon SJ, Nyquist SK, et al. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell. 2020; 181: 1016-1035 e1019.

[219]

Wu X, Yang X, Dai Y, et al. Single-cell sequencing to multi-omics: technologies and applications. Biomark Res. 2024; 12: 110.

[220]

Galeano Niño JL, Wu H, LaCourse KD, et al. Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature. 2022; 611: 810-817.

[221]

Green ED, Watson JD, Collins FS. Human genome project: twenty-five years of big biology. Nature. 2015; 526: 29-31.

[222]

Lareau CA, Parker KR, Satpathy AT. Charting the tumor antigen maps drawn by single-cell genomics. Cancer Cell. 2021; 39: 1553-1557.

[223]

Jagadeesh KA, Dey KK, Montoro DT, et al. Identifying disease-critical cell types and cellular processes by integrating single-cell RNA-sequencing and human genetics. Nat Genet. 2022; 54: 1479-1492.

[224]

Muslu O, Hoyt CT, Lacerda M, Hofmann-Apitius M, Frohlich HG. Prioritization of novel therapeutic targets with network representation learning. IEEE/ACM Trans Comput Biol Bioinform. 2022; 19: 491-500.

[225]

Gawel DR, Serra-Musach J, Lilja S, et al. A validated single-cell-based strategy to identify diagnostic and therapeutic targets in complex diseases. Genome Med. 2019; 11: 47.

[226]

Shifrut E, Carnevale J, Tobin V, et al. Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function. Cell. 2018; 175: 1958-1971. e1915.

[227]

Kinker GS, Greenwald AC, Tal R, et al. Pan-cancer single-cell RNA-seq identifies recurring programs of cellular heterogeneity. Nat Genet. 2020; 52: 1208-1218.

[228]

Shinozawa T, Kimura M, Cai Y, et al. High-fidelity drug-induced liver injury screen using human pluripotent stem cell-derived organoids. Gastroenterology. 2021; 160: 831-846 e810.

[229]

Krieger TG, Le Blanc S, Jabs J, et al. Single-cell analysis of patient-derived PDAC organoids reveals cell state heterogeneity and a conserved developmental hierarchy. Nat Commun. 2021; 12: 5826.

[230]

Iorio F, Knijnenburg TA, Vis DJ, et al. A landscape of pharmacogenomic interactions in cancer. Cell. 2016; 166: 740-754.

[231]

Wu L, Wen Y, Leng D, et al. Machine learning methods, databases and tools for drug combination prediction. Brief Bioinform. 2022; 23: bbab355.

[232]

Adam G, Rampášek L, Safikhani Z, Smirnov P, Haibe-Kains B, Goldenberg A. Machine learning approaches to drug response prediction: challenges and recent progress. NPJ Precis Oncol. 2020; 4: 19.

[233]

Hsieh C, Wen J, Lin S, et al. scDrug: from single-cell RNA-seq to drug response prediction. Comput Struct Biotechnol J. 2023; 21: 150-157.

[234]

Chen J, Wang X, Ma A, et al. Deep transfer learning of cancer drug responses by integrating bulk and single-cell RNA-seq data. Nat Commun. 2022; 13: 6494.

[235]

Fustero-Torre C, Jiménez-Santos MJ, García-Martín S, et al. Beyondcell: targeting cancer therapeutic heterogeneity in single-cell RNA-seq data. Genome Med. 2021; 13: 187.

[236]

Pellecchia S, Viscido G, Franchini M, Gambardella G. Predicting drug response from single-cell expression profiles of tumours. BMC Med. 2023; 21: 476.

[237]

Martin JC, Chang C, Boschetti G, et al. Single-cell analysis of Crohn's disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy. Cell. 2019; 178: 1493-1508. e1420.

[238]

Smillie CS, Biton M, Ordovas-Montanes J, et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell. 2019; 178: 714-730 e722.

[239]

Ediriwickrema A, Aleshin A, Reiter JG, et al. Single-cell mutational profiling enhances the clinical evaluation of AML MRD. Blood Adv. 2020; 4: 943-952.

[240]

Oren Y, Tsabar M, Cuoco MS, et al. Cycling cancer persister cells arise from lineages with distinct programs. Nature. 2021; 596: 576-582.

[241]

Rozenblatt-Rosen O, Regev A, Oberdoerffer P, et al. The human tumor atlas network: charting tumor transitions across space and time at single-cell resolution. Cell. 2020; 181: 236-249.

[242]

Aissa AF, Islam ABMMK, Ariss MM, et al. Single-cell transcriptional changes associated with drug tolerance and response to combination therapies in cancer. Nat Commun. 2021; 12: 1628.

[243]

Leader AM, Grout JA, Maier BB, et al. Single-cell analysis of human non-small cell lung cancer lesions refines tumor classification and patient stratification. Cancer Cell. 2021; 39: 1594-1609 e1512.

[244]

Färkkilä A, Gulhan DC, Casado J, et al. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer. Nat Commun. 2020; 11: 1459.

[245]

Zhang K, Erkan EP, Jamalzadeh S, et al. Longitudinal single-cell RNA-seq analysis reveals stress-promoted chemoresistance in metastatic ovarian cancer. Sci Adv. 2022; 8: eabm1831.

[246]

Vanhersecke L, Brunet M, Guégan J, et al. Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression. Nat Cancer. 2021; 2: 794-802.

[247]

Andrews TS, Hemberg M. False signals induced by single-cell imputation. F1000Res. 2018; 7: 1740.

[248]

Ascension AM, Arauzo-Bravo MJ, Izeta A. Challenges and opportunities for the translation of single-cell RNA sequencing technologies to dermatology. Life (Basel). 2022; 12: 67.

[249]

Heumos L, Schaar AC, Lance C, et al. Best practices for single-cell analysis across modalities. Nat Rev Genet. 2023; 24: 550-572.

[250]

Regev A, Teichmann SA, Lander ES, et al. The human cell atlas. Elife. 2017; 6: e27041.

[251]

Han X, Zhou Z, Fei L, et al. Construction of a human cell landscape at single-cell level. Nature. 2020; 581: 303-309.

[252]

Penter L, Borji M, Nagler A, et al. Integrative genotyping of cancer and immune phenotypes by long-read sequencing. Nat Commun. 2024; 15: 32.

[253]

Ding J, Sharon N, Bar-Joseph Z. Temporal modelling using single-cell transcriptomics. Nat Rev Genet. 2022; 23: 355-368.

[254]

La Manno G, Soldatov R, Zeisel A, et al. RNA velocity of single cells. Nature. 2018; 560: 494-498.

[255]

Bassez A, Vos H, Van Dyck L, et al. A single-cell map of intratumoral changes during anti-PD1 treatment of patients with breast cancer. Nat Med. 2021; 27: 820-832.

[256]

Erhard F, Baptista MAP, Krammer T, et al. scSLAM-seq reveals core features of transcription dynamics in single cells. Nature. 2019; 571: 419-423.

[257]

Hendriks G, Jung LA, Larsson AJM, et al. NASC-seq monitors RNA synthesis in single cells. Nat Commun. 2019; 10: 3138.

[258]

Qiu Q, Hu P, Qiu X, Govek KW, Cámara PG, Wu H. Massively parallel and time-resolved RNA sequencing in single cells with scNT-seq. Nat Methods. 2020; 17: 991-1001.

[259]

Byrnes LE, Wong DM, Subramaniam M, et al. Lineage dynamics of murine pancreatic development at single-cell resolution. Nat Commun. 2018; 9: 3922.

[260]

McKenna A, Findlay GM, Gagnon JA, Horwitz MS, Schier AF, Shendure J. Whole-organism lineage tracing by combinatorial and cumulative genome editing. Science. 2016; 353: aaf7907.

[261]

Chen W, Guillaume-Gentil O, Rainer PY, et al. Live-seq enables temporal transcriptomic recording of single cells. Nature. 2022; 608: 733-740.

[262]

Zeng Q, Mousa M, Nadukkandy AS, et al. Understanding tumour endothelial cell heterogeneity and function from single-cell omics. Nat Rev Cancer. 2023; 23: 544-564.

[263]

Cheng M, Jiang Y, Xu J, et al. Spatially resolved transcriptomics: a comprehensive review of their technological advances, applications, and challenges. J Genet Genomics. 2023; 50: 625-640.

[264]

Xia C, Fan J, Emanuel G, Hao J, Zhuang X. Spatial transcriptome profiling by MERFISH reveals subcellular RNA compartmentalization and cell cycle-dependent gene expression. Proc Natl Acad Sci U S A. 2019; 116: 19490-19499.

[265]

Eng CL, Lawson M, Zhu Q, et al. Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH. Nature. 2019; 568: 235-239.

[266]

Vickovic S, Eraslan G, Salmén F, et al. High-definition spatial transcriptomics for in situ tissue profiling. Nat Methods. 2019; 16: 987-990.

[267]

Chen A, Liao S, Cheng M, et al. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell. 2022; 185: 1777-1792 e1721.

[268]

Zhang G, Ji P, Xia P, et al. Identification and targeting of cancer-associated fibroblast signature genes for prognosis and therapy in Cutaneous melanoma. Comput Biol Med. 2023; 167: 107597.

[269]

Cassier PA, Navaridas R, Bellina M, et al. Netrin-1 blockade inhibits tumour growth and EMT features in endometrial cancer. Nature. 2023; 620: 409-416.

[270]

Garrido-Trigo A, Corraliza AM, Veny M, et al. Macrophage and neutrophil heterogeneity at single-cell spatial resolution in human inflammatory bowel disease. Nat Commun. 2023; 14: 4506.

[271]

Gui C, Wei J, Mo C, et al. Therapeutic implications for localized prostate cancer by multiomics analyses of the ageing microenvironment landscape. Int J Biol Sci. 2023; 19: 3951-3969.

[272]

Xie B, Gao D, Zhou B, Chen S, Wang L. New discoveries in the field of metabolism by applying single-cell and spatial omics. J Pharm Anal. 2023; 13: 711-725.

[273]

Dorgau B, Collin J, Rozanska A, et al. Deciphering the spatiotemporal transcriptional and chromatin accessibility of human retinal organoid development at the single-cell level. iScience. 2024; 27: 109397.

[274]

Wahle P, Brancati G, Harmel C, et al. Multimodal spatiotemporal phenotyping of human retinal organoid development. Nat Biotechnol. 2023; 41: 1765-1775.

[275]

Brorson J, Lin L, Wang J, et al. Complementing muscle regeneration-fibro-adipogenic progenitor and macrophage-mediated repair of elderly human skeletal muscle. Nat Commun. 2025; 16: 5233.

[276]

Cao J, Li C, Cui Z, et al. Spatial transcriptomics: a powerful tool in disease understanding and drug discovery. Theranostics. 2024; 14: 2946-2968.

[277]

Nativio R, Lan Y, Donahue G, et al. An integrated multi-omics approach identifies epigenetic alterations associated with Alzheimer's disease. Nat Genet. 2020; 52: 1024-1035.

[278]

Finotello F, Eduati F. Multi-omics profiling of the tumor microenvironment: paving the way to precision immuno-oncology. Front Oncol. 2018; 8: 430.

[279]

Najm P, Zhao P, Steklov M, et al. Loss-of-function mutations in TRAF7 and KLF4 cooperatively activate RAS-like GTPase signaling and promote meningioma development. Cancer Res. 2021; 81: 4218-4229.

[280]

Dixit A, Parnas O, Li B, et al. Perturb-Seq: dissecting molecular circuits with scalable single-cell rna profiling of pooled genetic screens. Cell. 2016; 167: 1853-1866 e1817.

[281]

Datlinger P, Rendeiro AF, Schmidl C, et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat Methods. 2017; 14: 297-301.

[282]

Jaitin DA, Weiner A, Yofe I, et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell. 2016; 167: 1883-1896 e1815.

[283]

Papalexi E, Mimitou EP, Butler AW, et al. Characterizing the molecular regulation of inhibitory immune checkpoints with multimodal single-cell screens. Nat Genet. 2021; 53: 322-331.

[284]

Duan B, Zhou C, Zhu C, et al. Model-based understanding of single-cell CRISPR screening. Nat Commun. 2019; 10: 2233.

[285]

Yang L, Zhu Y, Yu H, et al. scMAGeCK links genotypes with multiple phenotypes in single-cell CRISPR screens. Genome Biol. 2020; 21: 19.

[286]

Singer J, Kuipers J, Jahn K, Beerenwinkel N. Single-cell mutation identification via phylogenetic inference. Nat Commun. 2018; 9: 5144.

[287]

Wang R, Lin DY, Jiang Y. SCOPE: a normalization and copy-number estimation method for single-cell DNA sequencing. Cell Syst. 2020; 10: 445-452 e446.

[288]

Mallory XF, Edrisi M, Navin N, Nakhleh L. Methods for copy number aberration detection from single-cell DNA-sequencing data. Genome Biol. 2020; 21: 208.

[289]

Gao R, Bai S, Henderson YC, et al. Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes. Nat Biotechnol. 2021; 39: 599-608.

[290]

Patel AP, Tirosh I, Trombetta JJ, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014; 344: 1396-1401.

[291]

Petti AA, Williams SR, Miller CA, et al. A general approach for detecting expressed mutations in AML cells using single cell RNA-sequencing. Nat Commun. 2019; 10: 3660.

[292]

Vu TN, Nguyen H, Calza S, Kalari KR, Wang L, Pawitan Y. Cell-level somatic mutation detection from single-cell RNA sequencing. Bioinformatics. 2019; 35: 4679-4687.

[293]

Stubbington MJT, Lönnberg T, Proserpio V, et al. T cell fate and clonality inference from single-cell transcriptomes. Nat Methods. 2016; 13: 329-332.

[294]

Borcherding N, Bormann NL. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res. 2020; 9: 47.

[295]

Zhang Z, Xiong D, Wang X, Liu H, Wang T. Mapping the functional landscape of T cell receptor repertoires by single-T cell transcriptomics. Nat Methods. 2021; 18: 92-99.

[296]

Buenrostro JD, Wu B, Litzenburger UM, et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature. 2015; 523: 486-490.

[297]

Clark SJ, Smallwood SA, Lee HJ, Krueger F, Reik W, Kelsey G. Genome-wide base-resolution mapping of DNA methylation in single cells using single-cell bisulfite sequencing (scBS-seq). Nat Protoc. 2017; 12: 534-547.

[298]

Grosselin K, Durand A, Marsolier J, et al. High-throughput single-cell ChIP-seq identifies heterogeneity of chromatin states in breast cancer. Nat Genet. 2019; 51: 1060-1066.

[299]

Slavov N. Learning from natural variation across the proteomes of single cells. PLoS Biol. 2022; 20: e3001512.

[300]

Brinkerhoff H, Kang ASW, Liu J, Aksimentiev A, Dekker C. Multiple rereads of single proteins at single-amino acid resolution using nanopores. Science. 2021; 374: 1509-1513.

[301]

Hücker SM, Fehlmann T, Werno C, et al. Single-cell microRNA sequencing method comparison and application to cell lines and circulating lung tumor cells. Nat Commun. 2021; 12: 4316.

[302]

Seydel C. Single-cell metabolomics hits its stride. Nat Methods. 2021; 18: 1452-1456.

[303]

Zhang R, Zhou T, Ma J. Multiscale and integrative single-cell Hi-C analysis with Higashi. Nat Biotechnol. 2022; 40: 254-261.

[304]

Arrastia MV, Jachowicz JW, Ollikainen N, et al. Single-cell measurement of higher-order 3D genome organization with scSPRITE. Nat Biotechnol. 2022; 40: 64-73.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

7

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/