Analysis of Tumor-Infiltrating T-Cell Transcriptomes Reveal a Unique Genetic Signature across Different Types of Cancer
Abstract
:1. Introduction
2. Results
2.1. Profiling of Tissue-Infiltrating T-Cells from Different Types of Cancer
2.2. GO Annotations and Biological Pathways in T-Cells from Malignant and Nonmalignant Cancer
2.3. Exclusive Biological Pathways in T-Cells from Different Types of Cancer
2.4. Reactome Pathways in T-Cells from Different Types of Cancer
2.5. Validation of Biological Pathways Using Proteomics and RNA-seq Data
3. Discussion
4. Materials and Methods
4.1. Data Collection and Preprocessing
4.2. T-Cell Identification
4.3. Analysis of T-Cells Subpopulations
4.4. Proteomic Experiments
4.5. Analysis of RNA-seq
4.6. Pathways and GO Categories Analysis
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Tartey, S.; Takeuchi, O. Pathogen recognition and Toll-like receptor targeted therapeutics in innate immune cells. Int. Rev. Immunol. 2017, 36, 57–73. [Google Scholar] [CrossRef] [PubMed]
- McLane, L.M.; Abdel-Hakeem, M.S.; Wherry, E.J. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 2019, 37, 457–495. [Google Scholar] [CrossRef] [PubMed]
- Finotello, F.; Trajanoski, Z. New strategies for cancer immunotherapy: Targeting regulatory T cells. Genome Med. 2017, 9, 10. [Google Scholar] [CrossRef]
- Kumar, B.V.; Connors, T.J.; Farber, D.L. Human T cell development, localization, and function throughout life. Immunity 2018, 48, 202–213. [Google Scholar] [CrossRef] [PubMed]
- Luckheeram, R.V.; Zhou, R.; Verma, A.D.; Xia, B. CD4+ T cells: Differentiation and functions. Clin. Dev. Immunol. 2012, 2012, 925135. [Google Scholar] [CrossRef] [PubMed]
- Halim, L.; Romano, M.; McGregor, R.; Correa, I.; Pavlidis, P.; Grageda, N.; Hoong, S.J.; Yuksel, M.; Jassem, W.; Hannen, R.F.; et al. An atlas of human regulatory T helper-like cells reveals features of Th2-like Tregs that support a tumorigenic environment. Cell Rep. 2017, 20, 757–770. [Google Scholar] [CrossRef] [PubMed]
- Ostroumov, D.; Fekete-Drimusz, N.; Saborowski, M.; Kühnel, F.; Woller, N. CD4 and CD8 T lymphocyte interplay in controlling tumor growth. Cell. Mol. Life Sci. 2018, 75, 689–713. [Google Scholar] [CrossRef]
- Lee, H.L.; Jang, J.W.; Lee, S.W.; Yoo, S.H.; Kwon, J.H.; Nam, S.W.; Bae, S.H.; Choi, J.Y.; Han, N.I.; Yoon, S.K. Inflammatory cytokines and change of Th1/Th2 balance as prognostic indicators for hepatocellular carcinoma in patients treated with transarterial chemoembolization. Sci. Rep. 2019, 9, 3260. [Google Scholar] [CrossRef]
- Budhu, A.; Forgues, M.; Ye, Q.H.; Jia, H.L.; He, P.; Zanetti, K.A.; Kammula, U.S.; Chen, Y.; Qin, L.X.; Tang, Z.Y.; et al. Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell 2006, 10, 99–111. [Google Scholar] [CrossRef]
- Rabinovich, G.A.; Gabrilovich, D.; Sotomayor, E.M. Immunosuppressive strategies that are mediated by tumor cells. Annu. Rev. Immunol. 2007, 25, 267–296. [Google Scholar] [CrossRef] [Green Version]
- Gun, S.Y.; Lee, S.W.; Sieow, J.L.; Wong, S.C. Targeting immune cells for cancer therapy. Redox Biol. 2019, 25, 101174. [Google Scholar] [CrossRef] [PubMed]
- Iellem, A.; Mariani, M.; Lang, R.; Recalde, H.; Panina-Bordignon, P.; Sinigaglia, F.; D’Ambrosio, D. Unique chemotactic response profile and specific expression of chemokine receptors CCR4 and CCR8 by CD4+ CD25+ regulatory T cells. J. Exp. Med. 2001, 194, 847–854. [Google Scholar] [CrossRef]
- Łuksza, M.; Riaz, N.; Makarov, V.; Balachandran, V.P.; Hellmann, M.D.; Solovyov, A.; Rizvi, N.A.; Merghoub, T.; Levine, A.J.; Chan, T.A.; et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017, 551, 517–520. [Google Scholar] [CrossRef] [PubMed]
- Suvas, S.; Azkur, A.K.; Kim, B.S.; Kumaraguru, U.; Rouse, B.T. CD4+ CD25+ regulatory T cells control the severity of viral immunoinflammatory lesions. J. Immunol. 2004, 172, 4123–4132. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim, S.F.; Engh, G.V. Flow cytometry and cell sorting. Cell Separat. 2007, 106, 19–39. [Google Scholar]
- Santegoets, S.J.; Dijkgraaf, E.M.; Battaglia, A.; Beckhove, P.; Britten, C.M.; Gallimore, A.; Godkin, A.; Gouttefangeas, C.; de Gruijl, T.D.; Koenen, H.J.; et al. Monitoring regulatory T cells in clinical samples: Consensus on an essential marker set and gating strategy for regulatory T cell analysis by flow cytometry. Cancer Immunol. Immunother. 2015, 64, 1271–1286. [Google Scholar] [CrossRef] [PubMed]
- Radford, F.; Tyagi, S.; Gennaro, M.L.; Pine, R.; Bushkin, Y. Flow cytometric characterization of antigen-specific T cells based on RNA and its advantages in detecting infections and immunological disorders. Crit. Rev. Immunol. 2016, 36, 359–378. [Google Scholar] [CrossRef] [PubMed]
- Gawad, C.; Koh, W.; Quake, S.R. Single-cell genome sequencing: Current state of the science. Nat. Rev. Genet. 2016, 17, 175–188. [Google Scholar] [CrossRef]
- Tang, F.; Barbacioru, C.; Wang, Y.; Nordman, E.; Lee, C.; Xu, N.; Wang, X.; Bodeau, J.; Tuch, B.B.; Siddiqui, A.; et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat. Methods 2009, 6, 377–382. [Google Scholar] [CrossRef]
- Svensson, V.; Natarajan, K.N.; Ly, L.H.; Miragaia, R.J.; Labalette, C.; Macaulay, I.C.; Cvejic, A.; Teichmann, S.A. Power analysis of single-cell RNA-sequencing experiments. Nat. Methods 2017, 14, 381–387. [Google Scholar] [CrossRef]
- Kiselev, V.Y.; Andrews, T.S.; Hemberg, M. Challenges in unsupervised clustering of single-cell RNA-seq data. Nat. Rev. Genet. 2019, 20, 273–282. [Google Scholar] [CrossRef] [PubMed]
- Tirosh, I.; Izar, B.; Prakadan, S.M.; Wadsworth, M.H.; Treacy, D.; Trombetta, J.J.; Rotem, A.; Rodman, C.; Lian, C.; Murphy, G.; et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 2016, 352, 189–196. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; van der Leun, A.M.; Yofe, I.; Lubling, Y.; Gelbard-Solodkin, D.; van Akkooi, A.C.; van den Braber, M.; Rozeman, E.A.; Haanen, J.B.; Blank, C.U.; et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 2019, 176, 775–789. [Google Scholar] [CrossRef]
- Azizi, E.; Carr, A.J.; Plitas, G.; Cornish, A.E.; Konopacki, C.; Prabhakaran, S.; Nainys, J.; Wu, K.; Kiseliovas, V.; Setty, M.; et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 2018, 174, 1293–1308. [Google Scholar] [CrossRef]
- Chung, W.; Eum, H.H.; Lee, H.O.; Lee, K.M.; Lee, H.B.; Kim, K.T.; Ryu, H.S.; Kim, S.; Lee, J.E.; Park, Y.H.; et al. Single-cell RNA-seq enables comprehensive tumour and immune cell profiling in primary breast cancer. Nat. Commun. 2017, 8, 15081. [Google Scholar] [CrossRef] [PubMed]
- Szabo, P.A.; Levitin, H.M.; Miron, M.; Snyder, M.E.; Senda, T.; Yuan, J.; Cheng, Y.L.; Bush, E.C.; Dogra, P.; Thapa, P.; et al. A single-cell reference map for human blood and tissue T cell activation reveals functional states in health and disease. bioRxiv 2019, 555557. [Google Scholar] [CrossRef]
- Guo, X.; Zhang, Y.; Zheng, L.; Zheng, C.; Song, J.; Zhang, Q.; Kang, B.; Liu, Z.; Jin, L.; Xing, R.; et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 2018, 24, 978–985. [Google Scholar] [CrossRef]
- Zhang, L.; Yu, X.; Zheng, L.; Zhang, Y.; Li, Y.; Fang, Q.; Gao, R.; Kang, B.; Zhang, Q.; Huang, J.Y.; et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 2018, 564, 268–272. [Google Scholar] [CrossRef]
- Puram, S.V.; Tirosh, I.; Parikh, A.S.; Patel, A.P.; Yizhak, K.; Gillespie, S.; Rodman, C.; Luo, C.L.; Mroz, E.A.; Emerick, K.S.; et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 2017, 171, 1611–1624. [Google Scholar] [CrossRef]
- Ashburner, M.; Ball, C.A.; Blake, J.A.; Botstein, D.; Butler, H.; Cherry, J.M.; Davis, A.P.; Dolinski, K.; Dwight, S.S.; Eppig, J.T.; et al. Gene ontology: Tool for the unification of biology. Nat. Genet. 2000, 25, 25–29. [Google Scholar] [CrossRef]
- Gene Ontology Consortium. The gene ontology resource: 20 years and still GOing strong. Nucleic Acids Res. 2019, 47, D330–D338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bijay, J.; Matthews, L.; Viteri, G.; Gong, C.; Lorente, P.; Fabregat, A.; Sidiropoulos, K.; Cook, J.; Gillespie, M.; Haw, R.; et al. The reactome pathway knowledgebase. Nucleic Acids Res. 2020, 48, D498–D503. [Google Scholar]
- Pedregosa, F.; Varoquaux, G.; Gramfort, A.; Michel, V.; Thirion, B.; Grisel, O.; Blondel, M.; Prettenhofer, P.; Weiss, R.; Dubourg, V.; et al. Scikit-learn: Machine learning in Python. J. Mach. Learn. Res. 2011, 12, 2825–2830. [Google Scholar]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed]
- Ott, P.A.; Hodi, F.S.; Robert, C. CTLA-4 and PD-1/PD-L1 blockade: New immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin. Cancer Res. 2013, 19, 5300–5309. [Google Scholar] [CrossRef]
- Paul, S.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13, 2498–2504. [Google Scholar]
- Gabriela, B.; Mlecnik, B.; Hackl, H.; Charoentong, P.; Tosolini, M.; Kirilovsky, A.; Fridman, W.; Pagès, F.; Trajanoski, Z.; Galon, J. ClueGO: A Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 2009, 25, 1091–1093. [Google Scholar]
- Zhang, S. Comparisons of gene coexpression network modules in breast cancer and ovarian cancer. BMC Syst. Biol. 2018, 12, 75–87. [Google Scholar] [CrossRef]
- Emmert-Streib, F.; de Matos Simoes, R.; Glazko, G.; McDade, S.; Haibe-Kains, B.; Holzinger, A.; Dehmer, M.; Campbell, F.C. Functional and genetic analysis of the colon cancer network. BMC Bioinform. 2014, 15, S6. [Google Scholar] [CrossRef]
- Knutson, K.L.; Disis, M.L. Tumor antigen-specific T helper cells in cancer immunity and immunotherapy. Cancer Immunol. Immunother. 2005, 54, 721–728. [Google Scholar] [CrossRef]
- Marco, F.; Yáñez, M.; Sherman, M.; Llerena, F.; Hernandez, M.; Nourdin, G.; Urrizola, J.; Rivera, C.; Lamperti, L.; Nova, L.; et al. Immunomodulation of t helper cells by tumor microenvironment in oral cancer is associated with ccr8 expression and rapid membrane vitamin d signaling pathway. Front. Immunol. 2021, 12, 1668. [Google Scholar]
- Hon, C.-C.; Shin, J.W.; Carninci, P.; Stubbington, M.J.T. The Human Cell Atlas: Technical approaches and challenges. Brief. Funct. Genom. 2018, 17, 283–294. [Google Scholar] [CrossRef]
- Bacher, R.; Kendziorski, C. Design and computational analysis of single-cell RNA-sequencing experiments. Genome Biol. 2016, 17, 63. [Google Scholar] [CrossRef]
- Pierson, E.; Yau, C. ZIFA: Dimensionality reduction for zero-inflated single-cell gene expression analysis. Genome Biol. 2015, 16, 241. [Google Scholar] [CrossRef]
- Lin, P.; Troup, M.; Ho, J.W. CIDR: Ultrafast and accurate clustering through imputation for single-cell RNA-seq data. Genome Biol. 2017, 18, 59. [Google Scholar] [CrossRef]
- Rostom, R.; Svensson, V.; Teichmann, S.A.; Kar, G. Computational approaches for interpreting sc RNA-seq data. FEBS Lett. 2017, 591, 2213–2225. [Google Scholar] [CrossRef] [Green Version]
- McCarthy, D.J.; Campbell, K.R.; Lun, A.T.; Wills, Q.F. Scater: Pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics 2017, 33, 1179–1186. [Google Scholar] [CrossRef]
- Gierahn, T.M.; Wadsworth, M.H.; Hughes, T.K.; Bryson, B.D.; Butler, A.; Satija, R.; Fortune, S.; Love, J.C.; Shalek, A.K. Seq-Well: Portable, low-cost RNA sequencing of single cells at high throughput. Nat. Methods 2017, 14, 395–398. [Google Scholar] [CrossRef]
- Haque, A.; Engel, J.; Teichmann, S.A.; Lönnberg, T. A practical guide to single-cell RNA-sequencing for biomedical research and clinical applications. Genome Med. 2017, 9, 75. [Google Scholar] [CrossRef]
- Kohl, S.; Baumann, B.; Rosenberg, T.; Kellner, U.; Lorenz, B.; Vadala, M.; Jacobson, S.G.; Wissinger, B. Mutations in the cone photoreceptor G-protein α-subunit gene GNAT2 in patients with achromatopsia. Am. J. Hum. Genet. 2002, 71, 422–425. [Google Scholar] [CrossRef]
- Lerea, C.L.; Bunt-Milam, A.H.; Hurley, J.B. α transducin is present in blue-, green-, and red-sensitive cone photoreceptors in the human retina. Neuron 1989, 3, 367–376. [Google Scholar] [CrossRef]
- Bürckstümmer, T.; Baumann, C.; Blüml, S.; Dixit, E.; Dürnberger, G.; Jahn, H.; Planyavsky, M.; Bilban, M.; Colinge, J.; Bennett, K.L.; et al. An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasome. Nat. Immunol. 2009, 10, 266–272. [Google Scholar] [CrossRef]
- Chang, C.-C.; Su, K.-M.; Lu, K.-H.; Lin, C.-K.; Wang, P.-H.; Li, H.-Y.; Wang, M.-L.; Lin, C.-K.; Yu, M.-H.; Chang, C.-M. Key immunological functions involved in the progression of epithelial ovarian serous carcinoma discovered by the gene ontology-based immunofunctionome analysis. Int. J. Mol. Sci. 2018, 19, 3311. [Google Scholar] [CrossRef]
- Luo, J.; Nikolaev, A.Y.; Imai, S.; Chen, D.; Su, F.; Shiloh, A.; Guarente, L.; Gu, W. Negative control of p53 by Sir2α promotes cell survival under stress. Cell 2001, 107, 137–148. [Google Scholar] [CrossRef]
- Reggiori, F.; Ungermann, C. Autophagosome maturation and fusion. J. Mol. Biol. 2017, 429, 486–496. [Google Scholar] [CrossRef]
- Mathew, R.; Karantza-Wadsworth, V.; White, E. Role of autophagy in cancer. Nat. Rev. Cancer 2007, 7, 961–967. [Google Scholar] [CrossRef]
- Claesson-Welsh, L. Vascular permeability—The essentials. Ups. J. Med. Sci. 2015, 120, 135–143. [Google Scholar] [CrossRef]
- Bates, D.O.; Harper, S.J. Regulation of vascular permeability by vascular endothelial growth factors. Vascul. Pharmacol. 2002, 39, 225–237. [Google Scholar] [CrossRef]
- Villa, E.; Ali, E.S.; Sahu, U.; Ben-Sahra, I. Cancer cells tune the signaling pathways to empower de novo synthesis of nucleotides. Cancers 2019, 11, 688. [Google Scholar] [CrossRef]
- Buj, R.; Aird, K.M. Deoxyribonucleotide triphosphate metabolism in cancer and metabolic disease. Front. Endocrinol. 2018, 9, 177. [Google Scholar] [CrossRef]
- Sasada, T.; Azuma, K.; Ohtake, J.; Fujimoto, Y. Immune responses to epidermal growth factor receptor (EGFR) and their application for cancer treatment. Front. Pharmacol. 2016, 7, 405. [Google Scholar] [CrossRef]
- Laurent-Puig, P.; Lievre, A.; Blons, H. Mutations and response to epidermal growth factor receptor inhibitors. Clin. Cancer Res. 2009, 15, 1133–1139. [Google Scholar] [CrossRef]
- Schwartsburd, P. Cancer-induced reprogramming of host glucose metabolism: «Vicious Cycle» supporting cancer progression. Front. Oncol. 2019, 9, 218. [Google Scholar] [CrossRef]
- Wang, Z.; Dong, C. Gluconeogenesis in cancer: Function and regulation of PEPCK, FBPase, and G6Pase. Trends Cancer 2019, 5, 30–45. [Google Scholar] [CrossRef]
- Grasmann, G.; Smolle, E.; Olschewski, H.; Leithner, K. Gluconeogenesis in cancer cells–repurposing of a starvation-induced metabolic pathway? Biochim. Biophys. Acta-Rev. Cancer. 2019, 1872, 24–36. [Google Scholar] [CrossRef]
- Scott, S.A.; Mathews, T.P.; Ivanova, P.T.; Lindsley, C.W.; Brown, H.A. Chemical modulation of glycerolipid signaling and metabolic pathways. Biochim. Biophys. Acta Mol. Cell Biol. Lipids. 2014, 1841, 1060–1084. [Google Scholar] [CrossRef] [Green Version]
- Luo, X.; Cheng, C.; Tan, Z.; Li, N.; Tang, M.; Yang, L.; Cao, Y. Emerging roles of lipid metabolism in cancer metastasis. Mol. Cancer 2017, 16, 76. [Google Scholar] [CrossRef]
- Kurmi, K.; Haigis, M.C. Nitrogen metabolism in cancer and immunity. Trends Cell Biol. 2020, 30, 408–424. [Google Scholar] [CrossRef]
- Coutinho, A.E.; Chapman, K.E. The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol. Cell. Endocrinol. 2011, 335, 2–13. [Google Scholar] [CrossRef]
- Masri, S.; Sassone-Corsi, P. The emerging link between cancer, metabolism, and circadian rhythms. Nat. Med. 2018, 24, 1795–1803. [Google Scholar] [CrossRef]
- Keshvari, M.; Nejadtaghi, M.; Hosseini-Beheshti, F.; Rastqar, A.; Patel, N. Exploring the role of circadian clock gene and association with cancer pathophysiology. Chronobiol. Int. 2020, 37, 151–175. [Google Scholar] [CrossRef]
- Hoesel, B.; Schmid, J.A. The complexity of NF-κB signaling in inflammation and cancer. Mol. Cancer 2013, 12, 86. [Google Scholar] [CrossRef]
- Liu, B.; Yang, R.; Wong, K.A.; Getman, C.; Stein, N.; Teitell, M.A.; Cheng, G.; Wu, H.; Shuai, K. Negative regulation of NFκB signaling by PIAS1. Mol. Cell. Biol. 2005, 25, 1113–1123. [Google Scholar] [CrossRef]
- Zeligs, K.P.; Neuman, M.K.; Annunziata, C.M. Molecular pathways: The balance between cancer and the immune system challenges the therapeutic specificity of targeting nuclear factor-κB signaling for cancer treatment. Clin. Cancer Res. 2016, 22, 4302–4308. [Google Scholar] [CrossRef]
- Zhang, J.-M.; An, J. Cytokines, inflammation and pain. Int. Anesthesiol. Clin. 2007, 45, 27. [Google Scholar] [CrossRef]
- Conlon, K.C.; Miljkovic, M.D.; Waldmann, T.A. Cytokines in the treatment of cancer. J. Interferon Cytokine Res. 2019, 39, 6–21. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.; Margolin, K. Cytokines in cancer immunotherapy. Cancers 2011, 3, 3856–3893. [Google Scholar] [CrossRef]
- Dranoff, G. Cytokines in cancer pathogenesis and cancer therapy. Nat. Rev. Cancer 2004, 4, 11–22. [Google Scholar] [CrossRef]
- Oberholzer, A.; Oberholzer, C.; Moldawer, L.L. Cytokine signaling-regulation of the immune response in normal and critically ill states. Crit. Care Med. 2000, 28, N3–N12. [Google Scholar] [CrossRef]
- Akdis, M.; Aab, A.; Altunbulakli, C.; Azkur, K.; Costa, R.A.; Crameri, R.; Duan, S.; Eiwegger, T.; Eljaszewicz, A.; Ferstl, R.; et al. Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: Receptors, functions, and roles in diseases. J. Allergy Clin. Immunol. 2016, 138, 984–1010. [Google Scholar] [CrossRef]
- Jupe, S.; Ray, K.; Roca, C.D.; Varusai, T.; Shamovsky, V.; Stein, L.; D’Eustachio, P.; Hermjakob, H. Interleukins and their signaling pathways in the Reactome biological pathway database. J. Allergy Clin. Immunol. 2018, 141, 1411–1416. [Google Scholar] [CrossRef] [PubMed]
- Pancer, Z.; Cooper, M.D. The evolution of adaptive immunity. Annu. Rev. Immunol. 2006, 24, 497–518. [Google Scholar] [CrossRef] [PubMed]
- Grell, M.; Douni, E.; Wajant, H.; Löhden, M.; Clauss, M.; Maxeiner, B.; Georgopoulos, S.; Lesslauer, W.; Kollias, G.; Pfizenmaier, K.; et al. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 1995, 83, 793–802. [Google Scholar] [CrossRef]
- Shi, J.-H.; Sun, S.-C. Tumor necrosis factor receptor-associated factor regulation of nuclear factor κb and mitogen-activated protein kinase pathways. Front. Immunol. 2018, 9, 1849. [Google Scholar] [CrossRef]
- Andrews, S. FastQC: A quality control tool for high throughput sequence data. In Babraham Bioinformatics; Babraham Institute: Cambridge, UK, 2010. [Google Scholar]
- Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef] [PubMed]
- Hwang, B.; Lee, J.H.; Bang, D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp. Mol. Med. 2018, 50, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Van der Maaten, L.; Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 2008, 9, 2579–2605. [Google Scholar]
- Kong, A.T.; Leprevost, F.V.; Avtonomov, D.M.; Mellacheruvu, D.; Nesvizhskii, A.I. MSFragger: Ultrafast and comprehensive peptide identification in mass spectrometry–based proteomics. Nat. Methods 2017, 14, 513–520. [Google Scholar] [CrossRef]
- Teo, G.C.; Polasky, D.A.; Yu, F.; Nesvizhskii, A.I. Fast deisotoping algorithm and its implementation in the MSFragger search engine. J. Proteome Res. 2021, 20, 498–505. [Google Scholar] [CrossRef]
- Yu, F.; Haynes, S.E.; Nesvizhskii, A.I. IonQuant enables accurate and sensitive label-free quantification with FDR-controlled match-between-runs. Mol. Cell Proteomics 2021, 20, 100077. [Google Scholar] [CrossRef]
- Keller, A.; Nesvizhskii, A.I.; Kolker, E.; Aebersold, R. Empirical statistical model to estimate the accuracy of peptide identifications made by MS/MS and database search. Anal. Chem. 2002, 74, 5383–5392. [Google Scholar] [CrossRef] [PubMed]
- Tyanova, S.; Temu, T.; Sinitcyn, P.; Carlson, A.; Hein, M.Y.; Geiger, T.; Mann, M.; Cox, J. The Perseus computational platform for comprehensive analysis of (prote) omics data. Nat. Methods 2016, 13, 731–740. [Google Scholar] [CrossRef] [PubMed]
- Heberle, H.; Meirelles, G.V.; da Silva, F.R.; Telles, G.P.; Minghim, R. InteractiVenn: A web-based tool for the analysis of sets through Venn diagrams. BMC Bioinform. 2015, 16, 169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Data ID | Type of Cancer | Condition | CD4 | CD8 | Treg |
---|---|---|---|---|---|
GSE114727 | Breast | Malignant | 12,855 | 12,601 | 8924 |
GSE75688 | Breast | Malignant | 8878 | 2620 | 6835 |
GSE126030 | Lung | Nonmalignant | 8418 | 10,573 | 6620 |
GSE99254 | Lung | Malignant | 10,571 | 11,034 | 10,349 |
Nonmalignant | 9748 | 10,488 | 7532 | ||
GSE108989 | Colorectal | Malignant | 7324 | 7005 | 7283 |
Nonmalignant | 6270 | 7280 | 6725 | ||
GSE103322 | Head and neck | Malignant | 7574 | 8093 | 7517 |
GSE72056 | Melanoma | Malignant | 10,136 | 10,779 | 8732 |
GSE123139 | Melanoma | Malignant | 10,579 | 10,906 | 7187 |
Functional Enrichment | Malignant | Nonmalignant | ||||
---|---|---|---|---|---|---|
CD4 | CD8 | Treg | CD4 | CD8 | Treg | |
Biological process | 1335 | 950 | 1388 | 1282 | 1265 | 1270 |
Molecular function | 270 | 159 | 249 | 240 | 249 | 237 |
Cellular component | 376 | 271 | 385 | 322 | 339 | 342 |
Reactome pathway | 2059 | 1746 | 1966 | 2073 | 2138 | 2051 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Vidal, M.; Fraga, M.; Llerena, F.; Vera, A.; Hernández, M.; Koch, E.; Reyes-López, F.; Vallejos-Vidal, E.; Cabrera-Vives, G.; Nova-Lamperti, E. Analysis of Tumor-Infiltrating T-Cell Transcriptomes Reveal a Unique Genetic Signature across Different Types of Cancer. Int. J. Mol. Sci. 2022, 23, 11065. https://doi.org/10.3390/ijms231911065
Vidal M, Fraga M, Llerena F, Vera A, Hernández M, Koch E, Reyes-López F, Vallejos-Vidal E, Cabrera-Vives G, Nova-Lamperti E. Analysis of Tumor-Infiltrating T-Cell Transcriptomes Reveal a Unique Genetic Signature across Different Types of Cancer. International Journal of Molecular Sciences. 2022; 23(19):11065. https://doi.org/10.3390/ijms231911065
Chicago/Turabian StyleVidal, Mabel, Marco Fraga, Faryd Llerena, Agustín Vera, Mauricio Hernández, Elard Koch, Felipe Reyes-López, Eva Vallejos-Vidal, Guillermo Cabrera-Vives, and Estefanía Nova-Lamperti. 2022. "Analysis of Tumor-Infiltrating T-Cell Transcriptomes Reveal a Unique Genetic Signature across Different Types of Cancer" International Journal of Molecular Sciences 23, no. 19: 11065. https://doi.org/10.3390/ijms231911065
APA StyleVidal, M., Fraga, M., Llerena, F., Vera, A., Hernández, M., Koch, E., Reyes-López, F., Vallejos-Vidal, E., Cabrera-Vives, G., & Nova-Lamperti, E. (2022). Analysis of Tumor-Infiltrating T-Cell Transcriptomes Reveal a Unique Genetic Signature across Different Types of Cancer. International Journal of Molecular Sciences, 23(19), 11065. https://doi.org/10.3390/ijms231911065