AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1.7 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Single-cell RNA-seq reveals the immune escape and drug resistance mechanisms of mantle cell lymphoma

Liang Wang1,2 ( )Steven Mo3Xin Li1Yingzhi He4Jing Yang1
Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University and Capital Medical University, Beijing Tongren Hospital, Beijing 100730, China
Nanning Life-Ontology Biotechnology Co., Ltd., Nanning 530000, China
Department of Hematology, Zhujiang Hospital of Southern Medical University, Guangzhou 510280, China
Show Author Information

Abstract

Objective

Mantle cell lymphoma (MCL) is a rare subtype of non-Hodgkin lymphoma (NHL) with high heterogeneity and a high recurrence rate. How heterogenous cell populations contribute to relapse remains to be elucidated.

Methods

We performed single cell RNA sequencing (scRNA-seq) on approximately 4,000 bone marrow cells sampled from one patient with multidrug resistant MCL. We identified 10 subpopulations comprising 4 malignant B cell subtypes, 3 T cell subtypes, 2 dendritic cell subtypes and 1 natural killer (NK) cell subtype. Subsequently, we identified cell markers, including a series of genes associated with immune escape and drug resistance. In addition, we explored the roles of these genes in the mechanism of immune escape and drug resistance, and we verified the expression imbalance and clinical prognostic potential by using GEO datasets including 211 MCL samples.

Results

The major immune escape mechanisms of MCL included anti-perforin activity, decreased immunogenicity and direct inhibition of apoptosis and cell killing, as mediated by type Ⅰ and Ⅱ B cells. The drug resistance mechanisms of different cell clusters included drug metabolism, DNA damage repair, apoptosis and survival promotion. Type Ⅲ B cells closely communicate with other cells. The key genes involved in the resistance mechanisms showed dysregulated expression and may have significant clinical prognostic value.

Conclusion

This study investigated potential immune escape and drug resistance mechanisms in MCL. The results may guide individualized treatment and promote the development of therapeutic drugs.

Electronic Supplementary Material

Download File(s)
cbm-17-3-726_ESM.pdf (2.5 MB)

References

1

Herrmann A, Hoster E, Zwingers T, Brittinger G, Engelhard M, Meusers P, et al. Improvement of overall survival in advanced stage mantle cell lymphoma. J Clin Oncol. 2009; 27: 511-8.

2

Martin P, Chadburn A, Christos P, Furman R, Ruan J, Joyce MA, et al. Intensive treatment strategies may not provide superior outcomes in mantle cell lymphoma: overall survival exceeding 7 years with standard therapies. Ann Oncol. 2008; 19: 1327-30.

3

Corneth OBJ, Klein Wolterink RGJ, Hendriks RW. BTK signaling in B cell differentiation and autoimmunity. Curr Top Microbiol Immunol. 2016; 393: 67-105.

4

Mohamed AJ, Yu L, Backesjo CM, Vargas L, Faryal R, Aints A, et al. Bruton’s tyrosine kinase (Btk): function, regulation, and transformation with special emphasis on the PH domain. Immunol Rev. 2009; 228: 58-73.

5

Hershkovitz-Rokah O, Pulver D, Lenz G, Shpilberg O. Ibrutinib resistance in mantle cell lymphoma: clinical, molecular and treatment aspects. Br J Haematol. 2018; 181: 306-19.

6

Wang ML, Rule S, Martin P, Goy A, Auer R, Kahl BS, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013; 369: 507-16.

7

Zhang SQ, Smith SM, Zhang SY, Lynn Wang Y. Mechanisms of ibrutinib resistance in chronic lymphocytic leukaemia and nonHodgkin lymphoma. Br J Haematol. 2015; 170: 445-56.

8

Woyach JA, Ruppert AS, Guinn D, Lehman A, Blachly JS, Lozanski A, et al. BTK(C481S)-mediated resistance to ibrutinib in chronic lymphocytic leukemia. J Clin Oncol. 2017; 35: 1437-43.

9

Jones D, Woyach JA, Zhao W, Caruthers S, Tu H, Coleman J, et al. PLCG2 C2 domain mutations co-occur with BTK and PLCG2 resistance mutations in chronic lymphocytic leukemia undergoing ibrutinib treatment. Leukemia. 2017; 31: 1645-7.

10

Agarwal R, Chan YC, Tam CS, Hunter T, Vassiliadis D, Teh CE, et al. Dynamic molecular monitoring reveals that SWI-SNF mutations mediate resistance to ibrutinib plus venetoclax in mantle cell lymphoma. Nat Med. 2019; 25: 119-29.

11

McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017; 168: 613-28.

12

Zeisel A, Munoz-Manchado AB, Codeluppi S, Lonnerberg P, La Manno G, Jureus A, et al. Brain structure. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science. 2015; 347: 1138-42.

13

Pollen AA, Nowakowski TJ, Shuga J, Wang X, Leyrat AA, Lui JH, et al. Low-coverage single-cell mRNA sequencing reveals cellular heterogeneity and activated signaling pathways in developing cerebral cortex. Nat Biotechnol. 2014; 32: 1053-8.

14

Wills QF, Livak KJ, Tipping AJ, Enver T, Goldson AJ, Sexton DW, et al. Single-cell gene expression analysis reveals genetic associations masked in whole-tissue experiments. Nat Biotechnol. 2013; 31: 748-52.

15

Shalek AK, Satija R, Adiconis X, Gertner RS, Gaublomme JT, Raychowdhury R, et al. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature. 2013; 498: 236-40.

16

Grun D, van Oudenaarden A. Design and analysis of single-cell sequencing experiments. Cell. 2015; 163: 799-810.

17

Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018; 36: 411-20.

18

Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 2015; 33: 495-502.

19

Yu G, Wang LG, Han Y, He QY. Clusterprofiler: an R package for comparing biological themes among gene clusters. OMICS. 2012; 16: 284-7.

20

Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, et al. Tcgabiolinks: an R/bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016; 44: e71.

21

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res. 2013; 41: D991-5.

22

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015; 43: e47.

23

Law CW, Chen Y, Shi W, Smyth GK. Voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014; 15: R29.

24

Smyth GK. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004; 3: Article 3.

25
Wickham H. GGplot2: elegant graphics for data analysis. New York: Springer; 2009. http://dx.doi.org/10.1007/978-0-387-98141-3.
26

Malta TM, Sokolov A, Gentles AJ, Burzykowski T, Poisson L, Weinstein JN, et al. Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell. 2018; 173: 338-54 e15.

27

Stahl D, Lacroix-Desmazes S, Heudes D, Mouthon L, Kaveri SV, Kazatchkine MD. Altered control of self-reactive IgG by autologous IgM in patients with warm autoimmune hemolytic anemia. Blood. 2000; 95: 328-35.

28

Kasakura S.[A role for T-helper type 1 and type 2 cytokines in the pathogenesis of various human diseases]. Rinsho Byori. 1998; 46: 915-21.

29

Haas H, Schlaak M.[The th1/th2 concept – its importance for regulation of IgE]. Immun Infekt. 1994; 22: 88-93.

30

Romagnani S. Type 1 T helper and type 2 T helper cells: functions, regulation and role in protection and disease. Int J Clin Lab Res. 1991; 21: 152-8.

31

Raphael I, Nalawade S, Eagar TN, Forsthuber TG. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine. 2015; 74: 5-17.

32

Gardner A, Ruffell B. Dendritic cells and cancer immunity. Trends Immunol. 2016; 37: 855-65.

33

Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007; 449: 419-26.

34

Steinman RM. Decisions about dendritic cells: past, present, and future. Annu Rev Immunol. 2012; 30: 1-22.

35

Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998; 392: 245-52.

36

Agrawal S, Kishore MC. MHC class Ⅰ gene expression and regulation. J Hematother Stem Cell Res. 2000; 9: 795-812.

37

Brea EJ, Oh CY, Manchado E, Budhu S, Gejman RS, Mo G, et al. Kinase regulation of human MHC class Ⅰ molecule expression on cancer cells. Cancer Immunol Res. 2016; 4: 936-47.

38

Spicer BA, Conroy PJ, Law RHP, Voskoboinik I, Whisstock JC. Perforin-a key (shaped) weapon in the immunological arsenal. Semin Cell Dev Biol. 2017; 72: 117-23.

39

Voskoboinik I, Whisstock JC, Trapani JA. Perforin and granzymes: function, dysfunction and human pathology. Nat Rev Immunol. 2015; 15: 388-400.

40

Lopez JA, Susanto O, Jenkins MR, Lukoyanova N, Sutton VR, Law RH, et al. Perforin forms transient pores on the target cell plasma membrane to facilitate rapid access of granzymes during killer cell attack. Blood. 2013; 121: 2659-68.

41

Zhou F. Expression of multiple granzymes by cytotoxic T lymphocyte implies that they activate diverse apoptotic pathways in target cells. Int Rev Immunol. 2010; 29: 38-55.

42

Lopez-Herrera G, Vargas-Hernandez A, Gonzalez-Serrano ME, Berron-Ruiz L, Rodriguez-Alba JC, Espinosa-Rosales F, et al. Bruton’s tyrosine kinase–an integral protein of B cell development that also has an essential role in the innate immune system. J Leukoc Biol. 2014; 95: 243-50.

43

Kim E, Hurtz C, Koehrer S, Wang Z, Balasubramanian S, Chang BY, et al. Ibrutinib inhibits pre-BCR(+) B-cell acute lymphoblastic leukemia progression by targeting BTK and BLK. Blood. 2017; 129: 1155-65.

44

Deeks ED. Ibrutinib: a review in chronic lymphocytic leukaemia. Drugs. 2017; 77: 225-36.

45

Zuccolo J, Deng L, Unruh TL, Sanyal R, Bau JA, Storek J, et al. Expression of MS4A and TMEM176 genes in human B lymphocytes. Front Immunol. 2013; 4: 195.

46

Payandeh Z, Bahrami AA, Hoseinpoor R, Mortazavi Y, Rajabibazl M, Rahimpour A, et al. The applications of anti-CD20 antibodies to treat various B cells disorders. Biomed Pharmacother. 2019; 109: 2415-26.

47

Salles G, Barrett M, Foa R, Maurer J, O’Brien S, Valente N, et al. Rituximab in B-cell hematologic malignancies: a review of 20 years of clinical experience. Adv Ther. 2017; 34: 2232-73.

48

Lim SH, Vaughan AT, Ashton-Key M, Williams EL, Dixon SV, Chan HT, et al. Fc gamma receptor ⅡB on target B cells promotes rituximab internalization and reduces clinical efficacy. Blood. 2011; 118: 2530-40.

49

Sethi T, Rintoul RC, Moore SM, MacKinnon AC, Salter D, Choo C, et al. Extracellular matrix proteins protect small cell lung cancer cells against apoptosis: a mechanism for small cell lung cancer growth and drug resistance in vivo. Nat Med. 1999; 5: 662-8.

50

Shain KH, Tao J. The B-cell receptor orchestrates environmentmediated lymphoma survival and drug resistance in B-cell malignancies. Oncogene. 2014; 33: 4107-13.

51

Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS. Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines. Blood. 1999; 93: 1658-67.

52

Salehan MR, Morse HR. DNA damage repair and tolerance: a role in chemotherapeutic drug resistance. Br J Biomed Sci. 2013; 70: 31-40.

53

Sakthivel KM, Hariharan S. Regulatory players of DNA damage repair mechanisms: role in cancer chemoresistance. Biomed Pharmacother. 2017; 93: 1238-45.

54

Liu D, Mamorska-Dyga A. Syk inhibitors in clinical development for hematological malignancies. J Hematol Oncol. 2017; 10: 145.

55

Xiong S, Pant V, Zhang Y, Aryal NK, You MJ, Kusewitt D, et al. The p53 inhibitor Mdm4 cooperates with multiple genetic lesions in tumourigenesis. J Pathol. 2017; 241: 501-10.

56

Mohanty A, Sandoval N, Das M, Pillai R, Chen L, Chen RW, et al. CCND1 mutations increase protein stability and promote ibrutinib resistance in mantle cell lymphoma. Oncotarget. 2016; 7: 73558-72.

57

Belmar J, Fesik SW. Small molecule Mcl-1 inhibitors for the treatment of cancer. Pharmacol Ther. 2015; 145: 76-84.

58

Jares P, Colomer D, Campo E. Genetic and molecular pathogenesis of mantle cell lymphoma: perspectives for new targeted therapeutics. Nat Rev Cancer. 2007; 7: 750-62.

59

Smith MR. Should there be a standard therapy for mantle cell lymphoma? Future Oncol. 2011; 7: 227-37.

60

Dreyling M. Therapy of mantle cell lymphoma: new treatment options in an old disease or vice versa? Semin Hematol. 2011; 48: 145-7.

61

Tu SM, Lin SH, Logothetis CJ. Stem-cell origin of metastasis and heterogeneity in solid tumours. Lancet Oncol. 2002; 3: 508-13.

62

Alizadeh AA, Aranda V, Bardelli A, Blanpain C, Bock C, Borowski C, et al. Toward understanding and exploiting tumor heterogeneity. Nat Med. 2015; 21: 846-53.

63

Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012; 481: 306-13.

64

Marusyk A, Tabassum DP, Altrock PM, Almendro V, Michor F, Polyak K. Non-cell-autonomous driving of tumour growth supports sub-clonal heterogeneity. Nature. 2014; 514: 54-8.

65

Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, et al. Emergence of KRAS mutations and acquired resistance to antiEGFR therapy in colorectal cancer. Nature. 2012; 486: 532-6.

66

Voskoboinik I, Dunstone MA, Baran K, Whisstock JC, Trapani JA. Perforin: structure, function, and role in human immunopathology. Immunol Rev. 2010; 235: 35-54.

67

Schroter M, Lowin B, Borner C, Tschopp J. Regulation of fas(apo-1/CD95)- and perforin-mediated lytic pathways of primary cytotoxic T lymphocytes by the protooncogene bcl-2. Eur J Immunol. 1995; 25: 3509-13.

68

Hu X, Zhang Z. Understanding the genetic mechanisms of cancer drug resistance using genomic approaches. Trends Genet. 2016; 32: 127-37.

69

Longley DB, Johnston PG. Molecular mechanisms of drug resistance. J Pathol. 2005; 205: 275-92.

70

Shain KH, Dalton WS. Cell adhesion is a key determinant in de novo multidrug resistance (MDR): new targets for the prevention of acquired MDR. Mol Cancer Ther. 2001; 1: 69-78.

71

Jares P, Colomer D, Campo E. Molecular pathogenesis of mantle cell lymphoma. J Clin Invest. 2012; 122: 3416-23.

72

Wu SY, Lan SH, Liu HS. Degradative autophagy selectively regulates CCND1 (cyclin d1) and MIR224, two oncogenic factors involved in hepatocellular carcinoma tumorigenesis. Autophagy. 2019; 15: 729-30.

73

Baykara O, Dalay N, Bakir B, Bulut P, Kaynak K, Buyru N. The emsy gene collaborates with CCND1 in non-small cell lung carcinogenesis. Int J Med Sci. 2017; 14: 675-9.

74

Ma L, Wang X, Lan F, Yu Y, Ouyang X, Liu W, et al. Prognostic value of differential CCND1 expression in patients with resected gastric adenocarcinoma. Med Oncol. 2015; 32: 338.

75

Seiler R, Thalmann GN, Rotzer D, Perren A, Fleischmann A. CCND1/cyclind1 status in metastasizing bladder cancer: a prognosticator and predictor of chemotherapeutic response. Mod Pathol. 2014; 27: 87-95.

76

Chen Y, Shan Y, Lu M, DeSouza N, Guo Z, Hoffman R, et al. Alox5 blockade eradicates JAK2V617F-induced polycythemia vera in mice. Cancer Res. 2017; 77: 164-74.

Cancer Biology & Medicine
Pages 726-739
Cite this article:
Wang L, Mo S, Li X, et al. Single-cell RNA-seq reveals the immune escape and drug resistance mechanisms of mantle cell lymphoma. Cancer Biology & Medicine, 2020, 17(3): 726-739. https://doi.org/10.20892/j.issn.2095-3941.2020.0073

53

Views

0

Downloads

23

Crossref

N/A

Web of Science

24

Scopus

Altmetrics

Received: 21 February 2020
Accepted: 28 April 2020
Published: 15 August 2020
©2020 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return