eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3/2024
vol. 49
 
Share:
Share:
Review paper

New T-lymphocyte subpopulations and their characteristics: Challenges to the classical division of lymphocyte function

Filip Lewandowski
1, 2
,
Paulina Niedźwiedzka-Rystwej
1, 2

  1. Institute of Biology, University of Szczecin, Szczecin, Poland
  2. Center for Experimental Immunology and Immunobiology in Infectious Disease and Cancer, University of Szczecin, Szczecin, Poland
Cent Eur J Immunol 2024; 49 (3): 308-314
Online publish date: 2024/11/09
Article file
- New T-lymphocyte (1).pdf  [0.22 MB]
Get citation
 
PlumX metrics:
 

Introduction

In the field of immunology, our understanding of T-lymphocyte subpopulations and their functions has undergone significant advances in recent years [1]. These findings challenge the traditional categorization of lymphocyte function and call for a reevaluation of the characteristics attributed to different T-lymphocyte subsets. These new developments have been made possible by innovative reagents and techniques, which have allowed researchers to gain insights into thymic differentiation status and the phenotypes of both premalignant and malignant lymphocytic infiltrates [2]. Furthermore, the identification of previously unrecognized cells within lymphomas and the distinction between circulating and skin lymphocytes have provided valuable information. These discoveries not only expand our knowledge of T-lymphocyte biology, but also have potential therapeutic applications [3]. The study of T-lymphocyte subpopulations is a rapidly evolving field in immunology, challenging the conventional division of lymphocyte function [4]. Using advanced reagents and techniques, scientists have been able to gain a deeper understanding of T-lymphocyte subpopulations and their roles in immune responses.

Helper (Th) or memory (Tm) lymphocytes?

The classical division treats the two subpopulations as distinct. However, the introduction of single cell analysis techniques and the associated discovery of new lymphocyte surface markers disrupts this division. Increasingly, the newly discovered subpopulations of helper lymphocytes also show the function performed by the hitherto separate memory lymphocytes [5-7].

Helper lymphocytes, also known as CD4+ T cells, play a crucial role in regulating immune responses and maintaining immune homeostasis. Recent research in the field of immunology has uncovered several new variants of helper lymphocytes (Fig. 1), expanding our understanding of their diverse functions and potential therapeutic applications [8]. One of the emerging variants of helper lymphocytes is Tfh1, which stands for T follicular helper 1 cells. Tfh1 cells (Table 1) are a subset of helper lymphocytes that are characterized by their ability to provide help and support to B cells in the germinal centers of lymphoid structures and promote effective adaptive immune responses [9]. Another variant that has gained attention is Tfh2, which refers to T follicular helper 2 cells [10]. Tfh2 cells (Table 1) are involved in promoting humoral immunity and have been implicated in various diseases, including autoimmune disorders and allergic reactions [11]. In addition to Tfh1 and Tfh2 cells, another variant that has emerged is Tfh17 [10]. Tfh17 cells (Table 1) represent a specialized subset of helper lymphocytes that produce the cytokine interleukin (IL)-17 and are involved in promoting immune responses against extracellular pathogens and maintaining tissue barrier functions [12]. These Tfh subsets can be found in the germinal centers of lymphoid tissues, where they assist B cells in antibody production. However, they are not restricted to these sites. Studies have shown that Tfh1, Tfh2, and Tfh17 cells can also be detected in the peripheral blood, indicating their presence in circulation. This is essential for their role in systemic immune responses and their ability to migrate to different sites as needed [13-16]. Furthermore, the discovery of germinal center T follicular helper cells has shed light on a distinct population of helper lymphocytes that provide critical support to B cells in the germinal centers during the process of antibody production and affinity maturation [17]. These cells, known as GC-Tfh cells (Table 1), play a crucial role in shaping the humoral immune response and have been implicated in both protective and pathological immune responses [11]. Another noteworthy variant of helper lymphocytes is cTfh, which stands for circulating T follicular helper cells. cTfh cells (Table 1) are a subset of helper lymphocytes that circulate in the bloodstream and can migrate to secondary lymphoid organs, such as lymph nodes and the spleen, where they interact with B cells and contribute to the generation of antibody responses [18]. Specific subsets of circulating Tfh cells have also been identified, including cTfhCM, cTfh1, cTfh2, and cTfh17 [19] (Table 1). cTfhCM stands for circulating T follicular helper central memory cells, which are a subset of cTfh cells that possess memory-like properties and can provide long-lasting immune protection upon re-exposure to a specific antigen [20]. cTfh1 cells are a circulating subset of T follicular helper cells that primarily produce the cytokine interferon γ and have been associated with enhancing cellular immune responses and promoting antiviral immunity [17]. cTfh2 cells are another subset of circulating T follicular helper cells that produce the cytokine IL-4 and have been implicated in promoting humoral immune responses and supporting the generation of antibody production [18]. These subsets of cTfh cells have been found to play important roles in various diseases and immune responses [19]. Recent studies have highlighted the involvement of Tfh subsets in autoimmune disorders and allergic reactions. For example, Tfh17 cells have been shown to play a role in autoimmune diseases such as rheumatoid arthritis and multiple sclerosis, where they contribute to the activation of auto-reactive B cells and the production of pathogenic autoantibodies [21]. Additionally, Tfh subsets have also been implicated in certain types of cancer [9]. In the context of cancer, Tfh subsets have been found to infiltrate tumor tissues and play a dual role in either promoting or inhibiting anti-tumor immune responses, depending on the specific context [10]. Overall, the discovery of these new variants of helper lymphocytes has significantly expanded our understanding of their roles in immune responses and disease pathogenesis [22].

Table 1

Summary of Tfh lymphocytes and their surface markers

Name of the populationSurface markers
Tfh1CXCR5, CD4+CXCR3+CCR6- [29]
Tfh2CXCR3+CCR6- [29]
Tfh17CXCR3+CCR6+ [29]
GC-TfhCD4+ CXCR5high PD-1high BCL6high
cTfhCD4+ CD45RACXCR5+
cTfhCMCD4+ CXCR5+ CCR7high PD-1low
CD4+ CXCR5+ CCR7low PD-1high
cTfh1CD4+ CD45RACXCR5+ CXCR3+ CCR6
cTfh2CD4+ CD45RACXCR5+ CXCR3 CCR6-
cTfh17CD4+ CD45RACXCR5+ CXCR3 CCR6+
Fig. 1

Follicular lymphocytes with their molecular markers

/f/fulltexts/CEJOI/54976/CEJI-49-54976-g001_min.jpg

Cytotoxic lymphocytes (CTL)

Cytotoxic T lymphocytes play a vital role in the immune response against pathogens and cancer cells by recognizing and eliminating target cells [23]. In recent years, new subsets of cytotoxic T lymphocytes have been identified and characterized, including CD8+CD39+ and CD8+CD28+ T cells (Fig. 2) [24]. These subsets have garnered interest due to their unique functional properties and potential for therapeutic applications in cancer immunotherapy [25]. The CD8+CD39+ T-cell subset was found in various locations within the body, with significant presence in tumor microenvironments, especially within tumor-infiltrating lymphocytes (TILs). These cells have been observed in lymph node metastases from melanoma patients, where they are associated with the regulation of immune responses within the tumor context. In addition to tumor patients, CD8+CD39+ T cells have been found in the peripheral blood of healthy patients [26-28]. These cytotoxic T cells subset has been found to express high levels of the ectonucleotidase CD39, which hydrolyzes ATP to AMP and ADP [29]. CD73 converts it into immunosuppressive adenosine. Adenosine interacts with adenosine receptors on immune cells, leading to the suppression of T cell activation, proliferation, and cytokine production. CD73 expression on CD8+ T cells can enhance the immunosuppressive effects initiated by CD39, contributing to the inhibition of effective anti-tumor immune responses. In the tumor microenvironment (TME), high levels of CD73 are often associated with worse prognoses due to the increased production of adenosine, which dampens the cytotoxic functions of CD8+ T cells and supports tumor progression [29-31]. This immunosuppressive property allows CD8+CD39+ T cells to dampen immune responses and contribute to immune tolerance [24]. On the other hand, CD8+CD28+ T cells are characterized by the expression of the co-stimulatory molecule CD28 [23]. CD28 provides a critical signal for T cell activation and enhances the cytotoxicity of CD8+ T cells [24]. CD8+CD28+ T cells are primarily found in the peripheral blood and lymphoid tissues, playing a crucial role in the immune system by providing costimulatory signals necessary for T cell activation and survival [32]. The presence of CD8+CD28+ T cells has been associated with a more potent anti-tumor immune response and improved clinical outcomes in cancer patients receiving immunotherapy [33]. Several studies have highlighted the functional capabilities of CD8+CD39+ and CD8+CD28+ T cells in different disease contexts. For instance, CD8+CD39+ T cells have been implicated in the regulation of autoimmune diseases by suppressing excessive immune activation [23]. Additionally, CD8+CD28+ T cells have shown efficacy in tumor cell killing and have been associated with favorable outcomes in cancer patients [33]. In the context of HIV infection, CD8+CD28+ T cells play a crucial role in the immune response against the virus. These cells are involved in the early stages of infection, providing essential costimulatory signals for T cell activation and proliferation. CD28 is a key molecule that enhances T cell receptor (TCR) signaling, leading to increased cytokine production, cell survival, and proliferation [34, 35]. CD8+CD28+ T cells play a significant role in the pathogenesis and progression of rheumatoid arthritis (RA). These cells contribute to the autoimmune response, inflammation, and joint damage characteristic of RA. Their presence in synovial fluid indicates active participation in the local inflammatory response, with sustained cytokine production driving chronic inflammation. Modulating the activity of these T cells could help control RA, making them potential therapeutic targets [36, 37]. However, when discussing CD8+CD28+ lymphocytes, one cannot fail to mention CD8+CD28 immunosenescent T cells. Immunosenescent CD28 T cells play significant roles in the TME due to their unique characteristics and impact on tumor immunity [38]. These cells, which lack the costimulatory molecule CD28, are commonly found in various cancers and are associated with poor prognosis [39]. These cells contribute to the immunosuppressive environment of tumors by secreting inhibitory cytokines such as IL-10 and TGF-β, which suppress the activity of effector T cells [40]. Their presence in the TME can hinder effective immune responses against tumor cells. The presence of immunosenescent CD28- T cells poses a challenge for immunotherapy. These cells are less responsive to treatments such as checkpoint inhibitors, which aim to reinvigorate exhausted T cells. This reduced responsiveness necessitates alternative strategies to either target these cells for removal or restore their function [38-41]. Further research on these subsets of cytotoxic T lymphocytes is needed to fully understand their mechanisms of action and optimize their use in therapeutic approaches.

Fig. 2

Cytotoxic lymphocytes and their surface markers

/f/fulltexts/CEJOI/54976/CEJI-49-54976-g002_min.jpg

In describing the CD28 molecule and its role, terminally differentiated effector memory T cells (TEMRA) are worth mentioning. TEMRA are a subset of CD8+ T cells characterized by their unique phenotype and functional attributes, often associated with the expression of CD28 [42]. TEMRA cells typically exhibit markers of terminal differentiation, including CD45RA and the loss of CD27 and CD28. However, subsets of TEMRA cells can still express CD28, indicating some degree of functional variability within this population [43]. These cells are commonly found in the peripheral blood as well as within the tumor microenvironment, where they can adopt different phenotypes depending on their location and the signals they receive [42]. TEMRA cells are highly cytotoxic, possessing a potent ability to produce effector molecules such as perforin and granzyme B. This cytotoxic capacity allows them to effectively target and kill infected or malignant cells [44]. The loss of CD28, however, can impair their proliferative response and reduce their capacity to mount sustained immune responses, which is particularly relevant in chronic infections and cancer [42]. Within tumors, TEMRA cells often display an exhausted phenotype, characterized by the expression of inhibitory receptors such as PD-1. This exhaustion limits their effectiveness in anti-tumor responses [43]. Additionally, the presence of CD28 TEMRA cells in tumors is associated with poor prognosis, as these cells can contribute to an immunosuppressive environment that supports tumor growth and progression. Understanding the dual roles of CD28+ and CD28 TEMRA cells in cancer can provide insights into their contributions to immune evasion and tumor progression. Targeting the pathways that regulate the differentiation and function of TEMRA cells could enhance the effectiveness of immunotherapies, particularly in cancers where these cells are prevalent and functionally impaired [42-44].

CD20+ T lymphocytes

CD20 is a cell surface antigen found on B cells and plays a crucial role in the regulation of B cell development and differentiation [22, 45]. CD20+ T cells, a newly discovered subset of T cells expressing the CD20 antigen (Fig. 3), have gained significant interest in recent research [46]. These CD20+ T cells have been found to possess unique characteristics and effector functions that make them promising candidates for immunotherapeutic interventions in various diseases including cancer, autoimmune disorders, and infectious diseases [47]. The presence of CD20+ T cells was initially thought to be an anomaly, as CD20 was primarily considered a B cell-specific marker. However, recent studies have demonstrated that a small subset of human T cells expresses low levels of CD20 or a cross-reacting antigen [7]. Further investigations using different CD20 monoclonal antibodies, such as Leu16, B1, and 1F5, have confirmed the presence of CD20 on T cells [46]. CD20+ T lymphocytes have been identified in many human tissues, i.e. peripheral blood, spleen, bone marrow, thymus, cerebral spinal fluid and tonsils [48-50]. The functional significance of CD20 expression on T cells is a topic of ongoing research. Recent studies have revealed that CD20+ T cells possess cytotoxic effector programs and can mediate direct killing of tumor cells. These findings suggest that CD20+ T cells play a role in anti-tumor immunity and have the potential to be used as therapeutic targets in cancer immunotherapy. Furthermore, omics technologies have provided further insight into the clinical relevance of CD20+ T cells in cancer. Single-cell RNA sequencing analyses of intra-tumoral immune cells have identified CD4+ T cells expressing cytotoxic molecules, including CD20, in a broad range of cancer types [7]. These cytotoxic CD4+ T cells have been found to exhibit MHC-II-restricted direct cytotoxicity against patient tumor cells, and their presence has been associated with a favorable prognosis or clinical response to anti-PD-L1 therapy or therapeutic vaccination [48]. Moreover, the therapeutic importance of CD20+ T cells extends beyond cancer immunotherapy. CD20+ T cells have also shown potential in the treatment of autoimmune disorders and infectious diseases. For example, in autoimmune diseases such as multiple sclerosis, CD20+ T cells have been implicated in sustaining a local microenvironment that promotes disease progression. In infectious diseases, CD20+ T cells may have a role in modulating the immune response and potentially contributing to pathogen clearance [7]. In conclusion, the discovery of CD20+ T cells challenges the traditional view of CD20 as a B cell-specific marker. These findings highlight the need for further research to elucidate the functional significance of CD20 expression on T cells and explore its potential as a therapeutic target in anti- tumor immunity, autoimmune disorders, and infectious diseases [46].

Fig. 3

T and B lymphocytes with the surface marker CD20

/f/fulltexts/CEJOI/54976/CEJI-49-54976-g003_min.jpg

Conclusions

This review article highlights significant advances in the understanding of T-lymphocyte subpopulations and their functions, shedding new light on the complexity of the immune system and potential therapeutic implications. Discoveries of new variants of helper lymphocytes, such as Tfh1, Tfh2, Tfh17, GC-Tfh, and cTfh cells and their different subtypes, together with the characterization of newly identified subpopulations of cytotoxic T cells, open new pathways for understanding the mechanisms of the immune response and the development of diseases.

Also, the demonstration of specific functions and surface markers of these T-lymphocyte subpopulations points to the possibility of more targeted interventions in various pathological conditions, including cancer, autoimmune diseases, and infections.

In conclusion, these breakthroughs in the immunology of T-lymphocyte subpopulations not only expand our knowledge of the complex ecosystem of the immune system, but also open new perspectives for the development of new therapeutic strategies. Further research is needed to fully understand the clinical potential of these discoveries and their application in medical practice, which may lead to the development of new, more effective targeted therapies.

Funding

This research received no external funding.

Disclosures

Approval of the Bioethics Committee was not required.

Notes

[1] Conflicts of interest The authors declare no conflict of interest.

References

1 

Luckheeram RV, Zhou R, Verma AD, Xia B (2012): CD4+ T cells: Differentiation and functions. Clin Dev Immunol 2012: 1-12.

2 

Kisielow P (2014): Demonstration of functional heterogeneity of T lymphocytes and identification of their two major subsets. Front Immunol 5: 609.

3 

Raskov H, Orhan A, Christensen JP, Gögenur I (2021): Cyto-toxic CD8+ T cells in cancer and cancer immunotherapy. Br J Cancer 124: 359-367.

4 

Pistoia V, Tumino N, Vacca P, et al. (2018): Human Γδ T-cells: From surface receptors to the therapy of high-risk leukemias. Front Immunol 9: 984.

5 

Niedźwiedzka-Rystwej P, Tokarz-Deptuła B, Deptuła W (2013): Characteristics of T lymphocyte subpopulations. Postepy Hig Med Dosw 67: 371-379.

6 

Gold MC, Lewinsohn DM (2013): Co-dependents: MR1-restricted MAIT cells and their antimicrobial function. Nat Rev Microbiol 11: 14-19.

7 

Schmiedel BJ, Gonzalez-Colin C, Fajardo V, et al. (2022): Single-cell EQTL analysis of activated T cell subsets reveals activation and cell type–dependent effects of disease-risk variants. Sci Immunol 7: eabm2508.

8 

Velders MP, Markiewicz MA, Eiben GL, Kast WM (2003): CD4+ T cell matters in tumor immunity. Int Rev Immunol 22: 113-140.

9 

Bosisio FM, Cerroni L (2015): Expression of T-follicular helper markers in sequential biopsies of progressive mycosis fungoides and other primary cutaneous T-cell lymphomas. Am J Dermatopathol 37: 115-121.

10 

Thornhill JP, Fidler S, Klenerman P, et al. (2017): The role of CD4+ T follicular helper cells in HIV infection: From the germinal center to the periphery. Front Immunol 8: 46.

11 

Qin L, Waseem TC, Sahoo A, et al. (2018): Insights into the molecular mechanisms of T follicular helper-mediated immunity and pathology. Front Immunol 9: 1884.

12 

Crotty S (2019): T follicular helper cell biology: A decade of discovery and diseases. Immunity 50: 1132-1148.

13 

Che Y, Qiu J, Jin T, et al. (2016): Circulating memory T follicular helper subsets, Tfh2 and Tfh17, participate in the pathogenesis of Guillain-Barré syndrome. Sci Rep 6: 20963.

14 

De Monte L, Clemente F, Ruggiero E, et al. (2023): Pro-tumor Tfh2 cells induce detrimental IgG4 production and PGE2-dependent IgE inhibition in pancreatic cancer. EBioMedicine 97: 104819.

15 

Gao X, Luo K, Wang D, et al. (2023): T follicular helper 17 (Tfh17) cells are superior for immunological memory maintenance. Elife 12: e82217.

16 

Vella LA, Buggert M, Manne S, et al. (2019): T follicular helper cells in human efferent lymph retain lymphoid characteristics. J Clin Invest 129: 3185-3200.

17 

Qi J, Liu C, Bai Z, et al. (2023): T follicular helper cells and T follicular regulatory cells in autoimmune diseases. Front Immunol 14: 1178792.

18 

Gensous N, Charrier M, Duluc D, et al. (2018): T follicular helper cells in autoimmune disorders. Front Immunol 9: 1637.

19 

Cicalese MP, Salek-Ardakani S, Fousteri G (2020): Editorial: Follicular helper T cells in immunity and autoimmunity. Front Immunol 11: 1042.

20 

Asrir A, Aloulou M, Gador M, et al. (2017): Interconnected subsets of memory follicular helper T cells have different effector functions. Nat Commun 8: 847.

21 

Ueno H (2016): T follicular helper cells in human autoimmunity. Curr Opin Immunol 43: 24-31.

22 

Dobrzanski MJ (2013): Expanding Roles for CD4 T Cells and Their Subpopulations in Tumor Immunity and Therapy. Front Oncol 3: 63.

23 

García-Chagollán M, Ledezma-Lozano IY, Hernández-Bello J, et al. (2020): Expression patterns of CD28 and CTLA-4 in early, chronic, and untreated rheumatoid arthritis. J Clin Lab Anal 34: e23188.

24 

Simoni Y, Becht E, Fehlings M, et al. (2018): Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 557: 575-579.

25 

Raskov H, Orhan A, Christensen JP, Gögenur I (2021): Cyto-toxic CD8+ T cells in cancer and cancer immunotherapy. Br J Cancer 124: 359-367.

26 

Yost KE, Satpathy AT, Wells DK, et al. (2019): Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med 25: 1251-1259.

27 

Koppensteiner L, Mathieson L, Pattle S, et al. (2023): Location of CD39+ T cell subpopulations within tumors predict differential outcomes in non-small cell lung cancer. J Immunother Cancer 11: e006770.

28 

Attrill GH, Lee H, Tasker AT, et al. (2022): Detailed spatial immunophenotyping of primary melanomas reveals immune cell subpopulations associated with patient outcome. Front Immunol 13: 979993.

29 

Timperi E, Barnaba V (2021): CD39 regulation and functions in T cells. Int J Mol Sci 2021; 22: 8068.

30 

Whittaker S, Jones CL (2023): CD39-CD73-adenosine effects in Sézary syndrome. Blood 141: 9-10.

31 

Ortiz MA, Diaz-Torné C, De Agustin JJ, et al. (2024): Altered CD39 and CD73 expression in rheumatoid arthritis: Implications for disease activity and treatment response. Biomolecules 14: 1.

32 

Geng R, Tang H, You T, et al. (2023): Peripheral CD8+CD28+ T lymphocytes predict the efficacy and safety of PD-1/PD-L1 inhibitors in cancer patients. Front Immunol 14: 1125876.

33 

Liu C, Hu Q, Hu K, et al. (2019): Increased CD8+CD28+ T cells independently predict better early response to stereotactic ablative radiotherapy in patients with lung metastases from non-small cell lung cancer. J Transl Med 17: 120.

34 

Caruso A, Licenziati S, Canaris AD, et al. (1998): Contribution of CD4+, CD8+CD28+, and CD8+CD28-T cells to CD3+ lymphocyte homeostasis during the natural course of HIV-1 infection. J Clin Invest 101: 137-144.

35 

Cartwright EK, Pampusch MS, Rendahl AK, et al. (2022): HIV-specific CAR T cells with CD28 or 4-1BB signaling domains are phenotypically and functionally distinct and effective at suppressing HIV and simian immunodeficiency virus. Immunohorizons 6: 693-704.

36 

Körmendy D, Hoff H, Hoff P, et al. (2013): Impact of the CTLA-4/CD28 axis on the processes of joint inflammation in rheumatoid arthritis. Arthritis Rheum 65: 81-87.

37 

García-Chagollán M, Ledezma-Lozano IY, Hernández-Bello J, et al. (2020): Expression patterns of CD28 and CTLA-4 in early, chronic, and untreated rheumatoid arthritis. J Clin Lab Anal 34: e23188.

38 

Huff WX, Kwon JH, Henriquez M, et al. (2019): The evolving role of CD8+CD28- immunosenescent T cells in cancer immunology. Int J Mol Sci 20: 2810.

39 

Huff WX, Bam M, Shireman JM, et al. (2021): Aging-and tumor-mediated increase in CD8+CD28- T cells might impose a strong barrier to success of immunotherapy in glioblastoma. Immunohorizons 5: 395-409.

40 

Liu G, Yu Y, Feng F, et al. (2020): Human CD8+CD28- T suppressor cells expanded by common gamma chain (γc) cytokines retain steady allospecific suppressive capacity in vivo. BMC Immunol 21: 23.

41 

Chen X, Liu Q, Xiang AP (2018): CD8+CD28-T cells: not only age-related cells but a subset of regulatory T cells. Cell Mol Immunol 15: 734-736.

42 

Lee S, Choi HY, Lee G, et al. (2021): CD8+ TILs in NSCLC differentiate into TEMRA via a bifurcated trajectory: deciphering immunogenicity of tumor antigens. J Immunother Cancer 9: e002709.

43 

Guo L, Liu X, Su X (2023): The role of TEMRA cell-mediated immune senescence in the development and treatment of HIV disease. Front Immunol 14: 1284293.

44 

Türk L, Filippov I, Arnold C, et al. (2024): Cytotoxic CD8+ Temra cells show loss of chromatin accessibility at genes associated with T cell activation. Front Immunol 15: 1285798.

45 

Uckun FM, Burkhardt AL, Jarvis L, et al. (1993): Signal transduction through the CD19 receptor during discrete developmental stages of human B-cell ontogeny. J Biol Chem 268: 21172-21184.

46 

Chen Q, Yuan S, Sun H, Peng L (2019): CD3+CD20+ T cells and their roles in human diseases. Hum Immunol 80: 191-194.

47 

Semeniuk-Wojtaś A, Modzelewska M, Poddębniak-Strama K, et al. (2023): CD4, CD20 and PD-L1 as markers of recurrence in non-muscle-invasive bladder cancer. Cancers (Basel) 15: 5529.

48 

Lee AYS (2022): CD20+ T cells: An emerging T cell subset in human pathology. Inflamm Res 71: 1181-1189.

49 

de Bruyn M, Wiersma VR, Wouters MCA, et al. (2015): CD20+ T cells have a predominantly Tc1 effector memory phenotype and are expanded in the ascites of patients with ovarian cancer. Oncoimmunology 4: e999536.

50 

Schuh E, Berer K, Mulazzani M, et al. (2016): Features of human CD3+CD20+ T cells. J Immunol 197: 1111-1117.

Copyright: © 2024 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2025 Termedia Sp. z o.o.
Developed by Bentus.