Next Article in Journal
Extracellular Vesicles as Drivers of Immunoinflammation in Atherothrombosis
Previous Article in Journal
Hematopoietic Stem Cell Gene-Addition/Editing Therapy in Sickle Cell Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Differential Expression of CD45RO and CD45RA in Bovine T Cells

Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
*
Author to whom correspondence should be addressed.
Cells 2022, 11(11), 1844; https://doi.org/10.3390/cells11111844
Submission received: 22 April 2022 / Revised: 28 May 2022 / Accepted: 2 June 2022 / Published: 4 June 2022
(This article belongs to the Section Cellular Immunology)

Abstract

:
Effective vaccination induces immune memory to protect animals upon pathogen re-encounter. Despite contradictory reports, bovine memory T cells are identified based on two isoforms of CD45, expression of CD45RO plus exclusion of CD45RA. In this report, we contrasted CD45RA/RO expression on circulatory T cells with IFNγ and IL4 expression induced by a conventional method. To our surprise, 20% of cattle from an enclosed herd did not express CD45RO on T cells without any significant difference on CD45RA expression and IFNγ or IL4 induction. In CD45RO expressing cattle, CD45RA and CD45RO expressions excluded each other, with dominant CD45RO (>90%) expression on gamma delta (γδ) followed by CD4+ (60%) but significantly higher CD45RA expression on CD8+ T cells (about 80%). Importantly, more than 80% of CD45RO expressing CD4+ and CD8+ T cells failed to produce IFNγ and IL-4; however, within the cytokine inducing cells, CD4+ T cells highly expressed CD45RO but those within CD8+ T cells mostly expressed CD45RA. Hence, CD45RO is not ubiquitously expressed in cattle, and rather than with memory phenotype, CD45RA/RO expression are more associated with distinct T cell subtypes.

1. Introduction

Memory T cells mount rapid and robust immune responses and are the hallmark of effective vaccination. The quality and quantity of induced immune memory are therefore critical for evaluating the efficacy of vaccines. Conventionally, memory T cells were detected as cells producing cytokines such as IFNγ and IL4 after brief stimulation in vitro [1,2,3]. However, more recently, memory T cells are often identified based on the inclusion of CD45RO and exclusion of CD45RA on T cells, which is a relatively easy and practical approach for downstream treatments [3,4,5]. In humans and bovine T cells, CD45RA and CD45RO expressions are found to be mutually exclusive, while naïve cells are defined as CD45RA positive (CD45RA+) and CD45RO negative (CD45RO-), memory T cells are CD45RO positive (CD45RO+) and CD45RA negative (CD45RA-) [6,7,8]. The small population of double positive (DP) cells that co-express CD45RA and CD45RO (i.e., CD45RA+/CD45RO+) are believed to reflect the transition stage from naïve to memory induced by pathogen infection or vaccination [9,10,11]. Despite some controversial reports, the conventional memory markers CD45RO and CD45RA are used extensively to detect memory T cells in humans and cattle [5,12,13,14,15,16,17,18,19,20,21,22,23,24,25].
CD45 is a tyrosine phosphatase membrane protein expressed abundantly on the surface of a wide range of immune cells, including T lymphocytes [26,27]. In humans, CD45 is expressed in a combination of multiple isoforms generated through alternative splicing of the CD45 pre-mRNA at exons 4, 5, and 6 [26,28,29]. Inclusion and exclusion of these three exons differentially generate CD45 isoforms, such as the high molecular weight CD45RA and the low molecular weight CD45RO [26,30]. Specifically, the shortest CD45RO transcript is generated through the exclusion of exons (4, 5, and 6) while still retaining 3 and 7, which are shared by all the other isoforms [26,30]. Therefore, selective detection of CD45RO transcript by quantitative PCR (qPCR) is not practical. Instead, due to the highly conserved nature of CD45 protein across the mammalian species [28,31], bovine CD45RO expression is commonly examined at the protein level using a monoclonal antibody (clone # ILA116) in flow cytometry [5,8,24,30,32,33,34,35,36,37,38]. Bembridge et al. (1995) in their seminal research first used this monoclonal antibody (clone #ILA116) to precipitate bovine CD45RO, which had a molecular weight very close to that of humans (180 kDa), and suggested that this isoform could be associated with memory [8]. Subsequently, Sopp et al. (2005) reported that CD45RO+ CD4+ T cells were the dominant producer of IFNγ and IL4 [23]. These reports, along with several others, supported the idea that CD45RO expression is associated with memory phenotype in cattle [8,23,24,39].
In the past few decades, a considerable number of reports in humans and cattle challenged the classical CD45RA/RO paradigm that defines the memory phenotype as CD45RO+ and CD45RA- cells. Specifically, a large number of reports in humans reveal the existence of terminally differentiated CD45RA+ memory T cells both in vitro and in vivo, indicating that memory T cells could be found within both CD45RO+ and CD45RA+ subpopulations [13,16,18,19,21,22]. Interestingly, some data in bovine research also contrast this classical paradigm. For instance, Hagberg et al. (2008) suggested that CD45RO+ CD4+ and CD8+ T cells isolated from immune cattle failed to proliferate in response to the homogenate derived from the Dictyocaulus viviparous parasite [40,41]. Further, memory CD8+ T cells were detected in both CD45RO+ and CD45RA+ fractions, suggesting that CD45RO is not necessarily a memory marker for bovine CD8+ T cells [8,9,37,42,43]. In addition, Guerra Maupome et al. (2019) suggested that CD45RO is not an appropriate marker to detect bovine memory γδ T cells [44]. These contradictory reports warrant re-assessment of the classical paradigm in cattle to verify if CD45RA/RO isoforms are genuinely associated with memory phenotype in bovine T cells.
This project aimed to contrast the CD45RA/RO expression on circulatory T cells with memory T cell subtypes from the same cattle. Our data suggest that the classical CD45RA/RO paradigm may not be accurate in cattle. First, about 20% of cattle (six out of 28) did not express CD45RO. Furthermore, in those expressing CD45RO, the expression of this isoform was not correlated with IFNγ and IL4 induction. Instead, CD45RO and CD45RA clustered differentially in bovine T cell subtypes, suggesting that their expression is more associated with T cell subtypes than with memory phenotype.

2. Materials and Methods

2.1. Cattle

A total of 28 healthy cattle (Wye Angus), including 20 grass-fed and eight grain-fed, aged from one year to 24 months, were used. The closed herd was housed at the Wye Research and Education Center, University of Maryland Experimental Station (Queenstown, MD, USA) [45,46,47,48]. All calves were weaned at six months of age, then randomly assigned to either grain-fed or grass-fed groups. The grain-fed group was kept on a feedlot with a mixture diet [46], and the grass-fed group was maintained on the pasture as reported previously [49]. All the animals were examined monthly to ensure health. Animal Care and Use Protocols were approved by UMD (R-FEB-18-06 and R-JAN-21-02), Institutional Animal Care and Use Committee (IACUC), with the relevant guidelines and regulations.

2.2. Peripheral Blood Mononuclear Cells (PBMCs) Isolation and Stimulation

This procedure was similarly performed as in our previous reports [47,48]. Briefly, fresh blood was collected from the jugular vein using EDTA coated vacutainers (Becton Dickinson Vacutainer Systems, Franklin Lakes, NJ, USA) and transferred to 15 mL conical tubes (Fisher Scientific, Pittsburgh, PA, USA), which were centrifuged for 30 min at 1200× g. Following centrifugation, the buffy coat at the interface was gently collected into a new 15 mL tube and re-suspended with 1× phosphate-buffered saline (PBS) (Fisher Scientific, Fair Lawn, NJ, USA) to 8 mL. Then, it was overlaid with a 5 mL lymphocyte separation medium (LSM) with the density of 1.077 g/mL (Corning, Manassas, VA, USA), followed by centrifugation for 30 min at 900× g with break off. The second buffy coat at the interface was collected and washed twice with PBS. After the last wash, the cell pellet was re-suspended in 5 mL Allos medium (RPMI1640 (Corning, Manassas, VA, USA) plus 10% FBS), and a small aliquot was used for cell counting.
Following the purification, one million cells per sample were aliquoted and re-suspended in 1 mL Allos medium supplied with Brefeldin A (BFA) (BioLegend, San Diego, CA, USA) or cell activation cocktail (CT) (R&D Systems, Minneapolis, MN, USA) that contains monensin sodium salt (1.5 mM), Phorbol 12-myristate 13-acetate (0.0405 mM) and Ionomycin calcium salt (0.67 mM). The cell suspensions were incubated at 37 °C with 5% CO2 for 4 h before intracellular staining.

2.3. Antibodies and Reagents

All the antibodies used in this study are listed in the following tables: primary antibodies (Table 1) and secondary antibodies plus isotypes (Table 2). Staining buffer (SB) is PBS with 2% FBS, and fix solution is 4% paraformaldehyde (W/V) in PBS with pH 7.4. Intracellular staining permeabilization wash buffer (P/W) (BioLegend, San Diego, CA USA) was purchased and used following the manufacturer instructions.

2.4. Flow Cytometry Analysis

For surface staining, cells were stained with primary antibodies (Table 1), followed by secondary antibodies, or fluorescence conjugated antibodies (Table 2) at 4 °C for 25 min, then washed with SB to remove the unbounded antibodies. Cells were further incubated with fix solution (1:1 dilution) for 15 min and washed with SB. Finally, cells were re-suspended in SB before being analyzed in a flow cytometer. For intracellular staining, the cells were stimulated for 4 h as described in 2.2, and then fixed with 2% paraformaldehyde for 15 min. Following two washes, cells were incubated with permeabilization wash buffer (P/W) (BioLegend, San Diego, CA, USA) for additional 15 min at 4 °C. Similar to the surface staining, cells were incubated with antibodies at 4 °C for 25 min. Finally, cells were re-suspended in SB for reading. Staining of isotype and unstained controls was performed using the same method described above. Cells were examined by using FACSCalibur flow cytometer. The Flow cytometer data were analyzed with the FlowJo version 10 (Tree Star, Ashland, OR, USA).

2.5. Statistical Analysis

Statistical analyses were performed using Prism 9 (GraphPad Software, Inc., La Jolla, CA, USA), and details are described in the figure legends. Overall, all data passed the Anderson–Darling normality test and were analyzed via one-way ANOVA with Tukey’s Multiple Comparisons Test. Asterisks indicate statistical significance. * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001. NS indicates not significant.

3. Results

3.1. Bovine Lymphocytes Do Not Always Express CD45RO

Detection of an established memory T cell population is the gold standard for evaluating effectiveness of the vaccines [35,50,51]. In both humans and cattle, memory cells are identified using CD45RO as a marker [23,25,31,52,53]. We examined CD45RO and CD45RA expressions in bovine PBMCs. Almost all of the lymphocytes expressed high levels of either CD45RO(RO+) or CD45RA(RA+), and a small population expressed both at intermediate levels, which are referred to as double positive (DP) cells throughout this manuscript (Figure 1A,B). In general, much more lymphocytes expressed CD45RA than CD45RO (Figure 1B). To our surprise, four out of sixteen cattle did not express CD45RO (designated as RO null) but their CD45RA expression level was similar to those in CD45RO expressing cattle, labeled as CD45RO positive (RO+) (Figure 1A,B). It seems lack of CD45RO isoform does not necessarily affect CD45RA expression. Among the examined cattle, while half of the cattle were raised on pasture (grass-fed), the other half were raised in feedlot (grain-fed) [46,48]. Due to more exposure of grass-fed cattle to the environmental pathogens present in the pasture such as Ostertagia Ostertagi (OO) [46], the frequency of the memory cells in these animals could be greater than in grain-fed counterparts, despite being under the same vaccination procedure [46,54]. The data were further analyzed by comparing the grass-fed cattle with the grain-fed, but no significant difference was observed (Supplementary Figure S1).

3.2. CD45RO and CD45RA Are Expressed Differentially in T Cell Subtypes

There are three major T cell subtypes in cattle, CD4+, CD8+ and gamma delta (γδ) T cells, all of which could differentiate into memory cells, with CD45RO as the marker [8,23,55]. Consistent with previous literature, CD4+ and γδ were the biggest T cell populations in PBMCs, while the CD8+ population was significantly lower than that of CD4+ T cells (Figure 2B) [23,56]. Interestingly, CD45RO and CD45RA expression clustered differently in each T cell subtype with γδ T cells (90%) dominantly expressing CD45RO, followed by CD4+ (around 60%), and CD8+ (around 40%) as shown in Figure 2C. The pattern of CD45RA expression, as expected [8,44], was opposite to CD45RO, lowest in γδ T cells but highest in CD8+ T cells (Figure 2C). It seems that the expression of CD45RA and CD45RO is associated with specific T cell subtypes. With the reason unknown, CD45RO expression was higher in CD4+ from the grass-fed cattle than those from the grain-fed (Supplementary Figure S2B).

3.3. Differential Activation Status in CD45RO+ and CD45RA+ Fractions of CD4+, but Not of CD8+ T Cells

CD25, the α subunit of IL-2 receptor, has been a reliable marker for identifying activated T cells in humans and cattle [48,57,58,59], which is also expressed by regulatory CD4+ T cells in cattle [46]. In the PBMCs from healthy cattle, only a small fraction of CD4+ T cells expressed CD25, thus potentially activated (Figure 3A,B). Majority of the cells in the CD25+ CD4+ T cell subpopulation were expressing CD45RO (RO+), with a smaller fraction expressing CD45RA (RA+) (Figure 3B). The frequency of RA+ cells in CD25+ CD4+ T cells was similar between (RO+) and RO null cattle (Figure 3B). In contrast, the frequency of CD25+ cells was significantly lower in CD8+ than in CD4+ T cells (p < 0.001) (Figure 3B,C and data not shown). CD62L, an adhesion molecule related to trafficking into the lymphoid tissues [60], was expressed at a higher level in CD45RA+ than in the CD45RO+ subpopulation from both CD4+ and CD8+ T cells (Figure 3D,E). The expression of CD62L was not affected by the lack of CD45RO expression in the RO null cattle (Figure 3E).

3.4. Expression of IFNγ and IL4 Is Not Associated with CD45RO in CD4+ T Cells

Induction of IFNγ and or IL4 after brief in vitro stimulation is a conventional method to detect memory T cells in humans and cattle [1,2,3,61]. Using this method, about 15% of the CD4+ T cells expressed IFNγ, whereas a smaller population expressed IL4, plus an even smaller fraction produced both IFNγ and IL4 (Figure 4A,B). Interestingly, the lack of CD45RO expression did not affect the frequencies of these IFNγ or IL4 producing cells (Figure 4B), suggesting CD45RO may not be an exclusive marker for memory CD4+ T cells. To further examine the association of IFNγ and/or IL4 with CD45RO, data were analyzed in two different ways. First, we calculated the percentages of cytokine producing cells within CD45RA+ (RA+) and CD45RO+ (RO+) subpopulations; second, we analyzed the frequencies of CD45RA and CD45RO within the cytokine producing cells. Close to 20% of the RO+ subpopulation produced IFNγ, which was much lower (about 5%) in RA+ (Figure 4D). This trend was similarly reflected in IL4 producing cells but at a lower level, and, and lack of CD45RO expression did not affect the frequency of IFNγ/IL4 production in RO null cattle (Figure 4D). In the second analysis, more than 50% of IFNγ or IL4 producing CD4+ T cells expressed CD45RO, with a significantly small fraction also expressing CD45RA (Figure 4E). There was no significant difference in the frequencies of IFNγ or IL4 producing CD4+ T cells between grass-fed and grain-fed cattle (Supplementary Figure S4A). Additionally, the percentage of CD45RO+ or CD45RA+ cells within IFNγ or IL4 producing populations was also similar (Supplementary Figure S4B). These data suggested that most cytokine-producing CD4+ T cells express CD45RO, consistent with previous reports in cattle [8,23,24,62], but a majority of the CD45RO expressing cells are not producing effector cytokines.

3.5. Expression of IFNγ Is Not Associated with CD45RO in CD8+ T Cells

Vaccination against intracellular pathogens such as viruses induces memory CD8+ T cells, which are critical for rapid control of infections [63,64]. We detected memory CD8+ cells using a similar methodology as applied for identifying memory CD4+ T cells in Figure 4. Unlike in CD4+, CD8+ T cells only produced IFNγ, but essentially no IL4 (Figure 5A,B). This is in agreement with their cytotoxic function instead of the regulatory function defined for CD4+ T cells that expressed both cytokines [64,65]. However, consistent with the results for CD4+ (Figure 4), the lack of CD45RO expression did not affect IFNγ production in CD8+ T cells from RO null cattle (Figure 5B). In CD8+ T cells, CD45RO expressing cells had a significantly higher frequency of IFNγ+ cells than those expressing CD45RA (Figure 5C), which was similar to observations in CD4+ T cells (Figure 4D). However, within IFNγ+ CD8+ T cells, the frequency of CD45RA expressing cells was significantly higher than those expressing CD45RO, opposite to the data in CD4+ T cells (Figure 4E), suggesting that CD45RA/RO expression was different between cytokine-producing CD4+ and CD8+ T cell subtypes. These data further support the notion that CD45RA/RO expression is associated with distinct T cell subtypes.

3.6. Expression of IFNγ and IL4 Is Not Associated with CD45RO in γδ T Cells

γδ T cells have been suggested to be involved in both innate and adaptive immunity, thus their memory has also been identified with CD45RO in cattle [66,67]. Our data revealed that 2 out of 12 cattle were RO null, and, therefore, did not express CD45RO in the γδ T cells (Figure 6B and data not shown). In the RO+ cattle, consistent with Figure 2, more than 90% of γδ T cells expressed CD45RO with majority of them also positive for CD62L (Figure 6A,B). Despite almost all of the γδ T cells (>90%) expressed CD45RO, only a small fraction of them produced IFNγ, suggesting weak or no association between CD45RO and memory phenotype (Figure 6C,D). Among the IFNγ expressing γδ T cells, a small frequency of cells was double positive for IFNγ/IL4, which was not affected by the lack of CD45RO expression in the RO null cattle (Figure 6C and dotted boxes in Figure 6D). Previously, γδ T cells have been reported to express low levels of CD25, [68,69,70], being activated at least partially. In our data, only a small fraction of IFNγ+ and IL4+ cells expressed CD25 (Figure 6E–G), suggesting that these cytokine expressing cells were mostly memory but not activated cells.

4. Discussion

Vaccine induces pathogen-specific memory T cells to protect animals from re-infection. Memory cells in cattle are defined as CD45RO+ and CD45RA- T cells. However, our research reveals that a fraction of cattle does not express CD45RO on T cells but still differentiate into IFNγ and IL4 producing memory cells. Additionally, in those that expressed CD45RO, the clustering of CD45RO and CD45RA isoforms is uniquely related to bovine T cell subtypes rather than the memory phenotype.
The classical paradigm supports that naïve cells are CD45RA+ CD45RO- and memory cells are CD45RO+ CD45RA-. However, some results from humans and cattle, at least partially, contrast this characterization [8,13,16,18,19,21,22,40,41,44]. In fact, multiple publications have suggested that the memory CD4+ and CD8+ T cells could express CD45RA. Therefore, the relevance of CD45RO as a unique memory marker for T cells remains controversial [12,13,14,15,16,17,18,19,20,21,22,71,72,73,74]. In support to these reports, our data demonstrated that, in a fraction (about 20%) of healthy cattle, T cells failed to express CD45RO but expressed a normal level of CD45RA. Interestingly, the frequency of IFNγ and IL4 producing memory cells in RO null cattle was not different from those in RO+ cattle. This pattern was also consistent within cytokines producing memory CD4+, CD8+, and γδ T cell subtypes, indicating that in the RO null group, the memory T cell differentiation was not affected by the absence of CD45RO expression. Furthermore, we noticed that the average frequency of CD45RA+ T cells in the RO+ cattle was similar to those in the RO null, which suggests that CD45RA expression might not depend on CD45RO. In the RO+ cattle, only less than 20% of CD45RO+ CD4+ T cells produced IFNγ, which means that more than 80% of CD45RO+ cells did not produce effector cytokine upon in vitro stimulation. These experimental observations strongly suggest that using CD45RO as a marker for memory T cells in cattle might be overestimating and even misleading.
The classical paradigm assumes that switching of CD45 isoforms from CD45RA to CD45RO is an essential process associated with the memory T cell differentiation [7,8,53,75,76,77]. However, the data from the RO null cattle suggest that, at least in a fraction of cattle, switching of CD45 isoforms is not required for the induction of memory T cells. We noticed that the average percentage of CD45RA+ lymphocytes in the RO null cattle was similar to that in RO+ cattle. Further, the average frequencies of CD45RA+ cells within CD4+ and CD8+ T cell subtypes in the RO+ cattle were also close to those in the RO null. The evidence suggests that the isoform switch might not be necessary to induce memory T cell population in all cattle. Therefore, the relevance of CD45RO as a signature marker for memory bovine T cells is questionable.
Our data demonstrate that CD45RA/RO expression pattern differs in distinct bovine T cell subtypes. While the proportion of CD45RA+ cells in the total lymphocytes was always higher than that of CD45RO+ cells, the pattern varied significantly within the distinct bovine T cell subtypes. In γδ (more than 90%) and CD4+ (60%), CD45RO expression was dominant; however, in CD8+ T cells, CD45RO expression was relatively low (about 30%) but CD45RA expression was significantly high. These data indicate a distinct pattern of CD45RA/RO clustering in bovine T cell subtypes. The tendency of our data is in partial agreement with several previous reports, where γδ T cells were extensively CD45RO+, CD4+ T cells were predominantly CD45RO+, but CD8+ T cells were mostly CD45RO- [8,23,44]. Interestingly, this pattern has been similarly reported in human T cell subtypes [78,79,80,81,82]. Therefore, the distinct pattern of isomer clustering reflects that the CD45RA/RO expression is strongly associated with bovine T cell subtypes.
We noticed that even within the cytokine producing T cell population, the CD45RA/RO expression pattern was associated with distinct T cell subtypes, which is in agreement with several previous reports. It has been reported that the antigen-specific memory CD4+ population is within the CD45RO+ population [5,8,23,24,38,39], but that of CD8+ T cells contains both CD45RA+ and CD45RO+ fractions [8,34,35,37]. Previously, cytokine-producing ovalbumin (OVA)-specific memory bovine CD4+ T cells were found within the CD45RO+ sub-population, which is in line with another report, where the CD45RO+ fraction of the CD4+ T cells dominantly produced IFN-γ and IL4 in both blood and lymph nodes [8,23]. In contrast, a different pattern has been reported in the context of bovine CD8+ T cells. While the S. uberis and BCG-specific memory CD8+ T cells were mostly found within CD45RO+ population, those specific to Theileria parva were under both CD45RA+ and CD45RO+ fractions [8,34,35,37]. Our analysis suggests that the memory bovine T cells could be both CD45RO+ and CD45RA+ with the dominant expression of CD45RO in the CD4+ T cells but CD45RA in the CD8+ subtype. Therefore, the association between CD45RA/RO expression and bovine T cell subtypes was also reflected within IFNγ and IL-4 inducing cells.
As CD45RA/RO expression on T cells is more related to distinct subtypes than the memory phenotype, we speculate that their expression pattern could be associated with T cell subtype-specific functions, which has been reported in humans [72,83,84,85]. For instance, Dengue virus (DENV) specific CD45RA+ CD4+ T cells, Epstein-Barr virus (EBV) specific CD45RA+ CD8+ T cell, and M tuberculosis as well as cytomegalovirus (CMV) specific CD8+ γδ T cells expressing CD45RA demonstrated cytotoxic activity [72,83,84,85]. Additionally, CD45RA+ but not CD45RA- CD8+ T cells isolated from HIV-1 infected patients selectively demonstrated cytotoxicity under in vitro assay, indicating that killing ability could be related to CD45RA expression on these cells [74]. In contrast, CD45RO expression in the CD4+ T cells could be more related to immunoregulatory functions such as helping B cells to produce immunoglobulins. CD45RO+ CD4+ T cells showed an improved ability to adhere with immortalized B cell lines than the CD45RA+ cells [86]. Furthermore, CD45RO+ CD4+ T cell isolated from the blood of healthy humans stimulated B cells to produce high levels of IgM and IgG in vitro, which were drastically reduced when CD45RO+ CD4+ T cells were removed from the culture [11,87]. These lines of evidence provide a new perspective for interpreting the distinct pattern of CD45RA/RO clustering on T cells that their expression could be related to the distinct T cell subtypes and their specific effector functions. Therefore, more research is required to appropriately understand how distinct CD45RA/RO expression is related to the cell-specific functions in cattle. Nevertheless, more biomarkers are warranted to precisely identify the memory bovine T cell population in cattle.

5. Conclusions

Our data contrast the classical CD45RA/RO paradigm and suggest that CD45RO expression on CD45RA- T cells is not strongly correlated with memory identification. Rather than with memory phenotype, the pattern of CD45RA/RO distribution on the T cells is associated more with distinct T cell subtypes. Therefore, future research should target to identify novel markers for memory T cell population and also define the function of CD45RA/RO isoforms in cattle.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cells11111844/s1, Figure S1: CD45RO is not expressed by lymphocytes in some cattle; Figure S2: CD45RO and CD45RA are expressed differentially in T cell subtypes; Figure S3: Activation status varies between CD45RO+ and CD45RA+ subpopulations in CD4+, but not in CD8+ T cells; Figure S4: Expression of IFNg and IL-4 is not associated with CD45RO in CD4+ T cells; Figure S5: Expression of IFNg is not associated with CD45RO in CD8+ T cells; Figure S6: Expression of IFNg and IL4 is not associated with CD45RO in GD T cells.

Author Contributions

Z.X. conceived the study. Z.X. designed and coordinated the study. Z.X., A.K., A.H. and L.L. performed, and analyzed the experiments. Z.X., A.K., A.H. and L.L. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Research was supported by USDA NIFA Grant 2016-67015-24948 (to Z.X.) and Grant 2019-67015-29831 (to Z.X.), the Jorgensen Foundation (to Z.X.), and MAES program in University of Maryland (to Z.X.).

Institutional Review Board Statement

These studies have been reviewed and approved by the Institutional Animal Care and Use Committee at the University of Maryland (R-FEB-18-06 approved on 2 May 2018 and R-Jan-21-02 approved on 1 December 2021).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data are contained within the article and Supplemental File.

Acknowledgments

The authors are grateful to Ken Class, Edward Draper of UMD for their excellent technical assistance, Wenbin Tuo for scientific discussion and sharing some reagents.

Conflicts of Interest

The authors declare no competing interest.

References

  1. Picker, L.; Singh, M.; Zdraveski, Z.; Treer, J.; Waldrop, S.; Bergstresser, P.; Maino, V. Direct demonstration of cytokine synthesis heterogeneity among human memory/effector T cells by flow cytometry. Blood 1995, 86, 1408–1419. [Google Scholar] [CrossRef] [PubMed]
  2. Baran, J.; Kowalczyk, D.; Ozóg, M.; Zembala, M. Three-color flow cytometry detection of intracellular cytokines in peripheral blood mononuclear cells: Comparative analysis of phorbol myristate acetate-ionomycin and phytohemagglutinin stimulation. ASM J. Clin. Diagn. Lab. Immunol. 2001, 8, 303–313. [Google Scholar] [CrossRef] [Green Version]
  3. Phetsouphanh, C.; Zaunders, J.J.; Kelleher, A.D. Detecting antigen-specific T cell responses: From bulk populations to single cells. Int. J. Mol. Sci. 2015, 16, 18878–18893. [Google Scholar] [CrossRef] [PubMed]
  4. Suni, M.A.; Picker, L.J.; Maino, V.C. Detection of antigen-specific T cell cytokine expression in whole blood by flow cytometry. J. Immunol. Methods 1998, 212, 89–98. [Google Scholar] [CrossRef]
  5. Maggioli, M.F.; Palmer, M.V.; Thacker, T.C.; Vordermeier, H.M.; Waters, W.R. Characterization of effector and memory T cell subsets in the immune response to bovine tuberculosis in cattle. PLoS ONE 2015, 10, e0122571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Akbar, A.; Terry, L.; Timms, A.; Beverley, P.; Janossy, G. Unidirectional phenotypic changes within the T200 complex during activation of T cells. J. Immunol. 1988, 140, 2171. [Google Scholar]
  7. Sanders, M.E.; Makgoba, M.W.; Shaw, S. Human naive and memory T cells: Reinterpretation of helper-inducer and suppressor-inducer subsets. Immunol. Today 1988, 9, 195–199. [Google Scholar] [CrossRef]
  8. Bembridge, G.; MacHugh, N.; McKeever, D.; Awino, E.; Sopp, P.; Collins, R.; Gelder, K.; Howard, C. CD45RO expression on bovine T cells: Relation to biological function. Immunology 1995, 86, 537. [Google Scholar]
  9. Stabel, J.R.; Kimura, K.; Robbe-Austerman, S. Augmentation of secreted and intracellular gamma interferon following johnin purified protein derivative sensitization of cows naturally infected with Mycobacterium avium subsp. paratuberculosis. J. Vet. Diagn. Investig. 2007, 19, 43–51. [Google Scholar] [CrossRef] [Green Version]
  10. McInnes, E.; Sopp, P.; Howard, C.; Taylor, G. Phenotypic analysis of local cellular responses in calves infected with bovine respiratory syncytial virus. Immunology 1999, 96, 396. [Google Scholar] [CrossRef] [PubMed]
  11. Duni, A.; Markopoulos, G.S.; Mallioras, I.; Pappas, H.; Pappas, E.; Koutlas, V.; Tzalavra, E.; Baxevanos, G.; Priska, S.; Gartzonika, K. The Humoral Immune Response to BNT162b2 Vaccine Is Associated With Circulating CD19+ B Lymphocytes and the Naïve CD45RA to Memory CD45RO CD4+ T Helper Cells Ratio in Hemodialysis Patients and Kidney Transplant Recipients. Front. Immunol. 2021, 12, 760249. [Google Scholar] [CrossRef]
  12. Michie, C.A.; McLean, A.; Alcock, C.; Beverley, P.C. Lifespan of human lymphocyte subsets defined by CD45 isoforms. Nature 1992, 360, 264–265. [Google Scholar] [CrossRef]
  13. Wills, M.R.; Carmichael, A.J.; Weekes, M.P.; Mynard, K.; Okecha, G.; Hicks, R.; Sissons, J.P. Human virus-specific CD8+ CTL clones revert from CD45ROhigh to CD45RAhigh in vivo: CD45RAhighCD8+ T cells comprise both naive and memory cells. J. Immunol. 1999, 162, 7080–7087. [Google Scholar]
  14. Callan, M.; Tan, L.; Annels, N.; Ogg, G.; Wilson, J.; O’callaghan, C.; Steven, N.; McMichael, A.; Rickinson, A. Direct visualization of antigen-specific CD8+ T cells during the primary immune response to Epstein-Barr virus in vivo. J. Exp. Med. 1998, 187, 1395–1402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Khan, N.; Shariff, N.; Cobbold, M.; Bruton, R.; Ainsworth, J.A.; Sinclair, A.J.; Nayak, L.; Moss, P.A. Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals. J. Immunol. 2002, 169, 1984–1992. [Google Scholar] [CrossRef] [Green Version]
  16. Wills, M.R.; Okecha, G.; Weekes, M.P.; Gandhi, M.K.; Sissons, P.J.; Carmichael, A.J. Identification of naive or antigen-experienced human CD8+ T cells by expression of costimulation and chemokine receptors: Analysis of the human cytomegalovirus-specific CD8+ T cell response. J. Immunol. 2002, 168, 5455–5464. [Google Scholar] [CrossRef] [Green Version]
  17. Pinto, L.; Covas, M.J.; Victorino, R.M. Loss of CD45RA and gain of CD45RO after in vitro activation of lymphocytes from HIV-infected patients. Immunology 1991, 73, 147–150. [Google Scholar] [PubMed]
  18. Gattinoni, L.; Lugli, E.; Ji, Y.; Pos, Z.; Paulos, C.M.; Quigley, M.F.; Almeida, J.R.; Gostick, E.; Yu, Z.; Carpenito, C.; et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 2011, 17, 1290–1297. [Google Scholar] [CrossRef]
  19. Ahmed, R.; Roger, L.; Del Amo, P.C.; Miners, K.L.; Jones, R.E.; Boelen, L.; Fali, T.; Elemans, M.; Zhang, Y.; Appay, V. Human stem cell-like memory T cells are maintained in a state of dynamic flux. Cell Rep. 2016, 17, 2811–2818. [Google Scholar] [CrossRef] [Green Version]
  20. Lee, H.; Stabel, J.; Kehrli, M., Jr. Cytokine gene expression in ileal tissues of cattle infected with Mycobacterium paratuberculosis. Vet. Immunol. Immunopathol. 2001, 82, 73–85. [Google Scholar] [CrossRef]
  21. Hong, H.; Gu, Y.; Sheng, S.Y.; Lu, C.G.; Zou, J.Y.; Wu, C.Y. The Distribution of Human Stem Cell–like Memory T Cell in Lung Cancer. J. Immunother. 2016, 39, 233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Jung, J.H.; Rha, M.-S.; Sa, M.; Choi, H.K.; Jeon, J.H.; Seok, H.; Park, D.W.; Park, S.-H.; Jeong, H.W.; Choi, W.S. SARS-CoV-2-specific T cell memory is sustained in COVID-19 convalescent patients for 10 months with successful development of stem cell-like memory T cells. Nat. Commun. 2021, 12, 1–12. [Google Scholar] [CrossRef]
  23. Sopp, P.; Howard, C.J. IFN gamma and IL-4 production by CD4, CD8 and WC1 gamma delta TCR(+) T cells from cattle lymph nodes and blood. Vet. Immunol. Immunopathol. 2001, 81, 85–96. [Google Scholar] [CrossRef]
  24. Frie, M.C.; Sporer, K.R.; Kirkpatrick, B.W.; Coussens, P.M. T and B cell activation profiles from cows with and without Johne’s disease in response to in vitro stimulation with Mycobacterium avium subspecies paratuberculosis. Vet. Immunol. Immunopathol. 2017, 193, 50–56. [Google Scholar] [CrossRef] [PubMed]
  25. Machura, E.; Mazur, B.; Pieniążek, W.; Karczewska, K. Expression of naive/memory (CD45RA/CD45RO) markers by peripheral blood CD4+ and CD8+ T cells in children with asthma. Arch. Immunol. Et Ther. Exp. 2008, 56, 55–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Hermiston, M.L.; Xu, Z.; Weiss, A. CD45: A critical regulator of signaling thresholds in immune cells. Annu. Rev. Immunol. 2003, 21, 107–137. [Google Scholar] [CrossRef] [PubMed]
  27. Tonks, N.K.; Charbonneau, H.; Diltz, C.D.; Fischer, E.H.; Walsh, K.A. Demonstration that the leukocyte common antigen CD45 is a protein tyrosine phosphatase. Biochemistry 1988, 27, 8695–8701. [Google Scholar] [CrossRef]
  28. Trowbridge, I.S.; Thomas, M.L. CD45: An emerging role as a protein tyrosine phosphatase required for lymphocyte activation and development. Annu. Rev. Immunol. 1994, 12, 85–116. [Google Scholar] [CrossRef]
  29. Lynch, K.W. Consequences of regulated pre-mRNA splicing in the immune system. Nat. Rev. Immunol. 2004, 4, 931–940. [Google Scholar] [CrossRef]
  30. Jonsson, N.N.; Cox, D.K.; Piper, E.K.; Valdivieso, E.F.M.; Constantinoiu, C.; Jackson, L.A.; Stear, M.J.; Ross, E.M.; Tabor, A.E. Allelic Variation in Protein Tyrosine Phosphatase Receptor Type-C in Cattle Influences Erythrocyte, Leukocyte and Humoral Responses to Infestation With the Cattle Tick Rhipicephalus australis. Front. Immunol. 2021, 12, 675979:1–675979:10. [Google Scholar] [CrossRef]
  31. Valentine M, Song K, Maresh GA, Mack H, Huaman MC, Polacino P, Ho O, Cristillo A, Kyung Chung H, Hu SL, Pincus SH. Expression of the memory marker CD45RO on helper T cells in macaques. PLoS One. 2013, 8, e73969.
  32. Ballingall, K.T.; Waibochi, L.; Holmes, E.C.; Woelk, C.H.; MacHugh, N.D.; Lutje, V.; McKeever, D.J. The CD45 locus in cattle: Allelic polymorphism and evidence for exceptional positive natural selection. Immunogenetics 2001, 52, 276–283. [Google Scholar] [CrossRef] [PubMed]
  33. Blunt, L.; Hogarth, P.J.; Kaveh, D.A.; Webb, P.; Villarreal-Ramos, B.; Vordermeier, H.M. Phenotypic characterization of bovine memory cells responding to mycobacteria in IFNγ enzyme linked immunospot assays. Vaccine 2015, 33, 7276–7282. [Google Scholar] [CrossRef] [Green Version]
  34. Hogg, A.E.; Parsons, K.; Taylor, G.; Worth, A.; Beverley, P.; Howard, C.J.; Villarreal-Ramos, B. Characterization of age-related changes in bovine CD8+ T-cells. Vet. Immunol. Immunopathol. 2011, 140, 47–54. [Google Scholar] [CrossRef]
  35. Elnaggar, M.M.; Knowles, D.P.; Davis, W.C.; Fry, L.M. Flow Cytometric Analysis of the Cytotoxic T-Cell Recall Response to Theileria parva in Cattle Following Vaccination by the Infection and Treatment Method. Vet. Sci. 2021, 8, 114. [Google Scholar] [CrossRef] [PubMed]
  36. Endsley, J.J.; Endsley, M.A.; Estes, D.M. Bovine natural killer cells acquire cytotoxic/effector activity following activation with IL-12/15 and reduce Mycobacterium bovis BCG in infected macrophages. J. Leukoc. Biol. 2006, 79, 71–79. [Google Scholar] [CrossRef]
  37. Denis, M.; Lacy-Hulbert, S.J.; Buddle, B.M.; Williamson, J.H.; Wedlock, D.N. Streptococcus uberis-specific T cells are present in mammary gland secretions of cows and can be activated to kill S. uberis. Vet. Res. Commun. 2011, 35, 145–156. [Google Scholar] [CrossRef]
  38. Mitoma, S.; Carr, B.V.; Harvey, Y.; Moffat, K.; Sekiguchi, S.; Charleston, B.; Norimine, J.; Seago, J. The detection of long-lasting memory foot-and-mouth disease (FMD) virus serotype O-specific CD4+ T cells from FMD-vaccinated cattle by bovine major histocompatibility complex class II tetramer. Immunology 2021, 164, 266. [Google Scholar] [CrossRef]
  39. Silflow, R.M.; Degel, P.M.; Harmsen, A.G. Bronchoalveolar immune defense in cattle exposed to primary and secondary challenge with bovine viral diarrhea virus. Vet. Immunol. Immunopathol. 2005, 103, 129–139. [Google Scholar] [CrossRef]
  40. Hagberg, M.; Lundén, A.; Höglund, J.; Morrison, D.; Waller, K.P.; Wattrang, E. Characterization of bovine lymphocytes stimulated in vitro by Dictyocaulus viviparus homogenate. Parasite Immunol. 2008, 30, 342–353. [Google Scholar] [CrossRef]
  41. Hagberg, M. Immune Cell Responses to the Cattle Lungworm, Dictyocaulus Viviparus. Ph.D. Thesis, Swedish University of Agricultural Sciences, Uppsala, Sweden, 2008. Available online: https://pub.epsilon.slu.se/1769/1/200837_Kappan_med_bildtext.pdf (accessed on 10 March 2022).
  42. Hogg, A.E.; Worth, A.; Beverley, P.; Howard, C.J.; Villarreal-Ramos, B. The antigen-specific memory CD8+ T-cell response induced by BCG in cattle resides in the CD8+ γ/δTCR− CD45RO+ T-cell population. Vaccine 2009, 27, 270–279. [Google Scholar] [CrossRef] [PubMed]
  43. Howard, C.J.; Sopp, P.; Parsons, K.R.; McKeever, D.J.; Taracha, E.L.; Jones, B.V.; MacHugh, N.D.; Morrison, W.I. Distinction of naive and memory BoCD4 lymphocytes in calves with a monoclonal antibody, CC76, to a restricted determinant of the bovine leukocyte-common antigen, CD45. Eur. J. Immunol. 1991, 21, 2219–2226. [Google Scholar] [CrossRef] [PubMed]
  44. Guerra-Maupome, M.; Palmer, M.V.; Waters, W.R.; McGill, J.L. Characterization of gammadelta T Cell Effector/Memory Subsets Based on CD27 and CD45R Expression in Response to Mycobacterium bovis Infection. Immunohorizons 2019, 3, 208–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Zhao, C.; Tian, F.; Yu, Y.; Luo, J.; Hu, Q.; Bequette, B.J.; Baldwin VI, R.L.; Liu, G.; Zan, L.; Scott Updike, M. Muscle transcriptomic analyses in Angus cattle with divergent tenderness. Mol. Biol. Rep. 2012, 39, 4185–4193. [Google Scholar] [CrossRef]
  46. Tuo, W.; Li, L.; Lv, Y.; Carrillo, J.; Brown, D.; Davis, W.C.; Song, J.; Zarlenga, D.; Xiao, Z. Abomasal mucosal immune responses of cattle with limited or continuous exposure to pasture-borne gastrointestinal nematode parasite infection. Vet. Parasitol. 2016, 229, 118–125. [Google Scholar] [CrossRef] [Green Version]
  47. Mendez, J.; Sun, D.; Tuo, W.; Xiao, Z. Bovine neutrophils form extracellular traps in response to the gastrointestinal parasite Ostertagia ostertagi. Sci. Rep. 2018, 8, 1–12. [Google Scholar] [CrossRef]
  48. Li, L.; Si, H.; Wu, S.-W.; Mendez, J.O.; Zarlenga, D.; Tuo, W.; Xiao, Z. Characterization of IL-10-producing neutrophils in cattle infected with Ostertagia ostertagi. Sci. Rep. 2019, 9, 1–14. [Google Scholar] [CrossRef] [Green Version]
  49. Carrillo, J.A.; He, Y.; Li, Y.; Liu, J.; Erdman, R.A.; Sonstegard, T.S.; Song, J. Integrated metabolomic and transcriptome analyses reveal finishing forage affects metabolic pathways related to beef quality and animal welfare. Sci. Rep. 2016, 6, 1–16. [Google Scholar] [CrossRef]
  50. Panagioti, E.; Klenerman, P.; Lee, L.N.; Van Der Burg, S.H.; Arens, R. Features of effective T cell-inducing vaccines against chronic viral infections. Front. Immunol. 2018, 9, 276. [Google Scholar] [CrossRef] [Green Version]
  51. Flaxman, A.; Ewer, K.J. Methods for measuring T-cell memory to vaccination: From mouse to man. Vaccines 2018, 6, 43. [Google Scholar] [CrossRef] [Green Version]
  52. Craig, W.; Poppema, S.; Little, M.T.; Dragowska, W.; Lansdorp, P.M. CD45 isoform expression on human haemopoietic cells at different stages of development. Br. J. Haematol. 1994, 88, 24–30. [Google Scholar] [CrossRef] [PubMed]
  53. Clement, L.T. Isoforms of the CD45 common leukocyte antigen family: Markers for human T-cell differentiation. J. Clin. Immunol. 1992, 12, 1–10. [Google Scholar] [CrossRef] [PubMed]
  54. Kumar, N.; Rao, T.K.S.; Varghese, A.; Rathor, V.S. Internal parasite management in grazing livestock. J. Parasit. Dis. 2013, 37, 151–157. [Google Scholar] [CrossRef] [Green Version]
  55. Blumerman, S.L.; Herzig, C.T.; Baldwin, C.L. WC1+ γδ T cell memory population is induced by killed bacterial vaccine. Eur. J. Immunol. 2007, 37, 1204–1216. [Google Scholar] [CrossRef] [PubMed]
  56. Kulberg, S.; Boysen, P.; Storset, A.K. Reference values for relative numbers of natural killer cells in cattle blood. Dev. Comp. Immunol. 2004, 28, 941–948. [Google Scholar] [CrossRef]
  57. Xiao, Z.; Kandel, A.; Li, L. Synergistic Activation of Bovine CD4+ T Cells by Neutrophils and IL-12. Pathogens 2021, 10, 694. [Google Scholar] [CrossRef]
  58. Liao, W.; Lin, J.-X.; Leonard, W.J. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity 2013, 38, 13–25. [Google Scholar] [CrossRef] [Green Version]
  59. Nelson, B.H. IL-2, regulatory T cells, and tolerance. J. Immunol. 2004, 172, 3983–3988. [Google Scholar] [CrossRef] [Green Version]
  60. Schuster, K.; Gadiot, J.; Andreesen, R.; Mackensen, A.; Gajewski, T.F.; Blank, C. Homeostatic proliferation of naïve CD8+ T cells depends on CD62L/L-selectin-mediated homing to peripheral LN. Eur. J. Immunol. 2009, 39, 2981–2990. [Google Scholar] [CrossRef]
  61. Waldrop, S.L.; Pitcher, C.J.; Peterson, D.M.; Maino, V.C.; Picker, L.J. Determination of antigen-specific memory/effector CD4+ T cell frequencies by flow cytometry: Evidence for a novel, antigen-specific homeostatic mechanism in HIV-associated immunodeficiency. J. Clin. Investig. 1997, 99, 1739–1750. [Google Scholar] [CrossRef] [Green Version]
  62. Totté, P.; Duperray, C.; Dedieu, L. CD62L defines a subset of pathogen-specific bovine CD4 with central memory cell characteristics. Dev. Comp. Immunol. 2010, 34, 177–182. [Google Scholar] [CrossRef] [PubMed]
  63. Xiao, Z.; Casey, K.A.; Jameson, S.C.; Curtsinger, J.M.; Mescher, M.F. Programming for CD8 T cell memory development requires IL-12 or type I IFN. J. Immunol. 2009, 182, 2786–2794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Mescher, M.F.; Curtsinger, J.M.; Agarwal, P.; Casey, K.A.; Gerner, M.; Hammerbeck, C.D.; Popescu, F.; Xiao, Z. Signals required for programming effector and memory development by CD8+ T cells. Immunol. Rev. 2006, 211, 81–92. [Google Scholar] [CrossRef] [PubMed]
  65. Kandel, A.; Masello, M.; Xiao, Z. CD4+ T Cell Responses to Pathogens in Cattle; Abubakar, M., Ed.; IntechOpen: London, UK, 2021; Available online: https://www.intechopen.com/chapters/78918 (accessed on 18 June 2021).
  66. Holtmeier, W.; Kabelitz, D. gammadelta T cells link innate and adaptive immune responses. Chem. Immunol. Allergy 2005, 86, 151–183. [Google Scholar] [CrossRef]
  67. Baldwin, C.L.; Yirsaw, A.; Gillespie, A.; Le Page, L.; Zhang, F.; Damani-Yokota, P.; Telfer, J.C. γδ T cells in livestock: Responses to pathogens and vaccine potential. Transbound. Emerg. Dis. 2020, 67, 119–128. [Google Scholar] [CrossRef] [PubMed]
  68. Waters, W.; Rahner, T.; Palmer, M.; Cheng, D.; Nonnecke, B.; Whipple, D. Expression of L-selectin (CD62L), CD44, and CD25 on activated bovine T cells. Infect. Immun. 2003, 71, 317–326. [Google Scholar] [CrossRef] [Green Version]
  69. Toka, F.N.; Kenney, M.A.; Golde, W.T. Rapid and transient activation of γδ T cells to IFN-γ production, NK cell-like killing, and antigen processing during acute virus infection. J. Immunol. 2011, 186, 4853–4861. [Google Scholar] [CrossRef]
  70. Phalke, S.P.; Chiplunkar, S.V. Activation status of γδ T cells dictates their effect on osteoclast generation and bone resorption. Bone Rep. 2015, 3, 95–103. [Google Scholar] [CrossRef] [Green Version]
  71. Tian, Y.; Babor, M.; Lane, J.; Schulten, V.; Patil, V.S.; Seumois, G.; Rosales, S.L.; Fu, Z.; Picarda, G.; Burel, J. Unique phenotypes and clonal expansions of human CD4 effector memory T cells re-expressing CD45RA. Nat. Commun. 2017, 8, 1–13. [Google Scholar] [CrossRef]
  72. Carrasco, J.; Godelaine, D.; Van Pel, A.; Boon, T.; van der Bruggen, P. CD45RA on human CD8 T cells is sensitive to the time elapsed since the last antigenic stimulation. Blood 2006, 108, 2897–2905. [Google Scholar] [CrossRef]
  73. Faint, J.M.; Annels, N.E.; Curnow, S.J.; Shields, P.; Pilling, D.; Hislop, A.D.; Wu, L.; Akbar, A.N.; Buckley, C.D.; Moss, P.A. Memory T cells constitute a subset of the human CD8+ CD45RA+ pool with distinct phenotypic and migratory characteristics. J. Immunol. 2001, 167, 1473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hamann, D.; Baars, P.A.; Rep, M.H.; Hooibrink, B.; Kerkhof-Garde, S.R.; Klein, M.R.; Lier, R.A.v. Phenotypic and functional separation of memory and effector human CD8+ T cells. J. Exp. Med. 1997, 186, 1407–1418. [Google Scholar] [CrossRef] [PubMed]
  75. Deans, J.; Boyd, A.; Pilarski, L. Transitions from high to low molecular weight isoforms of CD45 (T200) involve rapid activation of alternate mRNA splicing and slow turnover of surface CD45R. J. Immunol. 1989, 143, 1233–1238. [Google Scholar]
  76. Birkeland, M.L.; Johnson, P.; Trowbridge, I.S.; Puré, E. Changes in CD45 isoform expression accompany antigen-induced murine T-cell activation. Proc. Natl. Acad. Sci. USA 1989, 86, 6734–6738. [Google Scholar] [CrossRef] [Green Version]
  77. Akbar, A.; Terry, L.; Timms, A.; Beverley, P.; Janossy, G. Loss of CD45R and gain of UCHL1 reactivity is a feature of primed T cells. J. Immunol. 1988, 140, 2171–2178. [Google Scholar] [PubMed]
  78. Zola, H.; Flego, L.; Macardle, P.; Donohoe, P.; Ranford, J.; Roberton, D. The CD45RO (p180, UCHL1) marker: Complexity of expression in peripheral blood. Cell. Immunol. 1992, 145, 175–186. [Google Scholar] [CrossRef]
  79. Prince, H.E.; York, J.; Jensen, E.R. Phenotypic comparison of the three populations of human lymphocytes defined by CD45RO and CD45RA expression. Cell. Immunol. 1992, 145, 254–262. [Google Scholar] [CrossRef]
  80. Sathaliyawala, T.; Kubota, M.; Yudanin, N.; Turner, D.; Camp, P.; Thome, J.J.; Bickham, K.L.; Lerner, H.; Goldstein, M.; Sykes, M. Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity 2013, 38, 187–197. [Google Scholar] [CrossRef] [Green Version]
  81. Yang, W.; Jia, X.; Su, Y.; Li, Q. Immunophenotypic characterization of CD45RO+ and CD45RA+ T cell subsets in peripheral blood of peripheral T cell lymphoma patients. Cell Biochem. Biophys. 2014, 70, 993–997. [Google Scholar] [CrossRef]
  82. Qin, Y.; van den Noort, S.; Kurt, J.; Gupta, S. Dual expression of CD45RA and CD45RO isoforms on myelin basic protein-specific CD4+ T-cell lines in multiple sclerosis. J. Clin. Immunol. 1993, 13, 152–161. [Google Scholar] [CrossRef]
  83. Chowdhury, R.R.; Valainis, J.R.; Kask, O.; Ohanyan, M.; Sun, M.; Huang, H.; Dubey, M.; von Boehmer, L.; Sola, E.; Huang, X. The role of antigen recognition in the γδ T cell response at the controlled stage of M. tuberculosis infection. bioRxiv 2021, 460324:1–460324:56. [Google Scholar]
  84. Sallusto, F.; Geginat, J.; Lanzavecchia, A. Central memory and effector memory T cell subsets: Function, generation, and maintenance. Annu. Rev. Immunol. 2004, 22, 745–763. [Google Scholar] [CrossRef] [PubMed]
  85. Gaballa, A.; Arruda, L.; Rådestad, E.; Uhlin, M. CD8+ γδ T cells are more frequent in CMV seropositive bone marrow grafts and display phenotype of an adaptive immune response. Stem Cells Int. 2019, 2019, 6348060:1–6348060:13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Lecomte, O.; Fischer, A. Antigen-independent adhesion of CD45RA (naive) and CD45RO (memory) CD4 T cells to B cells. Int. Immunol. 1992, 4, 191–196. [Google Scholar] [CrossRef]
  87. Frolova, E.; Scott, C.; Jones, R. CD45RO+ T-cells immunoregulate spontaneous in vitro immunoglobulin production by normal and chronic lymphocytic leukaemia B-cells. Leuk. Lymphoma 1995, 18, 103–111. [Google Scholar] [CrossRef]
Figure 1. CD45RO is not expressed by lymphocytes in some cattle. Lymphocytes were gated in PBMCs and examined for expression of CD45RO and CD45RA. (A) Gating strategy for lymphocytes; (B) Comparison of CD45RO and CD45RA expression in lymphocytes. Data were pooled together from two experiments and expressed as mean of 12 CD45RO positive (RO+) cattle samples, or four CD45RO negative (RO null) cattle with standard deviation (SD). Data in BLUE will indicate samples from RO null cattle in the rest of figures. Q1: lymphocytes only expressing high level of CD45RO, but not CD45RA, designated as RO+; Q2: lymphocytes both CD45RO and CD45RA, designated as double positive (DP). Q3: lymphocytes only expressing high level of CD45RA, but not CD45RO, designated as RA+. Background in isotype control was subtracted from all the data in B. Data were analyzed by one-way ANOVA with Tukey’s Multiple Comparisons Test. Asterisks indicate statistical significance. **** p < 0.0001. NS: not significant. The same analysis will be performed in the rest of figures unless mentioned otherwise.
Figure 1. CD45RO is not expressed by lymphocytes in some cattle. Lymphocytes were gated in PBMCs and examined for expression of CD45RO and CD45RA. (A) Gating strategy for lymphocytes; (B) Comparison of CD45RO and CD45RA expression in lymphocytes. Data were pooled together from two experiments and expressed as mean of 12 CD45RO positive (RO+) cattle samples, or four CD45RO negative (RO null) cattle with standard deviation (SD). Data in BLUE will indicate samples from RO null cattle in the rest of figures. Q1: lymphocytes only expressing high level of CD45RO, but not CD45RA, designated as RO+; Q2: lymphocytes both CD45RO and CD45RA, designated as double positive (DP). Q3: lymphocytes only expressing high level of CD45RA, but not CD45RO, designated as RA+. Background in isotype control was subtracted from all the data in B. Data were analyzed by one-way ANOVA with Tukey’s Multiple Comparisons Test. Asterisks indicate statistical significance. **** p < 0.0001. NS: not significant. The same analysis will be performed in the rest of figures unless mentioned otherwise.
Cells 11 01844 g001
Figure 2. CD45RO and CD45RA are expressed differentially in T cell subtypes. Lymphocytes were gated in PBMCs and examined for the expression of CD45RO and CD45RA in different T cell subtypes. (A) Gating strategy for T cell subtypes and expression of CD45RO and CD45RA; (B) Comparison of frequency of T cell subtypes in PBMCs. (C) Comparison of CD45RO and CD45RA expressions in CD4+, CD8+ and γδ T cells. The same samples were examined as in Figure 1. Blue dotted box: comparison of CD45RA+% among different T cells subtypes; Red dotted box: comparison of CD45RO+% among different T cells subtypes. Q5: T cell subpopulation not expressing CD45RA or RO. Q6: T cell subpopulation expressing CD45RA or RO. Percentage of CD45RA+ or CD45RO+ in T cell subtype was expressed as Q6/(Q5 + Q6), with background subtracted using isotype control. ** p < 0.01, **** p < 0.0001.
Figure 2. CD45RO and CD45RA are expressed differentially in T cell subtypes. Lymphocytes were gated in PBMCs and examined for the expression of CD45RO and CD45RA in different T cell subtypes. (A) Gating strategy for T cell subtypes and expression of CD45RO and CD45RA; (B) Comparison of frequency of T cell subtypes in PBMCs. (C) Comparison of CD45RO and CD45RA expressions in CD4+, CD8+ and γδ T cells. The same samples were examined as in Figure 1. Blue dotted box: comparison of CD45RA+% among different T cells subtypes; Red dotted box: comparison of CD45RO+% among different T cells subtypes. Q5: T cell subpopulation not expressing CD45RA or RO. Q6: T cell subpopulation expressing CD45RA or RO. Percentage of CD45RA+ or CD45RO+ in T cell subtype was expressed as Q6/(Q5 + Q6), with background subtracted using isotype control. ** p < 0.01, **** p < 0.0001.
Cells 11 01844 g002
Figure 3. Activation status varies in CD45RO+ and CD45RA+ of CD4+ (B), but not of CD8+ T cells (C). CD45RO+ cells are designated as RO+ and CD45RA+ are labeled as RA+ in the figure (B,C). Lymphocytes were gated on CD4+ or CD8+ T cells in PBMCs and examined for the expression of CD25 and CD62L in either CD45RO+ or CD45RA+ subpopulations. (A,D) Gating strategies for CD25 (A) or CD62L (D) and expression of CD45RO and CD45RA based on gated CD4+ or CD8+ T cells. (B,C) Comparison of CD25+% in RO+ and RA+ subpopulations in CD4+ (B) and CD8+ (C) T cells. The same samples were analyzed as in Figure 1. Percentage of CD25+ expression in RO+ and RA+ subpopulations was expressed as Q6/(Q6 + Q7). (E) Comparison of CD62L+% in RO+ and RA+ subpopulations in CD4+ and CD8+ T cells. Percentage of CD62L+ expressing in CD45RO or CD45RA subpopulations was expressed as Q14/(Q14 + Q15). Background in isotype control was subtracted from all the data in (B,D). * p < 0.05; *** p < 0.001; **** p < 0.0001.
Figure 3. Activation status varies in CD45RO+ and CD45RA+ of CD4+ (B), but not of CD8+ T cells (C). CD45RO+ cells are designated as RO+ and CD45RA+ are labeled as RA+ in the figure (B,C). Lymphocytes were gated on CD4+ or CD8+ T cells in PBMCs and examined for the expression of CD25 and CD62L in either CD45RO+ or CD45RA+ subpopulations. (A,D) Gating strategies for CD25 (A) or CD62L (D) and expression of CD45RO and CD45RA based on gated CD4+ or CD8+ T cells. (B,C) Comparison of CD25+% in RO+ and RA+ subpopulations in CD4+ (B) and CD8+ (C) T cells. The same samples were analyzed as in Figure 1. Percentage of CD25+ expression in RO+ and RA+ subpopulations was expressed as Q6/(Q6 + Q7). (E) Comparison of CD62L+% in RO+ and RA+ subpopulations in CD4+ and CD8+ T cells. Percentage of CD62L+ expressing in CD45RO or CD45RA subpopulations was expressed as Q14/(Q14 + Q15). Background in isotype control was subtracted from all the data in (B,D). * p < 0.05; *** p < 0.001; **** p < 0.0001.
Cells 11 01844 g003
Figure 4. Expression of IFNγ and IL4 is not associated with CD45RO in CD4+ T cells. Purified PBMCs were stimulated with the activation cocktail for 4 h before intracellular staining for IFNγ and IL4, plus surface staining with antibodies to identify CD4+ T cells, and their expression of CD45RO and CD45RA. (A) Gating strategy for IFNγ and IL4 gated on CD4+ T cells. Q17: IL4+ subpopulation not expressing IFNγ, designated as IL4. Q18: IFNγ and IL4 double positive subpopulation, designated as IFN/IL4. Q19: IFNγ+ subpopulation not expressing IL4, designated as IFN. (B) Comparison of IFNγ and or IL4 producing CD4+ T cells between RO+ and RO null cattle samples. (C) Gating strategies for IFNγ or IL4 producing CD4+ in CD45RO or CD45RA subpopulations. (D) Comparison of IFNγ or IL4 expression in RO+ or RA+ CD4+ T cells. Percentage of IFNγ+ was calculated as Q12/(Q11 + Q12), whereas IL4+ as Q26/(25 + 26). (E) Comparison of CD45RO+ or CD45RA+ in IFNγ+ or IL4+ CD4+ T cells. CD45RO+ or CD45RA+% in IFNγ+ was calculated as Q12/(Q12 + Q13), whereas in IL4+% as Q26/(26 + 27), as indicated in (C). Background in isotype control was subtracted from all the data in B, D, and E. **** p < 0.0001.
Figure 4. Expression of IFNγ and IL4 is not associated with CD45RO in CD4+ T cells. Purified PBMCs were stimulated with the activation cocktail for 4 h before intracellular staining for IFNγ and IL4, plus surface staining with antibodies to identify CD4+ T cells, and their expression of CD45RO and CD45RA. (A) Gating strategy for IFNγ and IL4 gated on CD4+ T cells. Q17: IL4+ subpopulation not expressing IFNγ, designated as IL4. Q18: IFNγ and IL4 double positive subpopulation, designated as IFN/IL4. Q19: IFNγ+ subpopulation not expressing IL4, designated as IFN. (B) Comparison of IFNγ and or IL4 producing CD4+ T cells between RO+ and RO null cattle samples. (C) Gating strategies for IFNγ or IL4 producing CD4+ in CD45RO or CD45RA subpopulations. (D) Comparison of IFNγ or IL4 expression in RO+ or RA+ CD4+ T cells. Percentage of IFNγ+ was calculated as Q12/(Q11 + Q12), whereas IL4+ as Q26/(25 + 26). (E) Comparison of CD45RO+ or CD45RA+ in IFNγ+ or IL4+ CD4+ T cells. CD45RO+ or CD45RA+% in IFNγ+ was calculated as Q12/(Q12 + Q13), whereas in IL4+% as Q26/(26 + 27), as indicated in (C). Background in isotype control was subtracted from all the data in B, D, and E. **** p < 0.0001.
Cells 11 01844 g004
Figure 5. Expression of IFNγ is not associated with CD45RO in CD8+ T cells. The same samples and treatment were applied as in Figure 4, but the analysis was gated on CD8+ T cells. (A) Gating strategies for IFNγ+ and IL4+, plus IFNγ+ and CD45RO/CD45RA expression gated on CD8+ T cells. (B) Comparison between IFNγ (designated as IFN: Q2 + Q3) and IL4 (Q1 + Q2) producing cells in CD8. CD45RO+ cells were designated as RO+ and CD45RA+ cells as RA+ in (C,D). (C) Percentage of IFNγ+ in RO+ or RA+ CD8+ T cells: Q12/(Q11 + Q12), as indicated in (A). (D) Percentage of RO+ or RA+ in IFNγ producing CD8+ T cells: Q12/(Q12 + Q13), as indicated in (A). Background in isotype control was subtracted from all the date in B–D. * p < 0.05; ** p < 0.01; **** p < 0.0001.
Figure 5. Expression of IFNγ is not associated with CD45RO in CD8+ T cells. The same samples and treatment were applied as in Figure 4, but the analysis was gated on CD8+ T cells. (A) Gating strategies for IFNγ+ and IL4+, plus IFNγ+ and CD45RO/CD45RA expression gated on CD8+ T cells. (B) Comparison between IFNγ (designated as IFN: Q2 + Q3) and IL4 (Q1 + Q2) producing cells in CD8. CD45RO+ cells were designated as RO+ and CD45RA+ cells as RA+ in (C,D). (C) Percentage of IFNγ+ in RO+ or RA+ CD8+ T cells: Q12/(Q11 + Q12), as indicated in (A). (D) Percentage of RO+ or RA+ in IFNγ producing CD8+ T cells: Q12/(Q12 + Q13), as indicated in (A). Background in isotype control was subtracted from all the date in B–D. * p < 0.05; ** p < 0.01; **** p < 0.0001.
Cells 11 01844 g005
Figure 6. Expression of IFNγ and IL4 is not associated with CD45RO in γδ T cells. PBMCs were isolated from 12 cattle, which were tested for expression of CD45RO (RO+), CD62L (CD62L+), IFNγ (IFN) and IL4 in a way similar to that in Figure 4. (A) Gating strategy for RO+ and CD62L+ based on γδ T cells. (B) Comparison of CD62L expression in RO+ cattle and RO null cattle. (C) Gating strategy for IFNγ and IL4+ gated on γδ T cells. (D) Comparison of IFNγ and IL4+ between RO+ and RO null cattle. IL4 or IFN: Q5 + Q6 + Q7. Q5: IL4 only; Q6: IFN/IL4; Q7: IFN only. Red dotted box: IL4 or IFN; blue box: IFN only; black box: IFN/IL4. (EG): Gating strategy for CD25+ and IFNγ+ or IL4+ based on γδ T cells (E), and comparison of their expression in both RO+ and RO null cattle (F,G). In F, CD25+% in all: Q9 + Q10; CD25+% in IFN: Q10/(Q10 + Q11); CD25+% in IL4+: Q14/(Q14 + Q15). In G, IFN in CD25+: Q10/(Q9 + Q10); IL4 in CD25+: Q14/(Q13 + Q14). Data were expressed as mean plus 95% confidence interval (CI) in (B,D,F,G). Background in isotype control was subtracted from all the date in (B,D,F,G). *** p < 0.001; **** p < 0.0001.
Figure 6. Expression of IFNγ and IL4 is not associated with CD45RO in γδ T cells. PBMCs were isolated from 12 cattle, which were tested for expression of CD45RO (RO+), CD62L (CD62L+), IFNγ (IFN) and IL4 in a way similar to that in Figure 4. (A) Gating strategy for RO+ and CD62L+ based on γδ T cells. (B) Comparison of CD62L expression in RO+ cattle and RO null cattle. (C) Gating strategy for IFNγ and IL4+ gated on γδ T cells. (D) Comparison of IFNγ and IL4+ between RO+ and RO null cattle. IL4 or IFN: Q5 + Q6 + Q7. Q5: IL4 only; Q6: IFN/IL4; Q7: IFN only. Red dotted box: IL4 or IFN; blue box: IFN only; black box: IFN/IL4. (EG): Gating strategy for CD25+ and IFNγ+ or IL4+ based on γδ T cells (E), and comparison of their expression in both RO+ and RO null cattle (F,G). In F, CD25+% in all: Q9 + Q10; CD25+% in IFN: Q10/(Q10 + Q11); CD25+% in IL4+: Q14/(Q14 + Q15). In G, IFN in CD25+: Q10/(Q9 + Q10); IL4 in CD25+: Q14/(Q13 + Q14). Data were expressed as mean plus 95% confidence interval (CI) in (B,D,F,G). Background in isotype control was subtracted from all the date in (B,D,F,G). *** p < 0.001; **** p < 0.0001.
Cells 11 01844 g006
Table 1. Primary antibodies.
Table 1. Primary antibodies.
SpecificityCloneIsotypeSource
bCD3MM1AIgG1WSUMAC
bCD4CC8IgG2aBio-Rad
bCD25LCTB2AIgG3WSUMAC
bTCRδGB21AIgG2bWSUMAC
bTCRδCACT61AIgMWSUMAC
bCD45RAGC6AIgMWSUMAC
bCD45ROILA116IgG3WSUMAC
bCD62LBAQ92AIgG1WSUMAC
bIFNγCC302IgG1Bio-Rad
bIL4CC303IgG2aBio-Rad
SpecificityCloneConjugated
Fluorescence
Source
bCD4CC8PEBio-Rad
bCD8CC63FITCBio-Rad
bCD8CC58PEBio-Rad
bCD25IL-A111PEBio-Rad
bCD25IL-A111FITCBio-Rad
bCD62LCC32FITCBio-Rad
bIFNγCC302PEBio-Rad
b, Bovine. WSUMAC, Washington State University Monoclonal Antibody Center. Bio-Rad, Bio-Rad Laboratories, Inc. Hercules, CA, USA. Note: All the primary antibodies are monoclonal and generated from mice.
Table 2. Secondary antibodies and isotype controls.
Table 2. Secondary antibodies and isotype controls.
SpecificitySecondary AntibodiesSource
IgG1Anti-mouse IgG1-APCBioLegend
IgG1Anti-mouse IgG1-BiotinBioLegend
IgG2aAnti-mouse IgG2a-APCBioLegend
IgG2bAnti-mouse IgG2b-PEBioLegend
IgG2bAnti-mouse IgG2b-BiotinBioLegend
IgG2bAnti-mouse IgG2b-FITCBioLegend
IgG3Anti-mouse IgG3-BiotinBioLegend
IgMAnti-mouse IgM-BV421BioLegend
BiotinStreptavidin Per-CPBioLegend
Isotype ControlsSource
Mouse IgG1BioLegend
Mouse IgG2aBioLegend
Mouse IgG2bBioLegend
Mouse IgG3BioLegend
Mouse IgMBioLegend
Mouse IgG1-FITCBioLegend
Mouse IgG1-PEBioLegend
Mouse IgG2a-FITCBioLegend
Mouse IgG2a-PEBioLegend
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kandel, A.; Li, L.; Hada, A.; Xiao, Z. Differential Expression of CD45RO and CD45RA in Bovine T Cells. Cells 2022, 11, 1844. https://doi.org/10.3390/cells11111844

AMA Style

Kandel A, Li L, Hada A, Xiao Z. Differential Expression of CD45RO and CD45RA in Bovine T Cells. Cells. 2022; 11(11):1844. https://doi.org/10.3390/cells11111844

Chicago/Turabian Style

Kandel, Anmol, Lei Li, Akanksha Hada, and Zhengguo Xiao. 2022. "Differential Expression of CD45RO and CD45RA in Bovine T Cells" Cells 11, no. 11: 1844. https://doi.org/10.3390/cells11111844

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop