throbber
Clinical Immunology (2008) 127, 107–118
`
`a v a i l a b l e a t w w w. s c i e n c e d i r e c t . c o m
`
`w w w. e l s e v i e r. c o m / l o c a t e / y c l i m
`
`T cell subset-specific susceptibility to aging
`Marta Czesnikiewicz-Guzik a, Won-Woo Lee a, Dapeng Cui a, Yuko Hiruma a,
`David L. Lamar a, Zhi-Zhang Yang b, Joseph G. Ouslander c,
`Cornelia M. Weyand a, Jörg J. Goronzy a,⁎
`
`a Kathleen B. and Mason I. Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, GA, USA
`b Division of Hematology, Mayo Graduate School, Rochester, MN, USA
`c Division of Geriatric Medicine and Gerontology, Emory University School of Medicine, Atlanta, GA, USA
`
`Received 18 October 2007; accepted with revision 7 December 2007
`Available online 28 January 2008
`
`KEYWORDS
`Immunosenescence;
`Aging;
`T-cell subset;
`T-cell homeostasis;
`CD4;
`CD8;
`Killer
`immunoglobulin-like
`receptors;
`CD85
`
`With increasing age, the competence of the immune system to fight infections and
`Abstract
`tumors declines. Age-dependent changes have been mostly described for human CD8 Tcells, raising
`the question of whether the response patterns for CD4 Tcells are different. Gene expression arrays of
`memory CD4 Tcells yielded a similar age-induced fingerprint as has been described for CD8 Tcells. In
`cross-sectional studies, the phenotypic changes were not qualitatively different for CD4 and CD8 T
`cells, but occurred much more frequently in CD8 Tcells. Homeostatic stability partially explained this
`lesser age sensitivity of CD4 Tcells. With aging, naïve and central memory CD8 Tcells were lost at the
`expense of phenotypically distinct CD8 effector Tcells, while effector CD4 Tcells did not accumulate.
`However, phenotypic shifts on central memory Tcells were also more pronounced in CD8 Tcells. This
`distinct stability in cell surface marker expression can be reproduced in vitro. The data show that CD8
`T cells are age sensitive by at least two partially independent mechanisms: fragile homeostatic
`control and gene expression instability in a large set of regulatory cell surface molecules.
`© 2007 Elsevier Inc. All rights reserved.
`
`Introduction
`
`Failure of adaptive immunity with age is a major cause for
`morbidity and mortality in the elderly [1,2]. As a highly
`dynamic organ, the immune system is in constant turnover,
`even in the absence of infections or obvious challenges with
`exogenous antigens [3]. Naïve T cells have a half-life of 6 to
`12 months and memory T cells of 15 to 45 days [4,5]. Thymic
`
`⁎ Corresponding author. Lowance Center for Human Immunology,
`Emory University School of Medicine, 101 Woodruff Circle #1003,
`Atlanta, GA 30322, USA. Fax: +1 404 727 7371.
`E-mail address: jgoronz@emory.edu (J.J. Goronzy).
`
`production of new Tcells dwindles with age and does not meet
`the replenishment demand during adulthood [6–8]. After the
`ages of 40 to 50 years old, virtually the entire T-cell supply is
`generated from existing naïve and memory T cells [9]. In this
`setting, insufficient homeostatic mechanisms may lead to a
`progressive loss of naïve and memory Tcells and contraction of
`T-cell receptor diversity [10–14]. In addition, the replicative
`stress associated with continuous turnover can induce cellular
`senescence and lead to phenotypic changes that impinge on
`the competence of the adaptive immune system [15–18].
`Both mechanisms contribute to failure to respond to new
`antigenic challenges, poor vaccine responses [19–22] and in-
`creased morbidity with newly arising infections, such as is seen
`with antigenic shift or drift of the influenza virus [23,24].
`
`1521-6616/$ – see front matter © 2007 Elsevier Inc. All rights reserved.
`doi:10.1016/j.clim.2007.12.002
`
`UPenn Ex. 2055
`Miltenyi v. UPenn
`IPR2022-00855
`107
`
`

`

`108
`
`M. Czesnikiewicz-Guzik et al.
`
`Moreover, memory T-cell responses to some persisting viruses
`wane—a prime example is the increased incidence of herpes
`zoster with age [25]. Epidemiological evidence suggests that
`signs of decreased immune competence first occur after the
`age of 50 and accelerate after the ages of 65 to 70 [20,23,25].
`It is currently unclear which functional and phenotypic chan-
`ges in the immune system occur at what ages and, in particular,
`whether different cell types are affected to distinct extents,
`possibly providing a model for the patterns of age-dependent
`susceptibilities for different viral infections.
`Circumstantial evidence suggests that CD4 and CD8 T cells
`behave differently in response to aging. Oligoclonal expansions
`in the CD8 T-cell compartment can be readily detected with
`age and appear to be induced by chronic persisting viruses, in
`particular CMV, but also arise from uneven homeostatic pro-
`liferation [12,15,26,27]. In contrast, oligoclonal expansions
`within the CD4 compartment are rare and preferentially found
`in patients with autoimmune diseases [28]; they do not reach
`the clonal size seen with CD8 Tcells [18]. Even at the age of 65,
`decades after the involution of the thymus, both naïve and
`memory CD4 Tcells are highly diverse and show no contraction
`in T-cell receptor diversity compared to young adults [10].
`Also, the classical phenotypic change of CD28 loss, frequently
`encountered in CD8 Tcells with age [17], is only inconsistently
`seen for CD4 T cells [11]. However, the molecular mechanisms
`regulating CD28 expression and loss appear to be identical in
`both T-cell subsets [29,30], suggesting a fundamental differ-
`ence in CD4 and CD8 T cells in response to aging, possibly in
`addition to distinct cell-specific transcriptional regulation.
`The objective of this study was to compare CD4 and CD8 Tcells
`and to determine whether differences in their phenotypic
`response pattern to aging are explained by differences in T-cell
`subset homeostasis or whether CD4 and CD8 T cells are in-
`trinsically different in controlling gene expression.
`
`Materials and methods
`
`Study population
`
`Peripheral blood was obtained from 140 individuals aged
`20–90 years and immediately processed. The study cohort
`included 68 individuals age 20 to 39 years, 31 age 40 to 59,
`and 41 age 60 to 90 years. Exclusion criteria included the
`presence or a history of cancer, uncontrolled hypertension,
`diabetes mellitus, any chronic inflammatory or autoimmune
`disease, or any acute disease. Appropriate written informed
`consent was obtained, and the study was approved by the
`Emory Institutional Review Board.
`
`Gene expression microarray studies
`
`CD4 T cells were negatively enriched with human CD4+ T-cell
`enrichment cocktail (RosetteSep; StemCell Technologies,
`Vancouver, Canada) from PBMC of five 20- to 30- and five
`70- to 75-year-old healthy volunteers. CD45RA−CD4+ memory
`T cells were isolated by depletion of CD45RA+ subsets with
`anti-CD45RA magnetic beads (Miltenyi Biotec, Auburn, CA).
`Purity was between 95% and 98%. Total RNA was extracted
`using an RNeasy Mini Kit (Qiagen, Valencia, CA), amplified,
`and labeled using a modification of the technique described
`by Baugh et al. [31]. A first round of in vitro transcription (IVT)
`
`was followed by a second round of reverse transcription,
`second strand synthesis, and IVT. aRNA was hybridized with
`Affymetrix U133A GeneChips by the Institute of System Bio-
`logy (Seattle, WA). Gene expression signal was summarized by
`robust multi-array average RMA [32]. Genes that showed a
`1.25-fold difference between young and old samples were
`identified.
`In a second microarray series, CD4+CD28+ and CD4+CD28− T
`cells were purified from three donors by cell sorting using FACS
`vantage (BD Biosciences, San Jose, CA). Cells were expanded
`by stimulation with anti-CD3 (OKT3; Ortho Diagnostics, Ro-
`chester, NY) immobilized at 1 μg/ml in IL-2-containing me-
`dium. RNA was extracted 2 weeks after stimulation. This time
`point was chosen because gene expression in T cells cultured
`under these conditions is stable and kinetic changes induced by
`the stimulation are no longer observed (data not shown).
`Labeled aRNA was hybridized with Affymetrix Hu-95Av2
`GeneChips (Mayo Cancer Microarray Core Facility, Mayo
`Foundation; Rochester, MN). Data were analyzed with Micro-
`array Analysis Software version 5.0 (Affymetrix). A gene was
`called differentially expressed if comparisons for all three
`individuals yielded a consistent change call, or if two com-
`parisons yielded consistent change calls and the third yielded a
`“no change” call.
`
`Immunophenotyping and flow cytometry analysis
`
`To confirm age-dependent differential expression of cell
`surface markers identified in the gene arrays, PBMC were
`stained with the following monoclonal antibodies in 5-color
`panels: FITC-conjugated anti-CD158b/j (GLI83; CH-L), PE-anti-
`CD85j (HP-F1), PerCP-Cy5.5-anti-CD28 (L293), APC-anti-CD8
`(RPA-T8), APC-Cy7-anti-CD3 (SK7), FITC-anti-CD57 (HNK-1),
`PE-anti-CD26 (M-A261), PerCP-anti-CD4 (L200), APC-anti-CD3
`(HIT3a); FITC-conjugated anti-HLA-DR (G46-6), PE-anti-CD3
`(HIT3a), PerCP-anti-CD4 (L200), APC-anti-CD45R0 (4CHL1), PE-
`Cy7-anti-CD69 (FN 50). FITC- or APC-anti-CD45RA (RA5H9,
`HI100), PE- or APC-anti-CCR7 (150503), and PerCP-anti-CD8
`(SK1) were used to define the functional subset distribution.
`To determine the frequency of cytomegalovirus (CMV)-
`specific CD8 Tcells in different age groups, PBMC from HLA-A2
`individuals were stained with APC-conjugated HLA-A⁎0201/
`CMV pp65495–504 (NLVPMVATV) pentamer (Proimmune Inc,
`Springfield, VA) at room temperature for 10 min. Following
`washing, the cells were incubated with PE-anti-CD85j, PerCP-,
`or PE-Cy7-anti-CD8; FITC- or PerCP-Cy5.5-anti-CD28; and APC-
`Cy7-anti-CD3 antibodies for 20 min.
`All samples were acquired with FACSort or LSRII (BD Bio-
`sciences), and data were analyzed by using CellQuest, FACS
`DIVA software (both BD Biosciences), or FlowJo (Tree Star, Inc.,
`Ashland, OR). All antibodies were from BD Biosciences, except
`PE-, APC-anti-CCR7 (R&D Systems, Minneapolis, MN, USA), and
`anti-CD85j (Beckman Coulter, Fullerton, CA, USA).
`
`In vitro cultures
`
`CD28 positive cells were sorted from PBMC with biotin-labeled
`anti-CD28 antibody (BD Biosciences), and anti-biotin labeled
`microbeads (Miltenyi). Isolated cells were labeled with CFSE
`and stimulated with anti-CD3 (OKT3, 30 ng/ml) in the presence
`of irradiated EBV transformed cells, rhIl-2 (50 U/ml) and rhIl-
`
`UPenn Ex. 2055
`Miltenyi v. UPenn
`IPR2022-00855
`108
`
`

`

`T cell subset-specific susceptibility to aging
`
`109
`
`15 (100 ng/ml). Cultures were split every 4 days. Cells were
`analyzed at 6- to 8-day intervals for the expression of CD85j,
`CD28 and HLA-DR on CD4 and CD8 T cells that had undergone
`the same number of divisions as determined by CSFE dilution.
`
`Statistical analysis
`
`Results are expressed as medians, 25th/75th percentiles as
`boxes and 10th/90th percentiles as whiskers. Groups were
`compared using ANOVA or non-parametric Mann–Whitney U
`test. p values b 0.05 were considered statistically significant.
`
`Results
`
`Differential loss of CD28 in CD8 versus CD4 T cells
`
`Loss of CD28 expression, the dominant phenotypic change of
`T cells with aging, is seen much more frequently in CD8 than
`in CD4 T cells. Figure 1 shows a cross-sectional study of CD28
`expression in different age groups. In the vast majority of
`individuals, lack of CD28 is a rare event in CD4 T cells,
`irrespective of age. There was a small increase in CD4+CD28−
`T-cell frequencies (p b 0.001) and increasing variance in
`healthy individuals older than 60 years; however, even in
`this age group only a median of 7.9% of CD4 T cells lost CD28
`expression, and individuals with more than 20% CD4+CD28− T
`cells were the rare exception. In contrast, 21% of CD8 T cells
`were already negative for CD28 expression in individuals
`between the age of 20 and 39 years. The frequency increased
`to 25.5% in 40- to 59-year-old healthy individuals (p b 0.001).
`After that age, CD28 loss accelerated to a median frequency
`of 53.9% in the 60–90-year cohort (p b 0.001). Again the
`variance increased with age, indicating higher heterogeneity
`in the older population.
`
`Age-dependent changes in the transcriptional profile
`of CD4 memory T cells
`
`Regulation of CD28 may be cell specific, and CD8 T cells may
`lose CD28 more easily. Indeed, the DNA-binding proteins con-
`trolling the initiation of CD28 transcription differ in composi-
`tion between CD4 and CD8 T cells [30]. However, the basic
`mechanism of CD28 repression is the same in both T-cell sub-
`sets, suggesting that factors not related to CD28 transcription
`
`play a role [29]. The CD4 memory T cells may be a more stable
`cell type, with lesser susceptibility to age-dependent pheno-
`typic and functional changes. To determine whether CD4 T
`cells principally undergo the same changes in gene expression
`with age as CD8 T cells but do so at a substantially reduced or
`delayed rate, we performed gene expression arrays on (a)
`CD45RA−CD4+ cells purified from PBMC of five 20- to 30-year-
`old and five 70- to 75-year-old healthy donors and (b)
`CD28+CD4+ and CD28−CD4+ T cells from the same donor and
`compared them to published gene profiles for CD8 Tcells [33];
`The gene expression profiles in young and old memory CD4 T
`cells highly correlated indicating that age-dependent changes
`in CD4 T-cell transcription were small or only involved a minor
`subpopulation. Because even small changes in gene expression
`or emergence of a low-frequent subpopulation could have
`physiological consequences, we used a low cut-off of a 1.25-
`fold difference in expression levels between the two age
`groups to screen for genes that warrant further studies. 536
`genes were identified, including the cell surface molecules
`listed in Table 1. Several of the genes over-expressed in the
`elderly mapped to the HLA region. CD28 was among the few
`genes that were less expressed in elderly memory CD4 cells.
`Hypothesizing that the differences were small because only a
`minor subset of CD4 T cells is affected by aging and these
`senescent cells are characterized by the loss of CD28, in ana-
`logy to CD8+CD28− T cells, we compared the gene expression
`profiles of CD4+CD28− to CD4+CD28+ T cells. In general,
`identified differences in this subset were of larger magnitude
`than the age-dependent differences observed in the global
`memory CD4 T-cell pool. Cell surface molecules identified as
`differentially expressed partially overlapped with the genes
`identified in the global memory cell populations (Table 1). MHC
`class II genes again were among those that were overexpressed
`in the cells with the senescent CD4+CD28− phenotype. Also,
`the KLRC2 genes were underrepresented in both the elderly
`CD4+ memory cells and CD4+CD28− T cells. As expected, CD27
`transcripts that are known to correlate with CD28 loss in CD4
`and CD8 T cells were reduced in CD4+CD28− T cells. The most
`striking feature of these cells was the overexpression of
`several genes encoded in the leukocyte regulatory cluster,
`including several KIR genes.
`In comparing our results with those of Fann et al., it is
`evident that aging CD4 and CD8 T cells undergo similar
`transcriptional changes. In agreement with their findings for
`senescent CD8 Tcells [33], we observed a significant decrease
`
`Influence of age on CD28 loss within CD4+ and CD8+ T cells. The frequency of CD28 loss in CD4 (left panel) and CD8 (right
`Figure 1
`panel) T-cell subpopulations was determined by FACS in healthy individuals of different ages. Results are shown as box plots with
`medians, 25th and 75th percentiles as boxes and 10th and 90th percentiles as whiskers for different age strata. Although decline was
`significant for both CD4 (p b 0.001) and CD8 T cells (p b 0.001), loss was much more dramatic and earlier in life in the CD8 population
`and was already significant for middle-aged individuals (p b 0.001).
`
`UPenn Ex. 2055
`Miltenyi v. UPenn
`IPR2022-00855
`109
`
`

`

`110
`
`Table 1
`
`Age of donor
`
`Cell type
`Differentially overexpressed cell surface molecules
`
`Influence of age on gene expression in CD4 T cells
`20–30 years
`CD4+CD45RA−
`KLRB1
`CD28
`KLRC2
`CCR6
`
`M. Czesnikiewicz-Guzik et al.
`
`65–75 years
`CD4+CD45RA−CD28+
`TCRβ-chain
`ICAM-2
`CD7
`KLRC2
`IL9R
`IL7R
`CD27
`
`70–75 years
`CD4+CD45RA−
`HLA-DRA
`CCR4
`CD26
`HLA-DPB1
`HLA-DPB5
`HLA-DPB4
`HLA-F
`HLA-DPA1
`CCR8
`CD58
`IL17RB
`LAIT1
`CD3E
`
`CD4+CD45RA−CD28−
`KIR2DL3
`KIR2DL4
`KIR3DL2
`CD70
`HLA-DRA
`HLA-DQA
`CD74
`HLA-DRB1
`HLA-DPB1
`HLA-DMA
`HLA-DPA
`CCR5
`HLA-DQB1
`CD2
`
`in CD28 expression in memory CD4 T cells from elderly donors.
`We found that expression of the costimulatory molecules
`CTLA4 and PD-1 did not differ with respect to age in CD4
`memory cells, a finding which mirrored Fann et al.'s results
`with respect to senescence phenotype in CD8 T cells. While
`CD40L is significantly downregulated in CD8+CD28− Tcells [33],
`we did not detect an age-dependent decline in memory CD4
`cells; however, we have previously shown that CD28−CD4+ T
`cells lack the ability to induce CD40L [34]. Furthermore, there
`are many similarities in the expression of NK cell receptors on
`CD28−CD8 and CD4 T cells. For example, our results showed
`significant downregulation of KLRB1 (CD161) expression in old
`memory CD4 cells, as Fann et al. demonstrated for CD8+CD28−
`cells. Similarly, the expression of KIR2DL2, KIR2DL3, and
`KIR2DL4 increased in CD4+CD28− cells, although these differ-
`ences did not reach the significance found in CD8+CD28− T
`cells. We did not detect any changes in expression of KLRD1,
`KLRG1, and KLRK1 between old and young memory CD4 cells,
`in contrast to the significant upregulation of these genes in
`CD8+CD28− cells. Several chemokine receptors exhibited ex-
`pression patterns in CD4 memory cells that are analogous to
`those found in CD8 cells. CCR2, CCR6, and CCR7, which are
`all significantly downregulated in CD8+CD28− cells [33],
`decrease with age in memory CD4 T cells, with CCR6 reaching
`significance.
`
`Preferential age-dependent expression of negative
`regulatory receptors on CD8 T cells
`
`Candidate cell surface molecules implicated in the gene arrays
`were examined for their age-dependent expression in a cohort
`of 140 healthy individuals. One of the most striking findings of
`the gene array studies was the overexpression of several KIR
`genes in CD4+CD28− T cells. KIR genes have allelic polymorph-
`isms, and we selected the antibody to CD158b/j that re-
`cognizes the most frequent genetic variants KIR2DL2, KIR2DL3,
`and KIR2DS2 for further analysis [35]. In addition, we included
`CD85j, which is encoded in the leukocyte cluster near the KIR
`genes, in our analysis. Like most KIR genes, CD85j is a negative
`regulatory receptor that is irregularly expressed on T cells.
`Cross-sectional studies showed preferential expression of
`CD85j and CD158b/j in CD8 T cells, while they were infre-
`
`quently found in CD4 T cells (Fig. 2A). Expression of both
`molecules, but particularly CD85j, increased with age; the in-
`crease was already apparent in the age group of 40- to 59-year
`olds, but clearly accelerated after that age. Compared to the
`20–39-year-old individuals, CD158b/j (p b 0.001) and CD85j
`(p b 0.001) expression was significantly more frequent in CD8
`T cells from individuals older than 60 years. In many of these
`individuals, the majority of CD8 Tcells were positive for CD85j.
`The age-dependent increase was much less striking in CD4 T
`cells and did not reach significance for either molecule. How-
`ever, in both T-cell subsets, the expression of these receptors
`was closely related to the loss of CD28. In both the CD4 and CD8
`populations, the majority of CD28− T cells expressed CD85j. In
`contrast, CD4+CD28+ Tcells expressed almost no CD85j. A small
`fraction of CD8+CD28+ Tcells, however, was consistently found
`to express CD85j (Fig. 2B). CD158b/j showed the same dis-
`tribution pattern but was much less frequent and exceeded
`10% expression in only a few individuals, even on CD28− Tcells.
`KIR and CD85j expression patterns resembled those of CD57,
`a generally accepted T-cell senescence marker [36]. Gain in
`CD57 expression showed a similar age dependence (Fig. 2A),
`with a median of 5.1% of CD4 T cells and 33.35% of CD8 T cells
`being CD57-positive in 60- to 90-year olds. CD57 expression was
`mainly restricted to the CD28− cell population and occurred
`only infrequently in CD28+ cells (Fig. 2B). Thus, in both CD4 and
`CD8 Tcells, the gains in CD57, CD85j, and CD158b/j, and loss of
`CD28 with age were highly correlated, suggesting that the
`changes are qualitatively similar. However, they occurred much
`more frequently in CD8 T cells. Table 2 shows the slopes of the
`linear regression curves examining the relationship between
`age and frequency of cell surface expression. For each
`biomarker shown, the slope is significantly steeper for CD8 T
`cells, documenting a fundamental difference in CD4 and CD8 T
`cells in their responses to aging.
`
`Defective T-cell homeostasis in CD8 Tcells contributes
`to the preferential acquisition of negative regulatory
`receptors
`
`Since CD28 loss is mostly seen on effector T cells, we
`addressed the question of whether the higher susceptibility
`of CD8 T cells to undergo age-dependent phenotypic changes
`
`UPenn Ex. 2055
`Miltenyi v. UPenn
`IPR2022-00855
`110
`
`

`

`T cell subset-specific susceptibility to aging
`
`111
`
`Age-dependent gain of CD57, CD85j, and CD158b/j expression in CD8+ T cells. (A) Frequencies of CD85j, CD158b/j, and
`Figure 2
`CD57 cells within CD4 (left panel) and CD8 (right panel) T cells were determined by FACS. Results are shown as box plots with medians,
`25th and 75th percentiles as boxes, and 10th and 90th percentiles as whiskers for healthy individuals representing different age strata.
`(B) In both CD4 and CD8 T cells, CD57 and CD85j were preferentially expressed on CD28− (shaded areas) and less on CD28+ (dark lines).
`
`reflected different stability in homeostatic control mechan-
`isms. Quantification of naïve, central, and effector memory
`cells using the CD45RA and CCR7 markers in different age
`groups indeed showed considerable differences between CD4
`and CD8 T cells. In both compartments, naïve T cells were
`reduced in elderly individuals ( p b 0.001); however, the loss
`was much more dramatic for CD8 (Fig. 3B) than for CD4 Tcells
`(Fig. 3A). The functional subset distribution within the CD4
`memory compartment was stable with age (Fig. 3C). In
`contrast, the subset of central CD8 memory T cells was
`
`Linear regression analysis of the age dependency
`Table 2
`of cell surface molecule expression in CD4 and CD8 T cells
`
`CD8 T cells
`CD4 T cells
`Δ frequency
`Δ frequency
`(%/year)
`(%/year)
`−0.03 ± 0.04 −0.6 ± 0.07
`CD28
`0.1 ± 0.07
`0.7 ± 0.07
`CD85j
`0.2 ± 0.07
`0.4 ± 0.07
`CD57
`CD158b/j −0.02 ± 0.01
`0.1 ± 0.03
`0.09 ± 0.07 −0.5 ± 0.1
`CD26
`HLA-DR
`0.2 ± 0.03
`0.6 ± 0.8
`
`F
`
`p
`
`F = 36.798 p b 0.0001
`p b 0.001
`F = 25.75
`F = 4.942
`p = 0.03
`F = 12.483 p = 0.0005
`F = 12.46
`p = 0.0006
`p b 0.0001
`F = 26.02
`
`diminished (p = 0.01), and the CD8 effector memory T cells
`increasingly acquired a CD45RA phenotype ( p b 0.001, Fig.
`3D) The CD4/CD8 ratio was not affected by age, contrary to
`the hypothesis that expansion of CD8 effector T cells
`outcompetes CD4 T cells.
`The altered subset distribution in part accounted for the
`preferential phenotypic changes in CD8 Tcells. CD158b/j was
`exclusively expressed on CD45RA terminal effector T cells,
`suggesting that the accumulation of this subset was
`responsible for the emergence of CD158b/j cells with age.
`CD57 and CD85j were also preferentially expressed on
`CD45RA effector T cells; however, expression was not subset
`specific, and all three memory subsets gained expression of
`these molecules with age (Fig. 3E).
`
`Expansion of CMV-specific T cells does not account
`for the preference of age-dependent changes for
`the CD8 compartment
`
`Several recent studies have attributed disordered CD8 T-cell
`homeostasis to chronic persistent anti-CMV responses and
`the accumulation of oligoclonal CMV-specific effector T cells
`[14,15,27]. To examine the relationship between CMV
`
`UPenn Ex. 2055
`Miltenyi v. UPenn
`IPR2022-00855
`111
`
`

`

`112
`
`M. Czesnikiewicz-Guzik et al.
`
`Influence of age on functional T-cell subset distribution. PBMC were isolated from healthy young (n = 31; 27.9 ± 5.5 years)
`Figure 3
`and elderly (n = 26; 73.1 ± 4.2 years) individuals and stained with anti-CD3, -CD4, -CD8, -CD45RA, and -CCR7 mAbs to determine
`frequencies of T-cell subsets. Results are shown for CD45RA+CCR7+ naïve CD4 (A) and CD8 (B), CD45RA−CCR7+ central memory (CM),
`CD45RA−CCR7− effector memory (EM) and CD45RA+CCR7− effector (CD45RA effector) CD4 (C) and CD8 cells (D). Expression of CD57,
`CD85j and CD158b/j in the elderly was highest in effector CD8 Tcells, but CD57 and CD85j was also present in a central memory Tcells.
`Representative histograms are shown (E).
`
`responses and age-dependent phenotypic changes, we
`selected HLA-A2+ individuals and determined the frequency
`and phenotype of HLA-A2-restricted T cells specific for the
`CMV peptide NLVPMVATV (A2/NLV). While the majority of
`individuals in our population did not have any detectable
`(b0.1%) CMV pp65495–504 peptide-specific clones irrespective
`of age, clonal expansions of more than 1% were seen in about
`
`one half of the middle-aged and elderly populations
`(Fig. 4A). In contrast to the global CD8 T-cell compartment,
`CMV-specific clones did not undergo phenotypic changes with
`age; approximately 60% had lost CD28 expression and 50%
`had gained CD85j expression in young and elderly individuals
`(Fig. 4B). Individuals with CMV-specific clones had overall
`higher frequencies of CD8+CD28−CD85j+ T cells than
`
`UPenn Ex. 2055
`Miltenyi v. UPenn
`IPR2022-00855
`112
`
`

`

`T cell subset-specific susceptibility to aging
`
`113
`
`Expansion of CMV-specific T cells does not account for the preference of age-dependent phenotypic changes for the CD8
`Figure 4
`compartment. PBMC were isolated from HLA-A2+ individuals and stained with HLA-A2-CMV peptide (NLVPMVATV) (A2/NLV) pentamer,
`anti-CD3, -CD8, -CD28, and -CD85j mAbs. (A) Frequencies of CMV peptide-specific CD8 T cells are shown in correlation to age. (B) CMV-
`peptide-specific (solid boxes) and total CD8 T cells (open boxes) from young and elderly individuals who had N0.1% of CMV-peptide
`specific cells, were compared. (C) Individuals who had (solid bars) or did not have expanded CMV-peptide specific T cells (open bars)
`were compared for the frequency of CD85j and CD28 expression on CD8 T cells.
`
`CD4+ and CD8+ T cells differ in CD26 expression with age. Results of CD26 expression within CD4 (left panel) and CD8 (right
`Figure 5
`panel) T-cell populations in relation to age in cross-sectional studies are shown as box plots (A). Representative histograms of CD26
`expression in CD28+ and CD28−CD4 and CD8 cells from a 27 (top row) and 73-year-old (bottom row) individual are shown in (B).
`
`UPenn Ex. 2055
`Miltenyi v. UPenn
`IPR2022-00855
`113
`
`

`

`114
`
`M. Czesnikiewicz-Guzik et al.
`
`individuals without detectable CMV T-cell responses; how-
`ever, both subject populations exhibited the same age-
`dependent phenotypic shift in their CD8 compartment
`(Fig. 4C). These data suggest that CMV-specific responses
`are not the sole and, in most individuals, not a major cause
`for the observed age-dependent phenotypic conversions and
`the disequilibrium in functional T-cell subset distribution
`that mostly affect CD8 T cells.
`
`Preference for phenotypic conversions in CD8 T cells
`independent of homeostatic imbalance
`
`Several genes including CD26 and MHC class II molecules were
`found to be overexpressed in the gene array of elderly
`memory T cells independent of CD28 loss, raising the issue of
`whether their expression was independent of homeostatic
`dysregulation. Flow cytometric studies were performed to
`compare the effect of age on CD4 and CD8 T cells for these
`
`markers. A complex CD26 expression pattern was found,
`particularly on CD8 T cells. Three separate populations of
`CD26-expressing CD8 T cells could be distinguished: high,
`medium, and low expression. Most CD8 cells that had lost
`CD28 expression were also negative for CD26 (Fig. 5B, right
`panel). In addition, CD26high CD8 Tcells virtually disappeared
`with age. Both phenomena contributed to an age-dependent
`decrease in CD26 expression in CD8 Tcells ( p b 0.001, Fig. 5A).
`CD4 T cells were mostly CD26intermediate (Fig. 5B, left panel)
`and, contrary to the gene array, expression did not change
`with age (Fig. 5A, left panel). Thus, again there were striking
`differences between CD4 and CD8 T cells in age-dependent
`phenotypic changes.
`Genes of the MHC class II region were found to be dif-
`ferentially expressed in both series of gene arrays. Flow
`cytometry studies confirmed that the expression of HLA-DR is
`age dependent (Fig. 6A). A small increase in the frequencies of
`HLA-DR+ cells was already seen between the ages of 20 and
`59 years and then accelerated in the age group of 60 years and
`
`Increased expression of HLA-DR on CD4 and CD8 T cells with age. (A) The frequencies of HLA-DR+ cells within CD4 (left
`Figure 6
`panel) and CD8 (right panel) T cells are shown as box plots for different age groups. HLA-DR in elderly individuals is expressed on CD28+
`as well as CD28− T cells (B) and on central as well as effector memory CD8 T cells (C). Representative histograms from a 27- (top row)
`and 70-year-old (bottom row) individual are shown.
`
`UPenn Ex. 2055
`Miltenyi v. UPenn
`IPR2022-00855
`114
`
`

`

`T cell subset-specific susceptibility to aging
`
`115
`
`Phenotypic changes of the surface molecules: CD28, CD85j, HLA-DR in CD8 and CD4 subsets caused by long-term culture of
`Figure 7
`CD28 positive T cells. CD28 positive cells were sorted, labeled with CFSE, and stimulated with irradiated, EBV transformed PBMC and
`OKT3. FACS analysis was carried out every seventh day beginning from the first stimulation. CD28, HLA-DR, and CD85j molecules
`surface expression changes over time and is much more pronounced in CD8 T cells compared to the CD4 subset, suggesting that the
`phenotypic instability is intrinsic to CD8 T cells.
`
`older who had significantly higher frequencies of HLA-DR-
`expressing CD4 and CD8 T cells compared to young adults
`( p b 0.001). In contrast to KIRs, CD57 and CD85j, HLA-DR ex-
`pression did not coincide with CD28 loss. Representative
`histograms of CD4 and CD8 T cells are shown in Figure 6B. In
`both CD4 and CD8 subsets, very few Tcells expressed HLA-DR in
`a 27-year-old individual. Only CD8+CD28− T cells had a slightly
`higher expression of 6%. In contrast, 30% of CD8+CD28+ and 44%
`of CD8+CD28− T cells in a 70-year-old individual were positive
`for HLA-DR. 10 and 13%, respectively, of CD4+CD28+ and CD28−
`T cells in the same individual expressed the molecule. HLA-DR
`is known to be expressed on activated T cells, raising the
`possibility that elderly individuals have a larger fraction of
`activated cells. However, HLA-DR-positive cells had morpho-
`logical characteristics similar to HLA-DR-negative cells in the
`forward-side scatter, and there was no correlation with other
`activation markers, such as CD69, which does not increase with
`age (data not shown). Increased HLA-DR expression can
`therefore not be explained as a result of constitutive
`activation of T cells in the elderly. Although the pattern of
`HLA-DR expression was strikingly different from those of CD57
`and CD85j, the influence of age on HLA-DR expression was
`again more pronounced in CD8 than in CD4 T cells (Table 2).
`These findings suggest that CD8 Tcells are more susceptible to
`phenotypic conversions with age independent of their pre-
`ferred differentiation into effector cells.
`
`Intrinsic increased susceptibility of CD8 T cells to
`undergo phenotypic conversions
`
`To examine whether the phenotypic instability is intrinsic to
`CD8 Tcells, we activated CD4 and CD8 Tcells in vitro, expanded
`them in the presence of IL-2 and IL-15 and determined the
`expression of CD28, HLA-DR, and CD85j over time. To control
`for differences in population doublings, cells were labeled
`with CFSE, and CD4 and CD8 Tcells with equal division numbers
`were compared. Representative histograms are shown in
`Figure 7. CD28 was more rapidly lost in CD8 than in CD4 T
`cells. On day 31, most CD8, but few CD4, Tcells had lost CD28.
`Expression of CD85j emerged in a subset of CD8 Tcells after 3 to
`4 weeks of culture, but was nearly absent in CD4 Tcells. Both T-
`cell subsets gained HLA-DR expression early during culture,
`and expression in CD8 Tcells again preceded that in CD4 Tcells.
`In general, phenotypic shifts were more pronounced and
`earlier in older individuals (data not shown).
`
`Discussion
`
`In this manuscript, we describe that CD4 T cells are more
`resistant to phenotypic and functional changes with aging
`than CD8 T cells. CD4 and CD8 T cells undergo the same
`principal phenotypic shifts; however, the rate at which they
`
`UPenn Ex. 2055
`Miltenyi v. UPenn
`IPR2022-00855
`115
`
`

`

`116
`
`M. Czesnikiewicz-Guzik et al.
`
`occur or accumulate with age is vastly different. The
`increased susceptibility of CD8 T

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket