Modelling T-cell memory by genetic marking of memory T cells in vivo

Nature ◽  
10.1038/21208 ◽  
1999 ◽  
Vol 399 (6736) ◽  
pp. 593-597 ◽  
Author(s):  
Joshy Jacob ◽  
David Baltimore
2013 ◽  
Vol 210 (7) ◽  
pp. 1463-1479 ◽  
Author(s):  
Salvador Iborra ◽  
Manuel Ramos ◽  
David M. Arana ◽  
Silvia Lázaro ◽  
Francisco Aguilar ◽  
...  

Signals from the TCR that specifically contribute to effector versus memory CD8+ T cell differentiation are poorly understood. Using mice and adoptively transferred T lymphocytes lacking the small GTPase N-ras, we found that N-ras–deficient CD8+ T cells differentiate efficiently into antiviral primary effectors but have a severe defect in generating protective memory cells. This defect was rescued, although only partly, by rapamycin-mediated inhibition of mammalian target of rapamycin (mTOR) in vivo. The memory defect correlated with a marked impairment in vitro and in vivo of the antigen-mediated early induction of T-box transcription factor Eomesodermin (Eomes), whereas T-bet was unaffected. Besides N-ras, early Eomes induction in vitro required phosphoinositide 3-kinase (PI3K)–AKT but not extracellular signal-regulated kinase (ERK) activation, and it was largely insensitive to rapamycin. Consistent with N-ras coupling Eomes to T cell memory, retrovirally enforced expression of Eomes in N-ras–deficient CD8+ T cells effectively rescued their memory differentiation. Thus, our study identifies a critical role for N-ras as a TCR-proximal regulator of Eomes for early determination of the CD8+ T cell memory fate.


2000 ◽  
Vol 74 (24) ◽  
pp. 11690-11696 ◽  
Author(s):  
Jan P. Christensen ◽  
Peter C. Doherty ◽  
Kristen C. Branum ◽  
Janice M. Riberdy

ABSTRACT The recall of CD8+ T-cell memory established by infecting H-2b mice with an H1N1 influenza A virus provided a measure of protection against an extremely virulent H7N7 virus. The numbers of CD8+ effector and memory T cells specific for the shared, immunodominant DbNP366epitope were greatly increased subsequent to the H7N7 challenge, and though lung titers remained as high as those in naive controls for 5 days or more, the virus was cleared more rapidly. Expanding the CD8+ memory T-cell pool (<0.5 to >10%) by sequential priming with two different influenza A viruses (H3N2→H1N1) gave much better protection. Though the H7N7 virus initially grew to equivalent titers in the lungs of naive and double-primed mice, the replicative phase was substantially controlled within 3 days. This tertiary H7N7 challenge caused little increase in the magnitude of the CD8+ DbNP366 + T-cell pool, and only a portion of the memory population in the lymphoid tissue could be shown to proliferate. The great majority of the CD8+ DbNP366 + set that localized to the infected respiratory tract had, however, cycled at least once, though recent cell division was shown not to be a prerequisite for T-cell extravasation. The selective induction of CD8+ T-cell memory can thus greatly limit the damage caused by a virulent influenza A virus, with the extent of protection being directly related to the number of available responders. Furthermore, a large pool of CD8+ memory T cells may be only partially utilized to deal with a potentially lethal influenza infection.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 600-600
Author(s):  
W. Nicholas Haining ◽  
D. Neuberg ◽  
H. Keczkemethy ◽  
J. Evans ◽  
S. Rivoli ◽  
...  

Abstract Despite profound T cell immunodeficiency, most patients treated with chemotherapy (CT) do not succumb to infection. The basis for residual protective immunity in lymphopenic patients is not known. Understanding the mechanisms underlying persistent immunity in some cancer patients might suggest treatment strategies to enhance immune competence in all. We therefore prospectively studied T cell homeostasis in 73 children (median age 4y; range 1–17) with ALL receiving a protracted, 2-year chemotherapy regimen. T cell frequency and phenotype and TREC levels were measured at diagnosis and every 5 months on therapy, and compared to an age-matched cohort of 805 healthy children. Twenty-three patients (32%) had high-risk features. All patients received a 5-drug induction, consolidation, and a continuation phase with pulses of vincristine and prednisone. High risk patients also received doxorubicin during consolidation. Patients received childhood vaccinations prior to diagnosis and standard PCP prophylaxis during therapy. Most patients had profound defects in CD4 and CD8 T cell compartments at diagnosis that failed to recover during the 2 years of therapy. Absolute counts of CD4 and CD8 T cells in patients remained below 10th %ile for age in 77% and 86% respectively at all timepoints. We evaluated whether this T cell defect affected both naive and memory compartments. Compared to healthy children, the fraction of CD4 cells with a naive (CD45RA/CD62L) phenotype was markedly reduced (77% of patients with <10th %ile values). Consistent with the reduction in naive T cells, thymopoiesis (measured by TREC levels) was significantly lower in ALL patients than in normal controls (p<0.0001). In contrast, the proportion of CD4 cells with a memory phenotype was elevated, and 60% of children had CD45RO% greater than 90th %ile for age. To confirm that this represented preservation of bona fide T cell memory, we studied functional T cell memory in vitro and in vivo. T cell responses to vaccine Ags administered prior to ALL therapy were measured in a sub-set of 10 patients using a novel CFSE-based assay. Proliferation to the vaccine Ags Tetanus and Varicella Zoster Virus was significantly higher in patients than in pediatric controls (p<0.05), suggesting that memory T cells specific for previously-encountered Ags were enriched in the T cell pool. As an in vivo measure of immunity we recorded the frequency of serious infections (positive blood culture or PCP infection). Despite profound T cell lymphopenia, the rate of infection following induction was low at 0.14 infections/patient-year, suggesting that pathogen-specific immunity was largely maintained. We demonstrate that naive and memory T cells show differential sensitivity to CT. Naive T cells and/or thymocytes are most profoundly affected, while the memory T cell pool is relatively spared. Although persistence of T cell memory offers protection from previously encountered pathogens, without replenishment of the T cell pool with naive cells, response to new antigens is likely to be limited. Interventions that specifically protect thymopoiesis and/or naive T cells such as IL7, KGF or androgen inhibitors may lessen the impact of CT on the peripheral T cell pool and improve functional immunity in cancer patients.


1991 ◽  
Vol 174 (5) ◽  
pp. 969-974 ◽  
Author(s):  
D Gray ◽  
P Matzinger

Immunological memory has generally been ascribed to the development of long-lived memory cells that can persist for years in the absence of renewed antigenic encounter. In the experiments reported here, we have adoptively transferred memory T cells in the presence and absence of priming antigen and assessed their functional survival. The results indicate that, in contrast to the traditional view, the maintenance of T cell memory requires the presence of antigen, suggesting that memory, like tolerance, is an antigen-dependent process rather than an antigen-independent state.


Blood ◽  
2005 ◽  
Vol 106 (5) ◽  
pp. 1749-1754 ◽  
Author(s):  
W. Nicholas Haining ◽  
Donna S. Neuberg ◽  
Heather L. Keczkemethy ◽  
John W. Evans ◽  
Stephen Rivoli ◽  
...  

Abstract Despite profound T-cell immunodeficiency, most patients treated with chemotherapy do not succumb to infection. The basis for residual protective immunity in lymphopenic patients is not known. We prospectively measured T-cell numbers, thymopoiesis, and T-cell memory in 73 children undergoing a 2-year chemotherapy regimen for acute lymphoblastic leukemia (ALL) and compared them to an age-matched cohort of 805 healthy children. Most patients had profound defects in CD4 and CD8 T-cell numbers at diagnosis that did not recover during the 2 years of therapy. Thymic output and the fraction of naive T cells were significantly lower than in healthy controls. However, the remaining T-cell compartment was enriched for antigen-experienced, memory T cells defined both by phenotype and by function. This relative sparing of T-cell memory may, in part, account for the maintenance of protective immunity in lymphopenic patients treated for ALL. Moreover, because the memory T-cell compartment is least affected by ALL and its treatment, strategies to induce immunity to pathogens or tumor antigens in cancer patients may be most successful if they seek to expand pre-existing memory T cells. (Blood. 2005; 106:1749-1754)


2007 ◽  
Vol 179 (3) ◽  
pp. 1988-1995 ◽  
Author(s):  
Tamara Tuuminen ◽  
Eliisa Kekäläinen ◽  
Satu Mäkelä ◽  
Ilpo Ala-Houhala ◽  
Francis A. Ennis ◽  
...  

2021 ◽  
Vol 23 (1) ◽  
pp. 37
Author(s):  
Anjuman Ara ◽  
Aizhang Xu ◽  
Khawaja Ashfaque Ahmed ◽  
Scot C. Leary ◽  
Md. Fahmid Islam ◽  
...  

Energy sensors mTORC1 and AMPKα1 regulate T-cell metabolism and differentiation, while rapamycin (Rapa)-inhibition of mTORC1 (RIM) promotes T-cell memory. However, the underlying pathway and the role of AMPKα1 in Rapa-induced T-cell memory remain elusive. Using genetic and pharmaceutical tools, we demonstrate that Rapa promotes T-cell memory in mice in vivo post Listeria monocytogenesis rLmOVA infection and in vitro transition of effector T (TE) to memory T (TM) cells. IL-2- and IL-2+Rapa-stimulated T [IL-2/T and IL-2(Rapa+)/T] cells, when transferred into mice, differentiate into short-term IL-7R−CD62L−KLRG1+ TE and long-lived IL-7R+CD62L+KLRG1− TM cells, respectively. To assess the underlying pathways, we performed Western blotting, confocal microscopy and Seahorse-assay analyses using IL-2/T and IL-2(Rapa+)/T cells. We determined that IL-2(Rapa+)/T cells activate transcription FOXO1, TCF1 and Eomes and metabolic pAMPKα1(T172), pULK1(S555) and ATG7 molecules and promote mitochondrial biogenesis and fatty-acid oxidation (FAO). We found that rapamycin-treated AMPKα-deficient AMPKα1-KO IL-2(Rapa+)/TM cells up-regulate transcription factor HIF-1α and induce a metabolic switch from FAO to glycolysis. Interestingly, despite the rapamycin treatment, AMPKα-deficient TM cells lost their cell survival capacity. Taken together, our data indicate that rapamycin promotes T-cell memory via transcriptional FOXO1-TCF1-Eomes programs and AMPKα1-ULK1-ATG7 metabolic axis, and that AMPKα1 plays a critical role in RIM-induced T-cell memory.


Sign in / Sign up

Export Citation Format

Share Document