Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Immunol. Aug 15, 2023; 13(2): 11-22
Published online Aug 15, 2023. doi: 10.5411/wji.v13.i2.11
Stem cell-like memory T cells: Role in viral infections and autoimmunity
Meenakshi Sachdeva, Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
Shivangi Taneja, Naresh Sachdeva, Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
ORCID number: Meenakshi Sachdeva (0000-0002-4062-3064); Naresh Sachdeva (0000-0001-6289-4749).
Author contributions: Sachdeva M co-conceptualized the idea, performed the literature search, and wrote the manuscript; Taneja S supported in literature search, manuscript writing, and formatting; Sachdeva N conceptualized the idea, and supervised and edited the manuscript; and all authors have read and approved the final manuscript.
Conflict-of-interest statement: The authors declare no competing interests.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Naresh Sachdeva, MSc, PhD, Professor, Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Sector-12, Chandigarh 160012, India. sachdeva.naresh@pgimer.edu.in
Received: April 28, 2023
Peer-review started: April 28, 2023
First decision: May 30, 2023
Revised: July 6, 2023
Accepted: July 27, 2023
Article in press: July 27, 2023
Published online: August 15, 2023
Processing time: 107 Days and 18.3 Hours

Abstract

Stem cell-like memory T (TSCM) cells possess stem cell properties including multipotency and self-renewal and are being recognized as emerging players in various human diseases. Advanced technologies such as multiparametric flowcytometry and single cell sequencing have enabled their identification and molecular characterization. In case of chronic viral diseases such as human immunodeficiency virus-1, CD4+ TSCM cells, serve as major reservoirs of the latent virus. However, during immune activation and functional exhaustion of effector T cells, these cells also possess the potential to replenish the pool of functional effector cells to curtail the infection. More recently, these cells are speculated to play important role in protective immunity following acute viral infections such as coronavirus disease 2019 and might be amenable for therapeutics by ex vivo expansion. Similarly, studies are also investigating their pathological role in driving autoimmune responses. However, there are several gaps in the understanding of the role of TSCM cells in viral and autoimmune diseases to make them potential therapeutic targets. In this minireview, we have attempted an updated compilation of the dyadic role of these complex TSCM cells during such human diseases along with their biology and transcriptional programs.

Key Words: Stem cell-like memory T cells, Viral infections, Autoimmune diseases, Effector T cells, Memory T cells

Core Tip: This article discusses the contrasting roles of stem cell-like memory T (TSCM) cells during chronic viral infections and autoimmune diseases. During chronic viral infections, such as human immunodeficiency virus-1, the TSCM cells serve as reservoirs for latent viruses, which can be activated to make them susceptible to cytotoxic T cell responses. However, during acute viral infections, the TSCM cells have the ability to replenish the diminished effector T cell population. In autoimmune diseases, like type-1 diabetes, these cells contribute to the disease pathogenesis by persistent generation of autoreactive effector T cells. A better understanding of the key signaling pathways and mediators regulating TSCM cells could lead to novel approaches to target or manipulate these cells for immunotherapeutic applications.



INTRODUCTION

Memory T cells with stem cell properties were identified more than a decade ago and since then they have gained tremendous attention as important players in several disease conditions owing to their self-renewal and multipotent properties[1,2]. Typically, the stem cell-like memory T (TSCM) represent the earliest developmental stage of memory T cells capable of differentiation upon antigen re-challenge[1]. These cells proliferate extensively and have long lifespans because of the maintenance of the length of their telomeres attributed to an active telomerase that preserves the potency of these cells[3]. TSCM cells express a combination of molecules on their surface including: CD45RA, CD45RO, CD62L, CCR7, CD27, CD28, CD127, CD95, and CD122. These cells also express CXCR3, Bcl-2, and LFA-1 and display characteristics that are intermediate between naive and central memory T-cells.

In chronic viral infections, TSCM cells have been known to harbor viruses like human immunodeficiency virus-1 (HIV-1) or hepatitis C virus (HCV) and serve as their long-term latent reservoirs (Figure 1). This T cell subset harbors the highest amount of proviral DNA and contributes significantly as an HIV-1 reservoir as compared to other reservoirs[4]. The stem cell signaling pathways operational in these cells have been elucidated in a few studies[5]. Studies on cancer stem cells have further shed light on the role of canonical Wnt/Notch signaling as a fundamental pathway mediating self-renewal of these cells which might be probably involved in latency during HIV-1 infection. Wnt proteins are secreted glycoproteins that bind and activate the seven transmembrane receptors, Frizzled. Binding of Wnt to Frizzled receptor results in the activation of cytosolic proteins belonging to the Dishevelled family of proteins, leading to internalization of the Frizzled receptor. Activation of Wnt after binding to its receptors leads to downstream signaling events that further lead to the translocation of cytosolic β-catenin proteins into the nucleus. These proteins act as a transcriptional coactivators of factors belonging to the T cell factor 1 (TCF-1)/lymphoid enhancer factor (LEF) family leading to the transcription of Wnt/β-catenin target genes[6,7]. Turning off these pathways may favor memory T cell differentiation, influencing HIV-1 replication and making these cells susceptible to either anti-retroviral drugs or cytotoxic T cells. Besides Wnt/β-catenin pathways, other pathways such as Notch or Sonic Hedgehog pathways might also be functional in these cells, however, this remains to be an important area of future research.

Figure 1
Figure 1 Varying roles of stem cell-like memory T cells during chronic (human immunodeficiency virus-1) and acute (COVID-19) viral infections and autoimmune diseases (type 1 diabetes). In chronic human immunodeficiency virus-1 (HIV-1) infection, the CD4+ stem cell-like memory T (TSCM) cells act as latent reservoirs of HIV-1, whereas the CD8+ TSCM cells exhibit an exhausted phenotype with upregulation of inhibitory molecules like programmed cell death 1. In case of post coronavirus disease 2019 infection or vaccination, viral specific TSCM cells are generated that provide long term protective immunity. In autoimmune responses, such as during type 1 diabetes, TSCM cells generate autoreactive effector-memory (TEM) and effector T (Teff) cell subsets that perpetuate the ongoing inflammatory responses. HIV-1: Human immunodeficiency virus-1; PD-1: Programmed cell death 1; T1D: Type 1 diabetes; TSCM: Stem cell-like memory T; COVID: Coronavirus disease 2019.

Unlike viral infections, autoimmune diseases are characterized by an exaggerated immune response against self-antigens involving various cell types including TSCM cells (Figure 1). Studies in the past few years have drawn attention to the role of TSCM cells in the sustenance of autoimmune cells for longer durations, posing a big hurdle toward immunotherapeutic approaches. It is believed that owing to their high differentiation potential these cells continue to generate effector T cells that further exacerbate the immune homeostasis and contribute to disease pathogenesis. An understanding of the key transcription factors and their signaling mechanisms could lead to the design of approaches to specifically target these cells.

Although it has been more than a decade since the TSCM cells were first identified, there is a scarcity of literature on the role of these cells in various diseases suggesting further research on this important T cell subset through a greater number of studies. In this minireview, we have tried to compile the available literature on the role of TSCM cells in common viral infections like coronavirus disease 2019 (COVID-19), HIV-1, HCV, and autoimmune diseases like type 1 diabetes (T1D) and attempted to suggest a few strategies to target these cells.

STEM CELL-LIKE MEMORY T CELLS AND COVID-19 INFECTION: EMERGING CONCEPTS

Severe acute respiratory syndrome virus (SARS-CoV-2), a new virus that emerged in 2019 was responsible for the COVID-19 pandemic which caused havoc worldwide. The era witnessed the rapid development of a wide variety of vaccines against COVID-19 for the induction of long-term protective immunity in the general population, an attribute of immunological memory. The infection subsides after clearance of the virus leaving a small proportion of T and B memory cells that have the potential to differentiate into virus-specific effector cells and neutralizing antibodies in response to a new episode of infection. Studies using MHC multimers and activation-induced markers have demonstrated that the TSCM cells are generated in both the CD4 and CD8 compartments of T cells peaking approximately at 120 d and persisting up to 8 mo to 10 mo post-infection[8,9].

Further, gene expression profiling and using computational framework methods, the host genetic responses that influence immune cell subsets including CD8+ memory T cells were studied in COVID-19[10]. The CXCR6+ memory CD8+T cells were enriched in genes involved in cytokine, chemokine, T cell activation, proliferation, and migration, indicating their potential contribution to protection from the severe form of COVID-19. In fact, a timely generation of TSCM cells determines the durability of vaccine-induced memory CD8+ T-cell responses[11]. Hence, there appears to be a direct correlation between protective immunity and memory stem cells either after natural infection with the virus or post-vaccination. Similarly, in another study, BNT162b2 vaccine induced various immune-competent cell populations including TSCM cells that serve as indicators of a protective immune response[12]. The frequencies of both CD4+ and CD8+ TSCM cells correlated positively with antibody titers as well[13]. Since the TSCM cells are associated with virus control and induction of long-lived immunity, it may be assumed that these cells are an indicator of success of vaccine efficacy. Whether the TSCM cells or their progenitors are able to migrate to regional lymph nodes or mucosal sites of viral entry is another parameter of durable protection. Nevertheless, a deeper understanding of the genesis and sustenance of TSCM cells could provide the basis for the improvement and development of newer vaccines against COVID-19 and its variants.

STEM CELL-LIKE MEMORY T CELLS AND HIV-1: A PARADIGM

During HIV-1 infection, the virus utilizes a variety of cells like tissue macrophages, myeloid cells, brain microglial cells, gut epithelial cells, and hematopoietic stem cells (HSCs) and more importantly CD4+ TSCM cells as its latent reservoir, making it inaccessible to the antiretroviral therapy (ART). Consequently, the patients need to be on life-long ART since any interruption in their therapy leads to virus rebound from these reservoirs. Hence, this reservoir poses a big hindrance to the cure of HIV infection. The establishment of reservoirs begins early during the acute phase of infection and keeps on accumulating during the subsequent phases[14]. While the establishment of viral reservoirs is almost inevitable, early and timely initiation of ART leads to a reduction in the size of this reservoir[15,16]. Earlier research in this regard suggested that prevention of reservoirs is not possible even if therapy is initiated before the presentation of symptoms, as early as 10 days despite the successful control of plasma viremia[17]. In this context, the expansion of the TSCM cells has been documented to be correlated to HIV disease progression[4]. The contribution of TSCM to the total HIV reservoir becomes appreciable when the total CD4+ T cell population contracts with disease progression and hence contributes largely to the long-term persistence of the virus[18]. Earlier, one of our own studies showed an increased frequency of TSCM cells in HIV patients with a break in ART as compared to patients on regular ART. It may be noted that a greater number of these cells might not be protective since the CD8+ TSCM also expressed high levels of inhibitory molecule, programmed cell death 1 (PD-1) on their surface making them functionally incompetent[19]. In this scenario, latency reversal agents (LRA) in combination with PD-1 blockade might serve to target viral reservoirs besides revival of cellular functions. CD8+ T cells expressing PD-1 and TCF-1 retain long-term survival capacities and proliferation potential, hence they constitute an important armory against the virus. These cells hold greater promise in T cell therapies since they have higher cytolytic potential of these cells and could be helpful in controlling viral replication. These cells have been associated with slow progression in pediatric HIV-positive patients[20]. The HIV-specific CD8+ T cell population expressing PD-1 and TCF-1 represents a promising target for T cell therapies because of the long-term survival and proliferative capacity of these stem-like cells.

The cell signaling pathways that aid in the establishment of latency, are incompletely understood. It is suggested that the process of formation of HIV reservoirs mimics those that are operational in the maintenance of T cell memory, hence insights from the studies in cancer stem cells might shed light into their development programs. Strategies to reactivate viruses from this reservoir have been continually evolving and various hypotheses to explain the generation and maintenance of these reservoirs have been put forward. After encountering the antigen, a state of chronic activation is achieved. During this stage, these TSCM may undergo a process of asymmetric division that replenishes the pool of latent reservoirs[21]. Another hypothesis is that gamma chain cytokines such as IL-7 and IL-15 may lead to the homeostatic proliferation of these cells owing to a higher expression of cytokine receptors on these cells[22]. The observation that the cells expressing PD-1 retain higher levels of HIV-DNA as compared to cells that are PD-1 Low, suggests that certain pathways collating with negative regulators or inhibitory receptors might be favoring the sustenance of TSCM cells as viral reservoirs[23].

Several strategies have been tried including activating the cells carrying the latent HIV-1 with some LRA so that these cells become targets of the cytolytic machinery of the host[24]. Many such agents have been identified and include histone deacetylase inhibitors[25,26]; disulfiram[27,28]; and the bromodomain-containing protein 4 inhibitor JQ1[29]. These agents either function through the activation of some epigenetic programs or cell signaling pathways that lead to cellular activation. There are other agents that act via activating T cells like protein kinase C agonists such as phorbol esters, prostratin[30], bryostatin-1[31] which reverse latency in cell models. Using 3 assay systems, Bullen et al[24] did a comparison of all these agents, however, none of the agents significantly interrupted the maintenance of latent reservoirs. This could be explained by the resistant nature of these cells to cell death which is inefficiently induced by these agents. The only agent that demonstrated relatively better results was the PKC agonist, bryostatin-1, however, its clinical application is limited due to a high degree of associated cellular toxicity and a lack of pharmacokinetic studies[24].

STEM CELL-LIKE MEMORY T CELLS AND HEPATITIS C INFECTION

Chronic HCV infection is found to be associated with a suboptimal interferon response and exhaustion of cytotoxic T cell responses leading to the viral escape of immune responses and persistence of the virus. Similar to HIV-1 infection, complete clearance of HCV also does not occur with antiviral agents with a consequent dysfunctional state of memory CD8+ T cells. In such a scenario, CD8+ TSCM cells with long life, multipotency, and self-renewal properties can rescue the dysfunctional immune state. It has been observed that higher frequencies of these cells were associated with low viral loads and less immune activation, indicating a role of TSCM cells in protective immunity. More recently, the same group depicted an increased proportion of CD8+ memory stem cells during HCV infection and in HCV/HIV dual-positive patients. In fact, the proportions of these cells had a positive correlation with central memory cells. Also, they were associated with lower viral load, a likely impact on controlling immune activation, and ultimately an effective and protective immunity[32].

It was also observed that vaccination in chimpanzees for HCV-induced memory T cells along with control of virus replication concurrent to attenuation of inhibitory receptors on T cells[33]. Therefore, it could be speculated that such a response would mean more differentiation of the effector T cells from the memory stem cell reservoir.

STEM CELL-LIKE MEMORY T CELLS AND AUTOIMMUNE DISEASES

Since memory stem cells are endowed with self-renewal properties and can differentiate into effector T cells with the maintenance of their pool size, these cells are proposed to play a substantial role in the progression of autoimmune diseases particularly type 1 diabetes, systemic lupus erythematosus (SLE) and juvenile idiopathic arthritis, aplastic anemia (AA) and autoimmune uveitis[34-36].

T1D

A chronic autoimmune disease characterized by a strong islet-associated inflammatory response cause the destruction of pancreatic β-cells leading to deficiency in insulin synthesis[37]. Genetic predisposition and environmental factors be linked although the mechanisms are not completely understood[38]. Another strong hypothesis suggested that underlying microbial infections exacerbate islet inflammation in genetically susceptible individuals[39,40]. The disease leads to the production of islet-specific autoantibodies as well as islet-specific autoreactive CD8+ T cells that are present in large numbers in the insulitis lesions. Such autoreactive CD8+ T cells are mainly responsible for the destruction of β-cells besides the contribution of other immune cells like B cells, macrophages, and NK cells[41].

An important observation in T1D patients is that some of the autoimmune cells display properties that coincide with the appearance of TSCM cells along with the expression of memory cell markers[42]. These cells probably act as stable reservoirs which can differentiate into other autoreactive T cells that might lead to disease progression. One of the earlier studies did show that T1D patients possess memory stem cells with a higher proliferative capacity to generate memory autoimmune T cell repertoire[43]. The mechanisms involved in the generation of memory stem cells are being elucidated. The cytokine, IL-7 has been proposed to play an important role during the differentiation of T cells from naïve precursors[44]. IL-7 promotes the uptake of glucose via overexpression of glucose transporter 1 (GLUT1) and upregulation of the glycolytic enzyme hexokinase 2. The process of pyruvate oxidation in the mitochondria has been linked to the differentiation of TSCM cells from the naïve cell precursors. The T1D patients had a higher expression of GLUT1, which is a hallmark of circulating memory stem cells. Moreover, it has been shown that in such patients, WZB117, an inhibitor of GLUT1 could be used to target glucose uptake and was found to interfere with the generation of memory stem cells[45]. A large number of TSCM cells can be obtained in vitro from the naïve precursors with the use of a combination of IL-7 and IL-15 through the engagement of CD3/CD28[46]. This will facilitate rapid exploitation of these cells in adoptive cell therapy. A higher frequency of both CD4+ and CD8+ circulating TSCM cells has been observed in T1D suggesting a possible association with disease pathogenesis[34]. The effector functions could be attributed to their capacities to secrete the cytokines tumour necrosis factor alpha (TNF-α), interferon-gamma (IFN-γ), and IL-2, enabling them high pathogenicity in T1D.

Besides memory stem cells, closely related cell types or more specifically stem-like autoimmune progenitor T cells have also been shown to play an important role in the pathogenesis of autoimmune diseases. These stem-like cells are CD44highTCF1+CXCR6- which convert into effector CD44highTCF1-CXCR6+ CD8+ T cells which mediate destruction of target cells have been identified in NOD mice[47]. Using single-cell RNA sequencing and clonal analysis, researchers discovered that autoimmune CD8+ T cells represent distinct T cell differentiation phases and distinct traits that drive the switch from autoimmune progenitor to the autoimmune mediator. It could be speculated that strategies that could be employed to target the stem-like progenitors could serve as powerful immunotherapeutic tools in the future for T1D as well[48].

SLE

SLE is a multiorgan systemic disease in which autoantibodies develop against nuclear antigens and affect multiple body organs. Immune complexes comprising antigens and their cognate anti-nuclear antibodies accumulate in various organs such as skin, kidneys, or brain that trigger activation of immune cells leading to increased pro-inflammatory cytokine production that ultimately damages the affected organ[49]. Immunosuppressive therapies are not completely effective in suppressing the formation of autoantibodies making the disease incurable and there is only partial remission[50]. Similar to T1D, an increased frequency of CD4+ TSCM cells exist in patients with SLE. A vicious series of events is initiated with enhanced secretion of cytokines that boost their differentiation of T follicular helper (Tfh) cells that also increase the production of autoantibodies from B cells that further perpetuate the disease[51]. Further, these TSCM cells are more proinflammatory in nature indicative of their contribution to disease pathogenesis.

Aplastic anemia

Aplastic anemia (AA) is an autoimmune disease in which cytotoxic CD8+ T-cells attack autologous HSCs and is mainly characterized by an acquired immune-mediated bone marrow failure syndrome. The disease is sustained by increased levels of type I interferons that polarize the immune system towards Th1 responses in the early stage of the disease. On the other hand, the late-stage and severe form of the disease is characterized by Th17 cells and effector memory CD8+ T-cells[52].

It has been reported that AA patients had a higher frequency of both CD4+ and CD8+ effector memory T cells. This increased frequency was observed in both circulation as well as bone marrow, contributing to abnormal immune status and correlating with disease pathogenesis[53]. Next, there was a correlation between the memory stem cells and disease severity, treatment response, disease relapse, and response to immunosuppressive therapy[35]. An inverse relation was observed between these TSCM cells and their effector memory or effector T cell counterparts since these cells serve as their progenitors. A hyperimmune activation seen in patients with autoimmune diseases might be the reason for an aberrant immune cell repertoire encompassing a higher number of memory stem cells, although this remains to be a hypothesis and needs to be confirmed further. In AA as well, the TSCM cells serve as a source of pro-inflammatory cytokines such as TNF-α, IFN-γ, and IL-2 in response to TCR stimulation. As compared to healthy controls, in vitro stimulation of TCR in AA patients resulted in a higher production of these cytokines from both CD4+ and CD8+ TSCM cells with a greater expansion of these cells as well[54].

As far as memory stem cells are concerned, they also expressed higher PD-1 levels and are thus implicated as an exhausted T cell subset[35]. The expression of PD-1 also gets upregulated during AA and is associated with aberrant regulation of T cell activation[55]. However, this does not suggest a lower functional capacity of these cells since in healthy controls, a higher PD-1 expression correlated with a higher differentiation capacity making these cells self-reactive rather than a dysfunctional state[56]. Therefore, these two properties of TSCM cells, i.e., higher PD-1 expression, and higher cytokine production are evidence of clonal expansion capacity, similar to those of tumor-infiltrating CD8+ T cells. A complete understanding of these cells is warranted in the future and may help in the identification of novel therapeutic targets against these cells which probably applies to other autoimmune diseases as well.

Other autoimmune diseases

There is a scarcity of literature on these cells in other autoimmune diseases such as autoimmune uveitis, immune thrombocytopenia, and juvenile idiopathic arthritis. Autoimmune uveitis is an inflammatory disease that occurs as a result of an autoimmune reaction to self-antigens with foci of ocular lesions. An earlier observation was an increased proportion of CD8+ lymphocytes at the site of inflammation in the retina during the later stage of the disease[57]. Only one study has so far reported a reduced number of CD4+ TSCM cells as compared to healthy controls, however an elevated frequency of CD8+ TSCM cells was observed in the same set of patients[35]. Another observation in these patients was a positive correlation between these T cell subsets. Therefore, TSCM cells could fuel ongoing immune activation and serve as biomarkers of disease severity.

Autoimmune thrombocytopenia (ITP) is characterized by a deficiency of platelets, purpura, and episodes of hemorrhages in which the antiplatelet autoantibodies mediate the destruction of platelets[58]. Evidence suggests that platelets express MHC class I molecules and therefore possess the ability of MHC-dependent antigen processing. These platelets could then be recognized by CD8+ T-cells initiating a series of events culminating in the apoptosis of platelets in ITP[59]. ITP patients have hyper-activated CD8+ T-cells with an increased degranulation and cytotoxic potential[60]. One study investigated the effect of TSCM cells on the pathogenesis of ITP[36]. This study depicted a higher percentage of circulating CD8+ TSCM cells in these patients as compared to healthy controls. These cells play an important role in immune activation and such an aberrant immune response could further upregulate the memory stem cell numbers exacerbating the disease. TSCM cells could also serve as an indicator of treatment efficacy since glucocorticoid treatment significantly decreased their numbers.

APPROACHES TO AUGMENT AND TARGET MEMORY STEM CELLS IN VARIOUS DISEASES

With increased recognition of the role of memory stem cells during infectious as well as autoimmune diseases, various approaches to manipulate these cells have been defined and are summarized in Table 1. Memory stem cells are only scarcely present in the circulation which limits their clinical potential, however, ex vivo expansion of these cells is possible. CD8+ TSCM cells are an important part of the cellular immune response against viruses like HIV-1 which maintain their pool through secretion of IL-2. Cytokines like IL-7 and IL-15 could be used to generate and expand CD8+ TSCM cells from the naïve precursor cells. Using a combination of beads coated with CD3/CD2/CD28-specific antibodies or a combination of cytokines such as IL-7 and IL-15, effective expansion could be achieved in vitro[2,46]. Good manufacturing protocols have led to their expansion making their utility possible in many immunodeficiency conditions. Recently, an ex vivo expansion of these cells has been attempted in a mouse model using urolithin A (UA), a natural metabolite present in pomegranates. The authors have unraveled a novel mechanism in which UA-induced memory stem cell formation through Pink-1 mediated mitophagy through induction of mitochondrial phosphatase Pgam5, that further dephosphorylates β-catenin and drive Wnt signaling in these cells[61].

Table 1 Summary of approaches to target memory stem cells in different disease conditions.
No.
Disease
Approach
Target molecules
Ref.
1HIV-1 infectionEx vivo expansion of CD8+ TSCM cells with IL-7 & IL-15 with CD3/CD28T cell receptor[46]
2Immunodeficiency, tumorEx vivo expansion of CD8+ TSCM cells with urolithin APink-1 mediated mitophagy. Dephosphorylation of β-catenin through action of Wnt signaling [61]
3Tumors and malignanciesUse of TSCM-like CAR-T cells (particularly CAR modified CD8+ T cells)Various tumor/lymphoma antigens[65-67,69]
4Chronic viral infectionsIncreased survival of CD8+ TSCM through Wnt agonistsIncreased expression of TCF1[64]
5HIV-1 infectionNanoparticles or aptamers targeting latent reservoirs by inducing differentiation of TSCM cells to effector memory subsetsKey molecules of Wnt-β-catenin pathway[1]
6HIV-1 infectionShock and kill strategy to reactivate HIV from CD4+ TSCM cells with LRAs + TLR agonists to activate CD8+ T cellsKilling of activated reservoirs by cytotoxic T cells[76]
7HIV-1 infectionBlock and Lock strategy by HIV Tat inhibitorsNo HIV replication by blocking of HIV promoter[77]
8Autoimmune diseasesNanoparticles or siRNA to suppress TSCM cellsInhibition of key molecules of NF-κB signaling[79,80]
9Autoimmune diseasesWnt agonists or shRNA or inhibitors such as PRI-724 to disrupt TSCM cells by inducing differentiation to EM cellsTCF7, CBP/β-catenin pathway[1,81]
10Autoimmune diseasesInduced differentiation of TSCM cells to short lived EM cellsMolecules of mTOR/AMPK pathway[1]
11Type-1 diabetesSelective inhibition of metabolic machinery of TSCM cellsDecreased glucose metabolism through inhibition of GLUT1[5]
12Type-1 diabetesAntibodies to block CCR7 to target TSCM cellsCCR7[86]

Blocking the inhibitory receptors on T cells augments the CD8+ T cell immunity and this kind of immunotherapy has already been applied in the field of cancer and holds great promise for infectious diseases as well[62]. It is widely held that the process of exhaustion drives CD8+ T cells into a transcriptionally distinct lineage that comprises “precursor exhausted” and “terminally exhausted” subsets. The precursor-exhausted CD8+ T cells infrequently express inhibitory receptors and more commonly express certain transcription factors, including TCF-1, encoded by T cell factor 7 (TCF7), which is the downstream transcription factor of the canonical Wnt signaling pathway[63]. CD8+ T cells expressing high levels of TCF1 (TCF1high) exhibit a stem-cell-like phenotype, which maintained a better proliferative capacity and could further differentiate into both TCF1high and TCF1low CD8+ T cell subsets[64]. Therefore, it could be speculated that certain agonists of Wnt signaling pathways can improve TCF-mediated signaling that may promote the survival of these functional cells and prevent their differentiation into an exhausted phenotype and reinvigorate their effector function. Most of the reported studies have been performed in animal models of chronic viral infections and strategies to augment the expression of TCF-1 have shown promising results in such models. This approach could represent a promising immunotherapeutic strategy against not only chronic HIV-1 infection but also other chronic viral infections by reinvigorating the effector function of exhausted CD8+ T cells.

In the case of tumors and malignancies, the same approach has been applied to impart stemness or stem-cell-like phenotype in chimeric antigen receptor (CAR)-T cells, especially CAR-modified CD8+ T cells that have shown great promise[65,66]. In the case of conventional CAR T cells, a major disadvantage was their lack of persistence owing to the acquisition of exhaustion phenotype and loss of proliferative abilities. With advances in genetic manipulation methods and identification of novel transcription factors, it is now possible to generate clinical grade CAR T cells that carry the stem-cell-like phenotype that is functionally more competent, carry the memory phenotype, and are enriched with genes to promote their sustained proliferation. These modified TSCM-like CAR-T cells have shown initial success in clinical trials in various solid tumors, lymphomas, and other malignancies. At the same time, TSCM-like CAR-T cells are being explored in the area of infections and autoimmune diseases as well[67-69]. In one such first preclinical study, hematopoietic stem/progenitor cells derived T cells bearing a protective CD4 chimeric antigen receptor (CD46CD4CAR) were used to redirect against simian HIV (SHIV) infection in pigtail macaques. Owing to their stem-cell-like phenotype, these infused CAR-T cells were found to persist for over two years, retaining their immunological memory and abilities of multilineage engraftment without any toxic side effects[70]. In a relatively recent study, genetically modified, anti-HIV duo-CAR-T cells comprising predominantly CCR7+ TSCM and central memory T cells expressing a few effector molecules were injected in the spleens of HIV-infected humanized mice. These cells were efficient in killing HIV-infected PBMCs including activated CD4+ T cells, monocytes, and macrophages[71]. Likewise in other viral infections, wherever CAR-T cells are developed, there are options for inducing stem-cell-like phenotype in these cells to generate long-term protection against the pathogenic viruses.

Researchers discovered a relationship between the size of the CD8+ TSCM compartment and HIV-1 disease progression in a group of chronically infected subjects. The findings indicate that ART restores a normal frequency of CD8+ TSCM and that natural retention of this subset in the context of untreated HIV-1 infection is associated with enhanced viral control and immunity. As a result, the CD8+ TSCM population may be a predictor of protection in chronic HIV-1 infection, which is directly relevant to the development of T cell-based vaccines, adoptive immunotherapy methods, or the pharmacologic induction of TSCM cells[72-75]. As far as the role of CD4+ TSCM cells as latent reservoirs of viral infections are concerned, they need to be destabilized if these pockets are to be reduced. It has been proposed that targeting key molecules of stem cell signaling pathways such as Wnt through antagonists or silencing RNAs could be used to skew the stem cell signaling machinery to induce differentiation to short-lived effector memory T cell subsets[1]. Similarly, nanoparticles or aptamers against the key molecules of the Wnt-β-catenin pathway, a key driver of stemness could also be utilized to target these reservoirs to reverse the latency. Another strategy could be concomitant activation of the immune system and the LRA so that as soon as the HIV-1 gets reactivated from latency, it becomes a target of CTLs. A combinatorial approach has been investigated in which a synergistic interplay between TLR 8-matured dendritic cells and LRAs is successful in reinvigorating the exhausted CD8+ T cell machinery to kill the latently infected cells[76]. Reactivation of latent HIV-1 could be prevented using a block and lock strategy in which compounds prevent the reactivation of latent HIV-1 from the reservoirs. For example, transcriptional expression of HIV-1 transactivator of transcription (Tat) protein can be inhibited using dCA or Tat inhibitors. In the absence of tat protein, transcription from the integrated HIV-1 promoter does not take place and hence HIV-1 does not replicate[77]. More such inhibitors are under development and active research is going on targeting Tat.

Another approach is to induce a state of deep latency in which the proviral HIV-1 is maintained in a dormant state in these reservoirs so that the virus would not rebound even after discontinuation of anti-retroviral therapy[78]. Since many strategies have been evaluated but none have proven completely successful. Intravenous immunoglobulin (IVIG) has been used as an adjuvant therapy to ART. As a result, there was viral blip and HIV-RNA remained undetectable even after several months of ART discontinuation. IVIG treatment activated HIV from the reservoirs, hence a transient increase in plasma viral load also occurred. This encouraging data envisages studies exploring combination therapies of immunomodulatory agents and LRAs in the future.

In the case of autoimmune disorders, anti-inflammatory and immunosuppressive therapies have limited success and are often associated with relapse, which could be attributed mainly to the persistence of immunological memory. The expression of inflammatory molecules by TSCM cells suggests that key inflammatory signaling molecules can be targeted. In this context, NF-κB signaling which is involved in the maintenance and generation of cytokines by TSCM cells offers the possibility of changing aggressive’ TSCM cells into ‘limited TSCM cells. While, systemic NF-κB blocking may have negative side effects, targeted strategies including the use of nanoparticles could be beneficial for the selective administration of anti-inflammatory agents such as benzoyl aconitine or siRNA targeting NF-κB signaling in inflammatory lesions[79,80].

Similar to latent viral infections, Wnt antagonists or short hairpin RNA (shRNA) can target the key molecules of their respective signaling pathways. One such molecule is TCF7 which offers another option in autoimmune diseases to induce the differentiation of long-lived memory stem cells to short-lived subsets to curtail their sustenance[1]. PRI-724, a Wnt/β-catenin pathway inhibitor, has been shown to prevent the connection between cyclic AMP-response element-binding protein-binding protein (CBP) and β-catenin to direct cellular differentiation instead of proliferation[81]. In one such attempt, TCF-1 deletion did not induce differentiation of TN cells into memory T cells, although effector differentiation was detected and this deletion did not cause any developmental abnormality[82]. These studies imply the need for a better target and timing of intervention in the Wnt signaling pathway while targeting TSCM cells.

Another major pathway, the mTOR/AMPK system regulates TSCM cell stress and survival by balancing glycolysis and mitochondrial metabolism. Reduced mTORC1 activity and increased AMPK activity promote the formation of memory T cells, which rely on mitochondrial metabolism and fatty acid oxidation (FAO)[83,84]. Earlier studies have demonstrated that TRAF6-deficient CD8+ T cells have altered the expression of genes that regulate fatty acid metabolism. In response to growth factor withdrawal, activated CD8+ T cells lacking TRAF6 exhibit impaired AMPK activation and mitochondrial fatty acid oxidation (FAO)[83]. The AMPK pathway thus offers another possibility to target TSCM cells.

Another opportunity to target TSCM cells is selective inhibition or manipulation of metabolic machinery. It has been found that T1D patients have β-cell-specific CD8+ TSCM cells with elevated expression of GLUT1. In vivo, IL-7 is necessary for the maintenance of self-reactive CD8+ TSCM cells. IL-7 increases glucose absorption by overexpressing GLUT1 and the glycolytic enzyme hexokinase 2. In one such attempt, WZB117, a GLUT-1 specific inhibitor, effectively inhibited TSCM cells in T1D patients by decreasing glucose metabolism[45]. Even though metabolism is dependent on glucose absorption, pyruvate oxidation in the mitochondria is required for the formation of TSCM cells from naive precursors. The mitochondrial pyruvate transporter inhibitor UK5099 has been utilized for metabolic manipulation in cancer cells, however, metabolic manipulation of TSCM cells using similar agents could be explored in the future[85].

The chemokine receptor CCR7 and its ligands CCL19 and CCL21 direct T-cell homing and recruitment in lymphoid organs. TSCM cells express high levels of CCR7 which can be targeted using anti-CCR7 antibodies and potential antagonists in localized sites of inflammation or autoimmunity. Treatment of humanized CCR7 mice with anti-human CCR7 monoclonal antibodies, 8H3-16A12, resulted in near-total inhibition of the development of collagen-induced arthritis[86].

Taken together, there are numerous approaches available to target, augment or manipulate TSCM cells under various pathological conditions and many such approaches are being tested in clinical trials.

CONCLUSION

Memory stem cells (TSCM cells) contribute to the latency of viruses on one hand and autoimmunity on the other hand. During chronic viral infections like HIV-1 or HCV, these cells, particularly CD4+ TSCM cells serve as reservoirs where viruses remain in a dormant state inaccessible to any pharmaceutical agent. However, with discontinuation of therapy, the virus rebounds from these pockets and there is an outburst of viremia that overwhelms the limits of our immune system. With the recent development of strategies like shock and kill, block and lock, and deep latency, it is possible to either restrict the virus in these pockets for a long time or activate the virus using LRAs and making them susceptible to cytotoxic T cell (CTL) armory. However, CTLs including TSCM cells are not entirely functional in immunodeficient conditions because of immune exhaustion. Strategies to boost these cells after the blockade of inhibitory receptors or reinvigoration of CTL functions have been experimentally successful. More recently, in acute viral infections such as COVID-19, these cells have been proposed to exacerbate the inflammatory conditions because of increased activation of signaling pathways that increase the proinflammatory cytokines. Therefore, depending on the type of viral infection and the state of immune cells, different strategies could be employed to either restrain or augment these cells.

Autoimmune diseases that result from the loss of tolerance to self-antigens are also influenced by TSCM cells, however, the molecular programs that result in the persistence of these cells are still incompletely elucidated. Experience from the field of cancer stem cells has unraveled strategies to manipulate the metabolic machinery of TSCM cells during autoimmunity to restrict the proliferation of these cells or skew their differentiation to short-lived subsets. More detailed understanding in the context of genetic or epigenetic evaluation in this regard is warranted in the future. The stem-like autoimmune progenitor T cells can continuously seed the niches with self-proliferating memory stem cells, therefore, strategies that target the progenitors could emerge as novel therapeutic approaches in the future.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author’s Membership in Professional Societies: Immunology of Diabetes Society, No. IDS9480; Indian Immunology Society, No. 262; The Cytometry Society of India (TCS), No. L1259; AIDS Society of India, No. ASI1007.

Specialty type: Medicine, research and experimental

Country/Territory of origin: India

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Liu D, China; Sipos F, Hungary S-Editor: Chen YL L-Editor: A P-Editor: Zhao S

References
1.  Gattinoni L, Speiser DE, Lichterfeld M, Bonini C. T memory stem cells in health and disease. Nat Med. 2017;23:18-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 271]  [Cited by in F6Publishing: 364]  [Article Influence: 52.0]  [Reference Citation Analysis (0)]
2.  Lugli E, Gattinoni L, Roberto A, Mavilio D, Price DA, Restifo NP, Roederer M. Identification, isolation and in vitro expansion of human and nonhuman primate T stem cell memory cells. Nat Protoc. 2013;8:33-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 150]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
3.  Ahmed R, Roger L, Costa Del Amo P, Miners KL, Jones RE, Boelen L, Fali T, Elemans M, Zhang Y, Appay V, Baird DM, Asquith B, Price DA, Macallan DC, Ladell K. Human Stem Cell-like Memory T Cells Are Maintained in a State of Dynamic Flux. Cell Rep. 2016;17:2811-2818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 53]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
4.  Buzon MJ, Sun H, Li C, Shaw A, Seiss K, Ouyang Z, Martin-Gayo E, Leng J, Henrich TJ, Li JZ, Pereyra F, Zurakowski R, Walker BD, Rosenberg ES, Yu XG, Lichterfeld M. HIV-1 persistence in CD4+ T cells with stem cell-like properties. Nat Med. 2014;20:139-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 343]  [Cited by in F6Publishing: 337]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
5.  Gao S, Liang X, Wang H, Bao B, Zhang K, Zhu Y, Shao Q. Stem cell-like memory T cells: A perspective from the dark side. Cell Immunol. 2021;361:104273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
6.  Ring A, Kim YM, Kahn M. Wnt/catenin signaling in adult stem cell physiology and disease. Stem Cell Rev Rep. 2014;10:512-525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 131]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
7.  Clevers H, Nusse R. Wnt/β-catenin signaling and disease. Cell. 2012;149:1192-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3740]  [Cited by in F6Publishing: 4112]  [Article Influence: 342.7]  [Reference Citation Analysis (0)]
8.  Dan JM, Mateus J, Kato Y, Hastie KM, Yu ED, Faliti CE, Grifoni A, Ramirez SI, Haupt S, Frazier A, Nakao C, Rayaprolu V, Rawlings SA, Peters B, Krammer F, Simon V, Saphire EO, Smith DM, Weiskopf D, Sette A, Crotty S. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science. 2021;371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1972]  [Cited by in F6Publishing: 1795]  [Article Influence: 598.3]  [Reference Citation Analysis (0)]
9.  Jung S, Appleton E, Ali M, Church GM, Del Sol A. A computer-guided design tool to increase the efficiency of cellular conversions. Nat Commun. 2021;12:1659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
10.  Ma Y, Qiu F, Deng C, Li J, Huang Y, Wu Z, Zhou Y, Zhang Y, Xiong Y, Yao Y, Zhong Y, Qu J, Su J. Integrating single-cell sequencing data with GWAS summary statistics reveals CD16+monocytes and memory CD8+T cells involved in severe COVID-19. Genome Med. 2022;14:16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
11.  Jung S, Jung JH, Noh JY, Kim WJ, Yoon SY, Jung J, Kim ES, Kim HB, Cheong HJ, Park SH, Song KH, Song JY, Shin EC. The generation of stem cell-like memory cells early after BNT162b2 vaccination is associated with durability of memory CD8(+) T cell responses. Cell Rep. 2022;40:111138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
12.  Guerrera G, Picozza M, D'Orso S, Placido R, Pirronello M, Verdiani A, Termine A, Fabrizio C, Giannessi F, Sambucci M, Balice MP, Caltagirone C, Salvia A, Rossini A, Battistini L, Borsellino G. BNT162b2 vaccination induces durable SARS-CoV-2-specific T cells with a stem cell memory phenotype. Sci Immunol. 2021;6:eabl5344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 139]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
13.  Agallou M, Koutsoni OS, Michail M, Zisimopoulou P, Tsitsilonis OE, Karagouni E. Antibody and T-Cell Subsets Analysis Unveils an Immune Profile Heterogeneity Mediating Long-term Responses in Individuals Vaccinated Against SARS-CoV-2. J Infect Dis. 2023;227:353-363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
14.  Chahroudi A, Silvestri G, Lichterfeld M. T memory stem cells and HIV: a long-term relationship. Curr HIV/AIDS Rep. 2015;12:33-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
15.  Vanhamel J, Bruggemans A, Debyser Z. Establishment of latent HIV-1 reservoirs: what do we really know? J Virus Erad. 2019;5:3-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 48]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
16.  Lo Tartaro D, Camiro-Zúñiga A, Nasi M, De Biasi S, Najera-Avila MA, Jaramillo-Jante MDR, Gibellini L, Pinti M, Neroni A, Mussini C, Soto-Ramírez LE, Calva JJ, Belaunzarán-Zamudio F, Crabtree-Ramirez B, Hernández-Leon C, Mosqueda-Gómez JL, Navarro-Álvarez S, Perez-Patrigeon S, Cossarizza A. Effective Treatment of Patients Experiencing Primary, Acute HIV Infection Decreases Exhausted/Activated CD4+ T Cells and CD8+ T Memory Stem Cells. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
17.  Chun TW, Engel D, Berrey MM, Shea T, Corey L, Fauci AS. Early establishment of a pool of latently infected, resting CD4(+) T cells during primary HIV-1 infection. Proc Natl Acad Sci U S A. 1998;95:8869-8873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 618]  [Cited by in F6Publishing: 618]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
18.  Lu X, Song B, Weng W, Xia H, Su B, Wu H, Zhang T, Li W, Gao Y. CD4(+) T Memory Stem Cells Correlate with Disease Progression in Chronically HIV-1-Infected Patients. Viral Immunol. 2017;30:642-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
19.  Sachdeva M, Sharma A, Arora SK. High frequency of memory stem cells with a distinct gene signature in HIV patients with treatment interruption. Scand J Immunol. 2023;97:e13262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
20.  Vieira V, Lim N, Singh A, Leitman E, Dsouza R, Adland E, Muenchhoff M, Roider J, Marin Lopez M, Carabelli J, Giandhari J, Groll A, Jooste P, Prado JG, Thobakgale C, Dong K, Kiepiela P, Prendergast AJ, Tudor-Williams G, Frater J, Walker BD, Ndung'u T, Ramsuran V, Leslie A, Kløverpris HN, Goulder P. Slow progression of pediatric HIV associates with early CD8+ T cell PD-1 expression and a stem-like phenotype. JCI Insight. 2023;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
21.  Rong L, Perelson AS. Asymmetric division of activated latently infected cells may explain the decay kinetics of the HIV-1 latent reservoir and intermittent viral blips. Math Biosci. 2009;217:77-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 89]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
22.  Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, Boucher G, Boulassel MR, Ghattas G, Brenchley JM, Schacker TW, Hill BJ, Douek DC, Routy JP, Haddad EK, Sékaly RP. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15:893-900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1248]  [Cited by in F6Publishing: 1360]  [Article Influence: 90.7]  [Reference Citation Analysis (0)]
23.  Dafonseca S, Chomont N, El Far M, Boulassel R, Routy J, Sékaly R. Purging the HIV-1 reservoir through the disruption of the PD-1 pathway. J Int AIDS Soc. 2010;13 Suppl 3:O15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
24.  Bullen CK, Laird GM, Durand CM, Siliciano JD, Siliciano RF. New ex vivo approaches distinguish effective and ineffective single agents for reversing HIV-1 latency in vivo. Nat Med. 2014;20:425-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 367]  [Cited by in F6Publishing: 391]  [Article Influence: 39.1]  [Reference Citation Analysis (0)]
25.  Shirakawa K, Chavez L, Hakre S, Calvanese V, Verdin E. Reactivation of latent HIV by histone deacetylase inhibitors. Trends Microbiol. 2013;21:277-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 162]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
26.  Bartholomeeusen K, Fujinaga K, Xiang Y, Peterlin BM. Histone deacetylase inhibitors (HDACis) that release the positive transcription elongation factor b (P-TEFb) from its inhibitory complex also activate HIV transcription. J Biol Chem. 2013;288:14400-14407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 66]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
27.  Doyon G, Zerbato J, Mellors JW, Sluis-Cremer N. Disulfiram reactivates latent HIV-1 expression through depletion of the phosphatase and tensin homolog. AIDS. 2013;27:F7-F11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 84]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
28.  Xing S, Bullen CK, Shroff NS, Shan L, Yang HC, Manucci JL, Bhat S, Zhang H, Margolick JB, Quinn TC, Margolis DM, Siliciano JD, Siliciano RF. Disulfiram reactivates latent HIV-1 in a Bcl-2-transduced primary CD4+ T cell model without inducing global T cell activation. J Virol. 2011;85:6060-6064.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 163]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
29.  Banerjee C, Archin N, Michaels D, Belkina AC, Denis GV, Bradner J, Sebastiani P, Margolis DM, Montano M. BET bromodomain inhibition as a novel strategy for reactivation of HIV-1. J Leukoc Biol. 2012;92:1147-1154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 206]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
30.  Korin YD, Brooks DG, Brown S, Korotzer A, Zack JA. Effects of prostratin on T-cell activation and human immunodeficiency virus latency. J Virol. 2002;76:8118-8123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 181]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
31.  DeChristopher BA, Loy BA, Marsden MD, Schrier AJ, Zack JA, Wender PA. Designed, synthetically accessible bryostatin analogues potently induce activation of latent HIV reservoirs in vitro. Nat Chem. 2012;4:705-710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 139]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
32.  Lu X, Song B, Weng W, Su B, Wu H, Cheung AKL, Zhang T, Gao Y. Characteristics of CD8(+) Stem Cell-Like Memory T Cell Subset in Chronic Hepatitis C Virus Infection. Viral Immunol. 2023;36:25-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (1)]
33.  Park SH, Shin EC, Capone S, Caggiari L, De Re V, Nicosia A, Folgori A, Rehermann B. Successful vaccination induces multifunctional memory T-cell precursors associated with early control of hepatitis C virus. Gastroenterology. 2012;143:1048-60.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
34.  Fazeli P, Talepoor AG, Faghih Z, Gholijani N, Ataollahi MR, Ali-Hassanzadeh M, Moravej H, Kalantar K. The frequency of CD4+ and CD8+ circulating T stem cell memory in type 1 diabetes. Immun Inflamm Dis. 2022;10:e715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
35.  Hosokawa K, Muranski P, Feng X, Townsley DM, Liu B, Knickelbein J, Keyvanfar K, Dumitriu B, Ito S, Kajigaya S, Taylor JG 6th, Kaplan MJ, Nussenblatt RB, Barrett AJ, O'Shea J, Young NS. Memory Stem T Cells in Autoimmune Disease: High Frequency of Circulating CD8+ Memory Stem Cells in Acquired Aplastic Anemia. J Immunol. 2016;196:1568-1578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 62]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
36.  Cao J, Zhang C, Han X, Cheng H, Chen W, Qi K, Qiao J, Sun Z, Wu Q, Zeng L, Niu M, Li L, Xu K. Emerging role of stem cell memory-like T cell in immune thrombocytopenia. Scand J Immunol. 2019;89:e12739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
37.  Campbell-Thompson M, Fu A, Kaddis JS, Wasserfall C, Schatz DA, Pugliese A, Atkinson MA. Insulitis and β-Cell Mass in the Natural History of Type 1 Diabetes. Diabetes. 2016;65:719-731.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 249]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
38.  Field LL, Larsen Z, Pociot F, Nerup J, Tobias R, Bonnevie-Nielsen V. Evidence for a locus (IDDM16) in the immunoglobulin heavy chain region on chromosome 14q32.3 producing susceptibility to type 1 diabetes. Genes Immun. 2002;3:338-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
39.  Op de Beeck A, Eizirik DL. Viral infections in type 1 diabetes mellitus--why the β cells? Nat Rev Endocrinol. 2016;12:263-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 185]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
40.  Anderson G. Type I Diabetes Pathoetiology and Pathophysiology: Roles of the Gut Microbiome, Pancreatic Cellular Interactions, and the 'Bystander' Activation of Memory CD8(+) T Cells. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
41.  Coppieters KT, Dotta F, Amirian N, Campbell PD, Kay TW, Atkinson MA, Roep BO, von Herrath MG. Demonstration of islet-autoreactive CD8 T cells in insulitic lesions from recent onset and long-term type 1 diabetes patients. J Exp Med. 2012;209:51-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 448]  [Cited by in F6Publishing: 489]  [Article Influence: 40.8]  [Reference Citation Analysis (0)]
42.  Danke NA, Yang J, Greenbaum C, Kwok WW. Comparative study of GAD65-specific CD4+ T cells in healthy and type 1 diabetic subjects. J Autoimmun. 2005;25:303-311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 72]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
43.  Monti P, Scirpoli M, Rigamonti A, Mayr A, Jaeger A, Bonfanti R, Chiumello G, Ziegler AG, Bonifacio E. Evidence for in vivo primed and expanded autoreactive T cells as a specific feature of patients with type 1 diabetes. J Immunol. 2007;179:5785-5792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 102]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
44.  Monti P, Bonifacio E. Interleukin-7 and type 1 diabetes. Curr Diab Rep. 2014;14:518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
45.  Vignali D, Cantarelli E, Bordignon C, Canu A, Citro A, Annoni A, Piemonti L, Monti P. Detection and Characterization of CD8(+) Autoreactive Memory Stem T Cells in Patients With Type 1 Diabetes. Diabetes. 2018;67:936-945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 48]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
46.  Cieri N, Camisa B, Cocchiarella F, Forcato M, Oliveira G, Provasi E, Bondanza A, Bordignon C, Peccatori J, Ciceri F, Lupo-Stanghellini MT, Mavilio F, Mondino A, Bicciato S, Recchia A, Bonini C. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood. 2013;121:573-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 358]  [Cited by in F6Publishing: 422]  [Article Influence: 35.2]  [Reference Citation Analysis (0)]
47.  Ciecko AE, Schauder DM, Foda B, Petrova G, Kasmani MY, Burns R, Lin CW, Drobyski WR, Cui W, Chen YG. Self-Renewing Islet TCF1(+) CD8 T Cells Undergo IL-27-Controlled Differentiation to Become TCF1(-) Terminal Effectors during the Progression of Type 1 Diabetes. J Immunol. 2021;207:1990-2004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
48.  Gearty SV, Dündar F, Zumbo P, Espinosa-Carrasco G, Shakiba M, Sanchez-Rivera FJ, Socci ND, Trivedi P, Lowe SW, Lauer P, Mohibullah N, Viale A, DiLorenzo TP, Betel D, Schietinger A. An autoimmune stem-like CD8 T cell population drives type 1 diabetes. Nature. 2022;602:156-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 82]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
49.  Lech M, Anders HJ. The pathogenesis of lupus nephritis. J Am Soc Nephrol. 2013;24:1357-1366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 324]  [Cited by in F6Publishing: 301]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
50.  Wilhelm TR, Magder LS, Petri M. Remission in systemic lupus erythematosus: durable remission is rare. Ann Rheum Dis. 2017;76:547-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 72]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
51.  Lee YJ, Park JA, Kwon H, Choi YS, Jung KC, Park SH, Lee EB. Role of Stem Cell-Like Memory T Cells in Systemic Lupus Erythematosus. Arthritis Rheumatol. 2018;70:1459-1469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
52.  Giudice V, Selleri C. Aplastic anemia: Pathophysiology. Semin Hematol. 2022;59:13-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
53.  Hu X, Gu Y, Wang Y, Cong Y, Qu X, Xu C. Increased CD4+ and CD8+ effector memory T cells in patients with aplastic anemia. Haematologica. 2009;94:428-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
54.  Sloand E, Kim S, Maciejewski JP, Tisdale J, Follmann D, Young NS. Intracellular interferon-gamma in circulating and marrow T cells detected by flow cytometry and the response to immunosuppressive therapy in patients with aplastic anemia. Blood. 2002;100:1185-1191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 166]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
55.  Wu H, Miao M, Zhang G, Hu Y, Ming Z, Zhang X. Soluble PD-1 is associated with aberrant regulation of T cells activation in aplastic anemia. Immunol Invest. 2009;38:408-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
56.  Legat A, Speiser DE, Pircher H, Zehn D, Fuertes Marraco SA. Inhibitory Receptor Expression Depends More Dominantly on Differentiation and Activation than "Exhaustion" of Human CD8 T Cells. Front Immunol. 2013;4:455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 187]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
57.  Calder VL, Zhao ZS, Wang Y, Barton K, Lightman SL. Effects of CD8 depletion on retinal soluble antigen induced experimental autoimmune uveoretinitis. Immunology. 1993;79:255-262.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Vrbensky JR, Nazy I, Clare R, Larché M, Arnold DM. T cell-mediated autoimmunity in immune thrombocytopenia. Eur J Haematol. 2022;108:18-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
59.  Qiu J, Liu X, Li X, Zhang X, Han P, Zhou H, Shao L, Hou Y, Min Y, Kong Z, Wang Y, Wei Y, Ni H, Peng J, Hou M. CD8(+) T cells induce platelet clearance in the liver via platelet desialylation in immune thrombocytopenia. Sci Rep. 2016;6:27445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 60]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
60.  Vrbensky JR, Arnold DM, Kelton JG, Smith JW, Jaffer AM, Larché M, Clare R, Ivetic N, Nazy I. Increased cytotoxic potential of CD8(+) T cells in immune thrombocytopenia. Br J Haematol. 2020;188:e72-e76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
61.  Denk D, Petrocelli V, Conche C, Drachsler M, Ziegler PK, Braun A, Kress A, Nicolas AM, Mohs K, Becker C, Neurath MF, Farin HF, Buchholz CJ, Andreux PA, Rinsch C, Greten FR. Expansion of T memory stem cells with superior anti-tumor immunity by Urolithin A-induced mitophagy. Immunity. 2022;55:2059-2073.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 35]  [Reference Citation Analysis (0)]
62.  Alsaab HO, Sau S, Alzhrani R, Tatiparti K, Bhise K, Kashaw SK, Iyer AK. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front Pharmacol. 2017;8:561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 860]  [Cited by in F6Publishing: 1106]  [Article Influence: 158.0]  [Reference Citation Analysis (0)]
63.  Yu Q, Sharma A, Sen JM. TCF1 and beta-catenin regulate T cell development and function. Immunol Res. 2010;47:45-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 54]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
64.  Wu T, Ji Y, Moseman EA, Xu HC, Manglani M, Kirby M, Anderson SM, Handon R, Kenyon E, Elkahloun A, Wu W, Lang PA, Gattinoni L, McGavern DB, Schwartzberg PL. The TCF1-Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci Immunol. 2016;1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 390]  [Article Influence: 48.8]  [Reference Citation Analysis (0)]
65.  Sabatino M, Hu J, Sommariva M, Gautam S, Fellowes V, Hocker JD, Dougherty S, Qin H, Klebanoff CA, Fry TJ, Gress RE, Kochenderfer JN, Stroncek DF, Ji Y, Gattinoni L. Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. Blood. 2016;128:519-528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 265]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
66.  Blaeschke F, Stenger D, Kaeuferle T, Willier S, Lotfi R, Kaiser AD, Assenmacher M, Döring M, Feucht J, Feuchtinger T. Induction of a central memory and stem cell memory phenotype in functionally active CD4(+) and CD8(+) CAR T cells produced in an automated good manufacturing practice system for the treatment of CD19(+) acute lymphoblastic leukemia. Cancer Immunol Immunother. 2018;67:1053-1066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 105]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
67.  Arcangeli S, Falcone L, Camisa B, De Girardi F, Biondi M, Giglio F, Ciceri F, Bonini C, Bondanza A, Casucci M. Next-Generation Manufacturing Protocols Enriching T(SCM) CAR T Cells Can Overcome Disease-Specific T Cell Defects in Cancer Patients. Front Immunol. 2020;11:1217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 68]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
68.  Wang Y, Qiu F, Xu Y, Hou X, Zhang Z, Huang L, Wang H, Xing H, Wu S. Stem cell-like memory T cells: The generation and application. J Leukoc Biol. 2021;110:1209-1223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
69.  Meyran D, Zhu JJ, Butler J, Tantalo D, MacDonald S, Nguyen TN, Wang M, Thio N, D'Souza C, Qin VM, Slaney C, Harrison A, Sek K, Petrone P, Thia K, Giuffrida L, Scott AM, Terry RL, Tran B, Desai J, Prince HM, Harrison SJ, Beavis PA, Kershaw MH, Solomon B, Ekert PG, Trapani JA, Darcy PK, Neeson PJ. T(STEM)-like CAR-T cells exhibit improved persistence and tumor control compared with conventional CAR-T cells in preclinical models. Sci Transl Med. 2023;15:eabk1900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 23]  [Reference Citation Analysis (0)]
70.  Zhen A, Peterson CW, Carrillo MA, Reddy SS, Youn CS, Lam BB, Chang NY, Martin HA, Rick JW, Kim J, Neel NC, Rezek VK, Kamata M, Chen ISY, Zack JA, Kiem HP, Kitchen SG. Correction: Long-term persistence and function of hematopoietic stem cell-derived chimeric antigen receptor T cells in a nonhuman primate model of HIV/AIDS. PLoS Pathog. 2018;14:e1006891.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
71.  Anthony-Gonda K, Ray A, Su H, Wang Y, Xiong Y, Lee D, Block A, Chilunda V, Weiselberg J, Zemelko L, Wang YY, Kleinsorge-Block S, Reese JS, de Lima M, Ochsenbauer C, Kappes JC, Dimitrov DS, Orentas R, Deeks SG, Rutishauser RL, Berman JW, Goldstein H, Dropulić B. In vivo killing of primary HIV-infected cells by peripheral-injected early memory-enriched anti-HIV duoCAR T cells. JCI Insight. 2022;7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 9]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
72.  Ribeiro SP, Milush JM, Cunha-Neto E, Kallas EG, Kalil J, Somsouk M, Hunt PW, Deeks SG, Nixon DF, SenGupta D. The CD8⁺ memory stem T cell (T(SCM)) subset is associated with improved prognosis in chronic HIV-1 infection. J Virol. 2014;88:13836-13844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
73.  Munusamy Ponnan S, Hayes P, Fernandez N, Thiruvengadam K, Pattabiram S, Nesakumar M, Srinivasan A, Kathirvel S, Shankar J, Goyal R, Singla N, Mukherjee J, Chatrath S, Gilmour J, Subramanyam S, Prasad Tripathy S, Swaminathan S, Hanna LE. Evaluation of antiviral T cell responses and TSCM cells in volunteers enrolled in a phase I HIV-1 subtype C prophylactic vaccine trial in India. PLoS One. 2020;15:e0229461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
74.  Li Y, Wu D, Yang X, Zhou S. Immunotherapeutic Potential of T Memory Stem Cells. Front Oncol. 2021;11:723888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
75.  Wu K, Zhang S, Zhang X, Li X, Hong Z, Yu F, Liu B, Pan T, Huang Z, Tang XP, Cai W, Xia J, Zhang H, Zhang Y, Li L. IL-21 Expands HIV-1-Specific CD8(+) T Memory Stem Cells to Suppress HIV-1 Replication In Vitro. J Immunol Res. 2019;2019:1801560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
76.  Rochat MA, Schlaepfer E, Speck RF. Promising Role of Toll-Like Receptor 8 Agonist in Concert with Prostratin for Activation of Silent HIV. J Virol. 2017;91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
77.  Mousseau G, Valente S. Strategies to Block HIV Transcription: Focus on Small Molecule Tat Inhibitors. Biology (Basel). 2012;1:668-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
78.  Elsheikh MM, Tang Y, Li D, Jiang G. Deep latency: A new insight into a functional HIV cure. EBioMedicine. 2019;45:624-629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 47]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
79.  Zhou HF, Yan H, Pan H, Hou KK, Akk A, Springer LE, Hu Y, Allen JS, Wickline SA, Pham CT. Peptide-siRNA nanocomplexes targeting NF-κB subunit p65 suppress nascent experimental arthritis. J Clin Invest. 2014;124:4363-4374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 77]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
80.  Gai W, Hao X, Zhao J, Wang L, Liu J, Jiang H, Jin H, Liu G, Feng Y. Delivery of benzoylaconitine using biodegradable nanoparticles to suppress inflammation via regulating NF-κB signaling. Colloids Surf B Biointerfaces. 2020;191:110980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
81.  Mavigner M, Zanoni M, Tharp GK, Habib J, Mattingly CR, Lichterfeld M, Nega MT, Vanderford TH, Bosinger SE, Chahroudi A. Pharmacological Modulation of the Wnt/β-Catenin Pathway Inhibits Proliferation and Promotes Differentiation of Long-Lived Memory CD4(+) T Cells in Antiretroviral Therapy-Suppressed Simian Immunodeficiency Virus-Infected Macaques. J Virol. 2019;94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
82.  Danilo M, Chennupati V, Silva JG, Siegert S, Held W. Suppression of Tcf1 by Inflammatory Cytokines Facilitates Effector CD8 T Cell Differentiation. Cell Rep. 2018;22:2107-2117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 84]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
83.  Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang LS, Jones RG, Choi Y. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009;460:103-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1075]  [Cited by in F6Publishing: 1200]  [Article Influence: 80.0]  [Reference Citation Analysis (0)]
84.  Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, Larsen CP, Ahmed R. mTOR regulates memory CD8 T-cell differentiation. Nature. 2009;460:108-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1105]  [Cited by in F6Publishing: 1214]  [Article Influence: 80.9]  [Reference Citation Analysis (0)]
85.  Zhong Y, Li X, Yu D, Li Y, Long Y, Yuan Y, Ji Z, Zhang M, Wen JG, Nesland JM, Suo Z. Application of mitochondrial pyruvate carrier blocker UK5099 creates metabolic reprogram and greater stem-like properties in LnCap prostate cancer cells in vitro. Oncotarget. 2015;6:37758-37769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 56]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
86.  Moschovakis GL, Bubke A, Friedrichsen M, Ristenpart J, Back JW, Falk CS, Kremmer E, Förster R. The chemokine receptor CCR7 is a promising target for rheumatoid arthritis therapy. Cell Mol Immunol. 2019;16:791-799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]