Open access peer-reviewed chapter

Thymic Rejuvenation: Are We There Yet?

Written By

Jamilah Abusarah, Fatemeh Khodayarian, Yun Cui, Abed El-Hakim El-Kadiry and Moutih Rafei

Submitted: 09 October 2017 Reviewed: 15 January 2018 Published: 04 July 2018

DOI: 10.5772/intechopen.74048

From the Edited Volume

Gerontology

Edited by Grazia D’Onofrio, Antonio Greco and Daniele Sancarlo

Chapter metrics overview

1,302 Chapter Downloads

View Full Metrics

Abstract

Vaccination is an appealing form of immunotherapy for frail senior patients. However, several studies have shown that in contrast to younger adults, older patients do not effectively respond to vaccines. This phenomenon is greatly attributed to immunosenescence, a hallmark of aging defined by a general decline in immunity caused by thymic involution. Historically, the study of thymic involution brought to attention several factors and components involved in thymopoiesis, as contributors to the phenomena. Depicting the underlying cause(s) of the dramatic changes in the production and properties of the naïve T-cell pool in the event of acute thymic injury or due to inovulation can therefore, help focus the efforts on the best strategy to reverse or overcome these hurdles. Here, we discuss some of the well-studied approaches for rejuvenating the thymus, and introduce interleukin-(IL) 21 as the most recent thymo-stimulatory agent in the field.

Keywords

  • thymus
  • thymocytes
  • thymic epithelial cells
  • thymic involution
  • interleukin-21

1. Introduction

With the advances in health sciences and services, the life expectancy of the general population is increasing. The U.S. Census Bureau estimates that by 2050, one quarter of the U.S. population will be over 60 years old [1]. A similar demographic trend is predicted for both developed and developing countries according to the world health organization [2]. A longer life expectancy is accompanied with the increase in chronic diseases [3], susceptibility to infections [4], prevalence of cancer [5], and certain forms of degenerative diseases [6]. All these challenges put a great burden not only on individuals, but on the whole population and health system [7]. Therefore, it is crucial to adopt new strategies aimed at keeping the aging population healthy.

Several of the prominent challenges facing the efforts to achieve healthy aging are being linked to changes in the immune system [6, 8, 9]. More specifically, the natural process of aging is concomitant with immunosenescence; a degeneration of the immune system caused by thymic atrophy or involution [10]. As a result, the production of T-cells through thymopoiesis is compromised, and the output of newly developed naïve T-cells endowed with the capacity of effectively responding to new antigenic challenges decreases dramatically [11]. For example, accumulated extrinsic and intrinsic T-cell receptor (TCR) signaling defects in aging naïve T-cells are found to impede their response to new antigenic challenges [12]. The elucidation of such changes and defects related to thymic aging can therefore help conclude that the elderly’s optimal responses to vaccines that may strongly rely on the development of strategies aimed at rejuvenating their weakened thinned-out T-cell repertoire. In addition, immunosenescence compromises the patients’ immunocompetency and makes the body more prone to the development of different pathological conditions such as cancer [13]. Although most cancer types can be removed by surgery followed by radiation and/or chemotherapy, these multimodality therapies are primarily effective against localized tumors as opposed to spread metastases [14]. Cancer vaccination represents therefore, an appealing alternative due to its limited toxicity compared to chemo- or radiotherapies and the ability to stimulate the host’s own immune system to seek and destroy metastatic cancer cells [15]. Unfortunately, however, vaccines are less effective at older age due to immunosenescence [16]. Thus, there is an urgent need for the development or improvement of immunotherapies to stimulate and rejuvenate the immune response in the elderly population.

The knowledge of the elements involved in the deterioration of thymic function can help identify potential approaches for protecting or rejuvenating the thymus. We will be summarizing in this chapter most of the concepts related to thymus aging and discuss the most promising developments aimed at stimulating thymic function as a mean to rejuvenate elders’ immunity. In addition, we will highlight the experimental models along with the outcomes of our work on IL-21 as a thymo-stimulatory agent [17, 18, 19]. Optimizing and adopting such concept will strengthen multi-disciplinary collaborations and lead to innovative IL-21-based strategies directed against two key elderly-related health problems: (i) providing superior responsiveness to protective and therapeutic vaccines, and (ii) reducing the incidence and/or relapse of cancer or severe infections in this vulnerable population.

Advertisement

2. Background

2.1. The thymus

Functional immunity relies on the balance between innate immunity as a first responder in the presence of an external insult and the development of a rather specific adaptive immune response [20]. Primary lymphoid organs such as the bone marrow (BM), the fetal liver, and the thymus are the major sites for lymphopoiesis, whereby the common lymphoid progenitors (CLP) proliferate and complete their differentiation into mature effector B and T lymphocytes. The uniqueness of the thymus stems from it being the sole specialized organ where the microenvironment and the cellular content are perfectly orchestrated to support the maturation and development of T-cells [20]. This bilobed organ is located in the thoracic cavity between the heart and the sternum. Histologically, the thymus was believed to be mainly composed of a perivascular space (PVS) and a thymic epithelial space (TES) [21]. The latter is the main site for thymopoiesis and is subdivided into two anatomically and functionally distinct compartments: the cortex and the medulla [22]. These two compartments are connected by a cortico-medullary junction (CMJ), where the entry of lymphoid progenitors and the exit of mature thymocytes take place [23]. Furthermore, each of these two compartments is characterized by the presence of specific thymic epithelial cells (TECs), which guide and support thymocytes at their different differentiation and maturation stages [24]. Interestingly, the cortical (c-) and medullary (m)TECs have distinct expression profile for chemokines and cytokines, transcription factors and costimulatory molecules [25, 26]. As a result, cTECs and mTECs create a unique profile and microenvironment within the cortex and the medulla, respectively, which in turn affect the development and selection of thymocytes [25, 26]. Specifically, within the same compartment, cTECs and mTECs show heterogeneity in their markers and secretary profiles creating distinct microenvironments suitable for specific stages of thymopoiesis [26, 27, 28]. For example, cTECs express the thymoproteasome component β5t [29], Cathepsin L [30], in addition to thymus-specific serine protease, which facilitate the generation of major histocompatibility complex (MHC)-associated self-peptides required for positive selection [30, 31]. Moreover, it is being proposed that the crosstalk between thymocytes and different TECs is fundamental not only for supporting thymopoiesis but for maintaining the thymus homeostasis as well [24, 25, 26, 32]. Particularly, the interaction of developing thymocytes with certain TEC subsets sustains the regeneration of cTECs and mTECs [32, 33, 34].

Thymopoiesis or T-cell development starts with the settling of BM-derived early lymphocyte progenitors (ELP) in the thymus where they proliferate to give rise to early thymic progenitors (ETP) [35, 36]. Upon their interaction with cTECs, rapidly dividing ETPs undergo differentiation to the double-negative (DN) stage while initiating complex gene rearrangements to express a candidate TCR. At that stage, both CD4 and CD8 co-receptors are expressed and the resulting double-positive (DP) thymocyte undergoes positive selection; a process aimed at testing the rearrangement of a functional TCR. If successful, DP thymocytes receive a survival signal and continue their migration to the medulla [37]. Interaction of DP thymocytes with dendritic cells or mTECs is essential to sustain central tolerance as it deletes autoreactive clones leading finally to self-tolerant single-positive (SP) CD4 or CD8 naïve T-cells, which are determined by their restriction to MHCII or MHCI, respectively [38]. Newly developed recent thymic emigrants (RTEs—e.g., naïve CD4 or CD8 T cells) egress to the periphery where they complete their maturation and contribute to establishing a competent peripheral T-cell pool [39, 40, 41].

Despite its undisputed role in the development of T-cells, the thymus is commonly portrayed as a temporary organ due to its gradual decrease in size (progressive and irreversible loss of TES). This natural thymic atrophy/involution consequently results in gradual thymopoiesis decay.

2.2. Acute thymic atrophy

Through history, the thymus has been considered as a mysterious organ with no obvious function or purpose until 1961 [42, 43]. In the late 1800 and early 1900, scientists searching for the cause of unexplained sudden death of healthy babies and children labeled the thymus as the villain [44]. The origin of the confusion started as an attempt to understand and find a cure for what is known today as sudden infant death syndrome (SIDS). For instance, several scientists observed that suddenly dying babies had a much larger thymus in comparison to normal subjects. These observations led to the establishment of a medical condition known as status thymicolymphaticus [45, 46, 47]. As a result, thousands of infants received prophylactic irradiation to reduce the size of their thymus [45, 46, 47]. Interestingly, studies conducted in the mid-1930s on cadavers from ill or malnourished patients revealed that the thymus of these subjects was indeed smaller than those of healthy individuals of the same age [47, 48, 49]. These new findings not only acquitted the thymus from any role in SIDS, but also opened the door to better understand the physiology and role of the thymus [50]. Furthermore, subjects that have undergone ‘precautious’ irradiation in their younger age were later shown to exhibit higher thyroid malignancy and breast cancer risk [51, 52].

Nonchronic thymic atrophy is the first indication for the presence of a given insult(s). There are common morphological characteristics of acute thymic atrophy such as loss of cortical thickness due to sharp decline in cortical thymocytes. Longer exposure to insults leads to thinner, irregular, and lower cortical to medulla ratio, while the CMJ becomes less apparent. Conversely, insult(s) removal is usually sufficient to reverse thymic atrophy and allow the rebound of the cortex and its contents [53, 54, 55]. Such insults include a variety of physiological factors such as pregnancy, malnutrition/starvation, stress as well as pathological factors such as infections, corticosteroids, or immunosuppressive therapy [49, 55, 56, 57, 58]. The exact mechanism(s) and pathways involved in acute thymic atrophy upon exposure to one or more of these conditions is poorly understood and may vary [55]. For instance, the pathophysiological stress from infections upregulates serum glucocorticoids (GC) secretion by the hypothalamus-pituitary adrenal axis and pro-inflammatory cytokines levels [59, 60]. Thymocytes are highly affected by changes in GC levels, which can promote DP thymocytes apoptosis through caspase 8 and caspase 9 activation resulting consequently in lower naïve T-cell output [61, 62]. On the other hand, medical interventions such as irradiation regimens coupled with chemotherapy induce death of thymocytes, DCs and TECs [63, 64]. These interventions also affect the stromal compartment leading to slower TEC renewal [65]. This explains the delay in immune reconstitution following preparative regimen for BM transplantation (BMT), which leaves the patient immunocompromised [66]. It is therefore essential to elucidate and understand the patho-physiological factors triggering thymic atrophy in order to design strategies aimed at reversing their effect.

2.3. Chronic thymic involution

The progressive age-dependant atrophy of the thymus starts at birth and reaches its peak during puberty before continuing at a stable rate throughout life [53, 67]. The two main factors forming the basis of thymic involution are: (i) defects in the pre-thymic hematopoietic progenitor pool or their migration to the thymus, and (ii) the loss of the stromal compartment [68]. The latter factor is particularly problematic as it implies reduction of cTECs [67], loss of tissue structure, and changes in the microenvironmental niche [69]. Consequently, the body’s homeostatic compensatory system helps maintain the existing T-cell pool in the periphery through the expansion of mature T-cells [70, 71], which restricts the ability of aged subjects to benefit from new vaccines [71, 72]. Moreover, aged naïve T-cells are smaller and show defects in antigen recognition, antigen-induced activation and in their ability to proliferate [69, 73, 74]. Such changes in the properties of naïve T-cells and the microenvironment of the thymus can also affect the negative selection process, which in turn, potentiates the emergence of autoreactive or defective T-cells [75]. Besides, the increased longevity of naïve T-cells and the accompanying accumulation of reactive oxygen species lead to reduced TCR diversity and impair the function of effector and memory T cells in the periphery [76, 77, 78] translating into weakened immune responses to infections and immunotherapies such as vaccines [79].

Several factors have been proposed to explain and understand the circumstances taking part in the involution of the thymus. One of the earliest factors to be investigated was the change in the level of sex hormones, which peaks during puberty. GCs are believed to negatively affect both TECs and thymocytes mostly by inducing their apoptosis [80, 81, 82]. In fact, surgical or chemical castration of male mice was sufficient to restore atrophied thymi back to their normal size and function [83]. However, the decline in androgen levels with age is not accompanied with a concomitant reversal of thymic involution [21], suggesting that additional players are involved in chronic thymic atrophy. Some of these factors include: (i) changes in BM-derived T cell progenitors [84], (ii) alterations in the profile of circulating cytokines and factors such as the leukemia inhibitory factor, interleukin (IL)-6, Oncostatin M, and stem cell factor [73], (iii) a blockade in TCR rearrangement, decreased proliferation and increased apoptosis of ETPs [85, 86], and (iv) a shift towards the myeloid lineage in the elderly [87].

Altogether, the aging of the immune system is an ongoing complex process and is related to numerous clinical challenges. To properly tackle these challenges, it is imperative to adequately understand and elucidate the defects in the aging immune system response. Ultimately, acquired knowledge will guide the efforts for designing counteracting therapies to rejuvenate the immune system.

Advertisement

3. Strategies to stimulate intrathymic T-cell development

T-lymphopoiesis remains functional at older age albeit to a limited extent [88]. Thus, enhancing thymic function in aged hosts remains a promising therapeutic goal. The thymus lacks self-renewing progenitors and relies heavily on sustained seeding with BM-derived ETPs [35]. Unfortunately, ETP numbers decline markedly with age due to increased apoptosis rates as well as reduced proliferative capacities [84, 88]. This in turn negatively impacts the delicate thymic stromal compartment, which depends heavily on cross talk interactions with thymic progenitors for its sustained survival [89, 90, 91]. As the magnitude of thymic output correlates closely with the overall number of DN and DP thymocytes [92], pre-conditioning of aged subjects to enhance the development/recovery of these thymic subsets is critical to actively stimulate de novo intrathymic naïve T-cell development.

In an attempt to reverse/block this vicious thymic involution cycle, a variety of strategies including cytokines, growth factors, hormonal therapies, in addition to adoptive transfer of precursor T cells or castration have been tested and reviewed in the literature [93, 94, 95, 96]. However, few of these interventions stoodout as the most propitious including: (i) sex-steroid ablation (SSA), (ii) keratinocyte growth factor (KGF), (iii) ghrelin (GRL), (iv) IL-7 as well as, and (v) IL-21.

3.1. SSA: to reverse aging-induced changes in the thymus

Steroid hormones elicit their effect in the body by interacting with their specific receptors, where they translocate to the nucleus and directly affect gene transcription [97, 98]. In the thymus, both thymocytes and TECs express receptors for steroid hormones and are affected by changes in their levels [99, 100, 101]. For example, testosterone induces DP thymocyte apoptosis through tumor necrosis factor-α upregulation, whereas estrogen binding to receptors expressed by DN thymocytes inhibits their proliferation [100, 102]. Due to the concurrent decline in the size of thymus around puberty, sex steroids are being widely studied as a main factor in thymus involution. For instance, studies designed to evaluate the effect of androgens on T-cell output showed that androgens do indeed have a strong effect on the output of T-cells [94, 101]. Specifically, the reported decline in thymic output is mediated mainly by the sensitivity of TECs to androgens [101, 102, 103]. Therefore, adopting a plan to promote thymic activity by antagonizing or preventing the effect of sex hormones on the thymus is perceived as both rational and reasonable [96]. SSA can indeed be achieved either surgically or chemically. Both approaches show positive impact on thymic function, cellularity, as well as architecture, and can significantly enhance thymopoiesis [96, 104]. For instance, castration conducted in different animal models resulted in an increase in thymic output, T-cell responses and helped sustain the size and function of the thymus [105, 106, 107, 108]. More specifically, surgical castration in old mice led to rapid reversal of thymic atrophy and restoration of SP and DP thymocyte to comparable levels in young mice [107]. In addition, this approach increased T-cell responsiveness and facilitated the immune recovery of aged mice after chemotherapy and/or BMT [96, 109]. On the other hand, chemical SSA offers a transient and reversible effect by either targeting the upstream signaling events or by directly blocking steroid receptors [96, 110]. Therapeutic agents initially developed for prostate and uterine cancer such as the luteinizing hormone releasing hormone agonist or androgen receptor blocker increased both lymphoid and myeloid progenitors and accelerated thymic recovery after allogeneic BMT [102, 104]. Clinical studies on SSA have further shown increased repertoire diversity in the CD4 and CD8 T-cell populations [111]. Collectively, these preclinical data validated the idea that using SSA to enhance thymic function could lead to beneficial outcomes in both aged and immunocompromised patients [112].

Overall, SSA has a profound positive effect on the thymus. However, the long-term systemic effects of both surgical and chemical SSA are neither yet clear nor are the potential side effects. Therefore, more in depth investigations are required to assess the safety of SSA-based therapy. Moreover, the failure to restore thymic function by the natural decline in sex hormones levels in senile patients provides an evident that thymic involution is a complex process [21, 113]. Therefore, a more comprehensive approach involving different factors and mechanisms should be further investigated.

3.2. KGF: to guard the thymic epithelium

Also known as fibroblast growth factor 7, KGF is a member of the fibroblast growth factor family. It is mainly produced by mesenchymal cells [114, 115] and elects its activity through binding to its receptor fibroblast growth factor R2-IIIb (FgfR2-IIIb or KGFR) [116]. KGF has a notable effect on triggering the proliferation of epithelial cells [116]. Therefore, Palifermin®, a truncated form of KGF is clinically prescribed for patients undergoing stem cell replacement after high-dose chemotherapy and radiation [117]. Specially, Palifermin® is administered to reduce the incidence and duration of mucositis; an inflammation and ulceration of the mucous membranes lining the digestive tract [117].

In the thymus, both TECs and thymocytes produce KGF. However, only TECs express the KGFR, which explains the protective effect of KGF on thymic epithelium [116, 118]. Furthermore, studies conducted on KGFR-deficient mice support a major role for KGF in the thymus. Particularly, these deficient mice, which die at birth due to the absence of functional lungs, present with severely hypoplastic thymus [119]. In the same context, KGFR deficiency is found to be associated with significant defects in thymopoiesis accompanied by a decrease in thymic cellularity, in addition to signs of TEC proliferation blockade [119, 120]. Interestingly, the protective effect of KGF was evaluated in mouse models for allogeneic BMT where KGF pre-treatment was found to enhance the immune development post-BMT by improving the function of thymic microenvironment [118]. In the same study, Min et al. reported that KGF pre-treatment did improve thymopoiesis and positively affect the functional T-cells pool in the periphery [118].

As KGFR is expressed by several organs targeted by alloreactive T-cells, it was compelling to evaluate the effect of KGF in the setting of acute graft-versus-host disease (GVHD). In fact, current data have shown that KGF administration post-allogeneic BMT can: (i) facilitate alloengraftment [121], (ii) alleviate GVHD [122, 123, 124], (iii) protect epithelial cell in the gut mucosa while enhancing its repair [125], (iv) reduce the release of inflammatory cytokine [126], and (v) diminish allogeneic T-cell responses [121, 127]. A subsequent study by Berent-Maoz et al. further demonstrated that KGF not only protects TECs, but could also stimulate thymopoiesis indirectly by triggering them to secrete soluble factors [128]. Conversely, data from two separate phase I/II clinical trials conducted on patients undergoing allogeneic BMT revealed ameliorated mucositis with no significant improvements on the incidence and severity of acute GVHD, T-lymphopoiesis, infections, overall survival, or cancer relapse rates [129, 130]. As such, KGF can improve mucotoxicity following allogeneic BMT without exhibiting beneficial outcome on immune recovery in BMT patients.

3.3. GRL: increasing thymopoiesis appetite

The polypeptide hormone GRL, also named the hunger hormone, is normally released by specialized cells within the stomach into the circulation where it induces hunger and the release of growth hormone by stimulating the hypothalamus and the pituitary gland, respectively [131]. Moreover, GRL is known to play an important role in maintaining energy homeostasis [132, 133]. Interestingly, the GRL receptor (GRLR) is expressed by the pituitary gland, the central nervous system as well as on various immune cells including resting and activated T-cells [134]. Moreover, GRL deficiency in mice is associated with reduced thymopoiesis and increased thymic adiposity [135, 136]. Therefore, a potential role for GRL has been proposed in reversing age-related thymic involution [95]. In fact, scientists believe that GRL represents the next generation of hormonal-based rejuvenation therapy due to its astonishing effect on the aging thymus. Specifically, GRL administration to aged mice (14–22 months) resulted in improved TCR diversity, increased thymic cellularity (including cTECs, mTECs, and ETPs) and RTE output [134, 137]. In addition, Dixit et al. demonstrated that GRL administration inhibits adipogenesis and production of pro-inflammatory cytokines in the thymus; two important characteristics of thymic involution [134, 135, 138]. Despite these promising pleiotropic thymopoietic-stimulating effects, the progressive loss of GRLR expression with aging overshadows the potential use of GRL as a thymo-stimulatory therapy [134].

3.4. IL-7: a toolkit for thymus rejuvenation

Discovered in 1988 as a stimulator for murine B-cell progenitors, IL-7 is a member of the type-I-cytokine-family and signals through its heterodimer IL-7 receptor (IL-7R) [139]. Unlike other cytokines, IL-7 is mainly produced by nonhematopoietic cells [140]. Soon after its discovery, the profound and nonredundant effect of IL-7 on T-lymphopoiesis was underlined as it could: (i) enhance the expansion of naïve peripheral CD4 and CD8 SP T-cells [141], (ii) ameliorate the antiviral/antitumor activity of cytotoxic T-cells [142, 143], and (iii) support the survival/proliferation of CD8 memory T-cells [143, 144, 145]. These unique properties made IL-7 a central research topic in the context of viral infections, cancer therapies, and in T-cell reconstitution following BMT [140, 145]. However, the outstanding role of IL-7 as the cornerstone of T-lymphopoiesis in preclinical studies was challenged by contradicting observations made in higher species. For example, data from an autologous hematopoietic stem cell transplantation study in nonhuman primates [146] and from two IL-7 phase I clinical trials conducted on cancer patients failed to provide evidence for significant de novo thymopoiesis [147, 148]. This discrepancy with the results obtained from rodent models indicates the need to further elucidate the role of IL-7 in higher species. Nonetheless, IL-7 may still be clinically relevant as a potent adjuvant to stimulate T-cell effector functions in various illnesses.

3.5. IL21: a new thymostimulatory agent

IL-21 is the most recently identified member of the type-I-cytokine-family [149]. Produced mainly by activated CD4 T-cells [150], IL-21 was found to: (i) promote CD4 T-cell differentiation down the Th17 pathway [151], (ii) co-stimulate activated NK and CD8 lymphocytes [152], (iii) desensitize responding cells to the inhibitory effects of regulatory T-cells [150], and (iv) act as a switch for IgG production in B-cells [153]. In addition, similar to other type-I-cytokine-family members, IL-21 signals through its heterodimer IL-21 receptor (IL21-R), which is expressed by different hematopoietic cells such as, natural killers, B and T lymphocytes [154]. Moreover, we have recently showed that in vitro peptide-mediated TCR-engagement triggers potent cell surface expression of the IL-21R on DP thymocytes [18]. Although not required for hematopoiesis, BM progenitors expanded in response to IL-21 overexpression in vivo [155]. Likewise, IL-21 did not seem to be essential for thymopoiesis due to normal T-cell development in IL-21R−/− mice [149]. Moreover, IL-21 supplementation to positively selected DP thymocytes did not trigger their differentiation to CD8 SP T-cells as did IL-7 [156]. Instead, it led to DP thymocyte expansion and a 3–4-fold increase in the absolute number of in vitro differentiated CD8 T-cells when combined with other differentiation-inducing cytokines such as IL-4, IL-7, or IL-13 [157, 158]. These findings prompt further investigations using three pre-clinical settings with impaired thymopoiesis: (i) ensuing pharmacologically-induced thymic atrophy, (ii) in age-related thymic involution, and (iii) for T-cell reconstitution following allogeneic BMT.

So far, no treatments are available to protect against acute thymic atrophy or to accelerate thymic recovery under such circumstances, thereby leaving the immune system compromised. Of note, sepsis caused by bacterial infections can trigger GCs (corticosterone in rodents and cortisol in humans) secretion by the hypothalamus-pituitary adrenal axis. As a result, DP thymocytes undergo apoptosis due to their inability to express the anti-apoptotic molecule Bcl-2 [82]. Such acute thymosuppressive state can be easily replicated in mice via injection of the synthetic corticosteroid dexamethasone (DEX) [159]; hence, the utility of this model in evaluating the capacity of IL-21 in accelerating thymic recovery. One day following intraperitoneal (IP) injection of wild-type C57Bl/6 mice (4–6 months old) with PBS or DEX, IL-21R was found to be expressed on total or fractionated DN as well as CD4/CD8 SP thymocytes in both animal groups [18]. In contrast, DP thymocytes expressed the IL-21R exclusively following DEX injection [18]. To further ascertain these observations, a functional in vitro proliferation assay was conducted using sorted DN and DP subsets. Cell counts revealed that DN thymocytes derived from PBS or DEX-treated mice proliferated similarly in a dose-dependent manner in response to IL-21 [18]. In contrast, only positively selected (CD69+) or DEX-derived DP thymocytes expanded in response to IL-21 [18]. To directly assess its effect on thymopoiesis in vivo, DEX-treated mice were IP-injected with IL-21 versus equivalent volume of PBS. Thymi derived from the IL-21 group displayed accelerated size and cellularity recovery compared to control DEX/PBS-injected animals. Analysis of the percentages obtained by flow-cytometry revealed that IL-21 treatments enhance the recovery of DP thymocytes. The response to IL-21 administration also culminated in a noticeable remodeling of the thymic architecture [18]. Furthermore, extrapolation of absolute numbers using flow-cytometry percentages revealed a significant increase in the DN and DP populations within the IL-21 group with no observed effects on the TEC compartment. This is not surprising as TECs do not express the IL-21R. To confirm that IL-21-mediated effects are due to proliferation as opposed to survival of thymic progenitors, DN and DP thymocytes were analyzed for their expression of the anti-apoptotic molecules Bcl-2 (expressed only in DN thymocytes) and Bcl-XL (expressed normally in both DN and DP thymocytes) [18]. Interestingly, none of these anti-apoptotic proteins were up-regulated in both populations supporting the notion that IL-21 administration leads to thymocyte expansion as opposed to their enhanced survival. Pertinent to this project, IL-21 administration did not skew the TCR repertoire diversity as T-cells derived from PBS, DEX/PBS-, or DEX/IL-21-treated mice displayed comparable TCRVβ distribution within intrathymic and peripheral CD4/CD8 SP T-cell populations. This point is particularly important as it clearly demonstrates that IL-21 administration under acute thymic atrophy does not lead to non-specific mono- or oligoclonal T-cell proliferation. Finally, IL-21 signaling is characterized by the preferential activation of the JAK-STAT pathways [160]. Biochemical responsiveness of sorted thymic subsets in response to IL-21 stimulation in vitro clearly demonstrated phosphorylation of STAT1, STAT3, and STAT5 in all thymic subsets except for DP thymocytes; unless they were derived from DEX-treated mice. This was expected as nonpositively selected (CD69) or nonDEX-treated DP thymocytes do not express the IL-21R [18].

In contrast to acute thymic atrophy, chronic age-related involution is characterized by a gradual expansion of the PVS and reduction of the stromal compartment capable of supporting thymopoiesis [74]. To further explore the thymopoietic potential of IL-21 in such model, assessment of IL-21R expression with aging was conducted to discard the possibility that IL-21R expression declines progressively akin to the GRLR situation [134]. Analysis performed on total and fractionated thymocytes (DN, DP, and SPs) revealed that IL-21R expression profiles are comparable between young and aged mice (unpublished data). Next, wild type-aged C57Bl/6 mice (14–18 months old) received three IP injections of IL-21 prior to their analysis on the following day. This treatment led to increased thymic size, weight, and cellularity. Analysis of thymic subsets showed a drastic increase in the proportion of DN thymocytes as it reached 15% in contrast to 3% in PBS control mice. Likewise, a notable increase in the frequency of CD4 and CD8 SP T cells was also observed in the IL-21 group. This outcome was further reflected in the overall absolute number of DN, DP, and SP thymocytes. As one of the predominant deficiencies occurring with chronic thymic atrophy is decreased migration, proliferation, and survival of ETPs, we assessed the effect of IL-21 administration on the scarcity of this central progenitor population in the thymus. Flow-cytometry analysis of ETPs showed a significant increase in their frequencies within the IL-21 group suggesting preferential expansion of ETPs in response to IL-21 administration. As the aim of using IL-21 focuses on rejuvenating T-cell immunity of aged mice, T-cell output was analyzed weekly in PBS- vs. IL-21-treated aged mice over a total period of 3 weeks. Interestingly, aged mice undergoing IL-21 treatment showed distinctively improved T-cell responses [18]. In addition, while IL-21 administration had no impact on the absolute number of all peripheral lymphocytes, the frequency of peripheral GFP+ T-cells increased substantially in comparison with control mice [18]. This can also explain the improved anti-tumoral response observed in IL-21-pre-conditionned aged mice prior to cancer vaccination [18]. The sum of these results serves as the basis to investigate the use of IL-21 as an elderly pre-conditioning therapy to rejuvenate immunity, and thus, improve T-cell responsiveness to vaccination.

The above findings provided the impetus to investigate the use of IL-21 in T-cell reconstitution post-BMT. Although this medical procedure is adopted as a life-saving procedure for specific malignant and non-malignant conditions [161], it remains unfortunately associated with dangerous life-threatening complications. The high morbidity and mortality associated with BMT persist as a major clinical problem associated with increased risks of: (i) relapse or development of secondary malignancies [162, 163], (ii) infection [162, 164, 165], and (iii) reduced responsiveness to vaccination [166]. The primary factors associated with these complications are the significant delay and improper reconstitution of T-cells post-BMT [165, 167] mostly due to the significant damage inflicted to TECs [168, 169, 170, 171]. Therefore, the ability of IL-21 to improve de novo T-cell reconstitution post-BMT was investigated. For this purpose, T-cell-depleted RAG2p-GFP-derived BM cells (H2-Kb) were transplanted into irradiated LP/J (H2-Kb/c) recipient animals followed by IL-21 administration. In this experiment, GFP expression driven by the Rag2 promoter allows direct assessment of de novo peripheral T-cell reconstitution [172]. Analysis of transplanted mice revealed that IL-21: (i) accelerates lymphocyte reconstitution including T-cells (also observed in NOD scid gamma (NSG) mice receiving human IL-21 following cord-blood transplantation), (ii) slightly improved TECs recovery, (iii) regenerates a naïve peripheral T-cell pool with a diverse TCR repertoire, (iv) enhances regulatory B-cell development, and (v) protects from GHVD while retaining the graft-versus-tumor effect. Of note, IL-21 was originally believed to specifically affect the thymus [154]. In line with a previous study, however, BM analyses conducted on transplanted mice revealed increased counts of LinSca1+cKit+ (LSK) cells in IL-21-treated mice [17, 155]. This observation is particularly interesting as HSC/HSC progenitors capable of generating lineages of the hematopoietic system are enriched within the LSKs population [173].

Advertisement

4. Conclusion

So far, the use of IL-21 in the clinic remains limited to cancer immunotherapy (IL-21-based stimulation) or in the setting of autoimmune diseases (IL-21 inhibition). The latter clinical objective is particularly important as it raises major concerns related to the use of IL-21 in patients prone to develop autoimmune ailments. Therefore, additional studies are warranted to specifically address the clinical use of IL-21 in indications such as immunotherapies. Alternatively, all pre-clinical observations related to IL-21 strongly suggest that this cytokine could be exploited either as a monotherapy or in combination with other standards of care. This would ensure, at least in the context of thymopoiesis, improved de novo T-cell development in aged subjects as a mean to reverse thymic involution or post-BMT to accelerate the regeneration of naive T cells.

Advertisement

Acknowledgments

The work completed by the group of Dr. Rafei and summarized in this book chapter was supported by the Canadian Cancer Society (Innovation grant #2013-701623; the Louisa Gale Scholar), the Cole Foundation (Transition grant), the Cancer Research Society (Operating grant #18278 and Operating grant #21012) and by a generous donation from the Gagnon family. Jamilah Abusarah is a Vanier scholar. Moutih Rafei holds a Fonds de la Recherche en Santé du Québec Junior I and II Awards.

References

  1. 1. National Center for Health, S., Health, United States, in Health, United States, 2007: With Chartbook on Trends in the Health of Americans. 2007, National Center for Health Statistics (US): Hyattsville (MD)
  2. 2. Organization, W.H. Ageing and health. 2015 [cited 2017 09-22]; Available from: http://www.who.int/mediacentre/factsheets/fs404/en/
  3. 3. Denton FT, Spencer BG. Chronic health conditions: Changing prevalence in an aging population and some implications for the delivery of health care services. Canadian Journal on Aging. 2010;29(1):11-21
  4. 4. Effros RB. Role of T lymphocyte replicative senescence in vaccine efficacy. Vaccine. 2007;25(4):599-604
  5. 5. Peto J. Cancer epidemiology in the last century and the next decade. Nature. 2001;411(6835):390-395
  6. 6. Deleidi M, Jaggle M, Rubino G. Immune aging, dysmetabolism, and inflammation in neurological diseases. Frontiers in Neuroscience. 2015;9:172
  7. 7. Dorshkind K, Montecino-Rodriguez E, Signer RA. The ageing immune system: Is it ever too old to become young again? Nature Reviews. Immunology. 2009;9(1):57-62
  8. 8. Pera A et al. Immunosenescence: Implications for response to infection and vaccination in older people. Maturitas. 2015;82(1):50-55
  9. 9. Derhovanessian E et al. Immunity, ageing and cancer. Immunity & Ageing. 2008;5:11
  10. 10. Hakim FT, Gress RE. Immunosenescence: Deficits in adaptive immunity in the elderly. Tissue Antigens. 2007;70(3):179-189
  11. 11. Goronzy JJ et al. Naive T cell maintenance and function in human aging. Journal of Immunology. 2015;194(9):4073-4080
  12. 12. Larbi A et al. Impact of age on T cell signaling: A general defect or specific alterations? Ageing Research Reviews. 2011;10(3):370-378
  13. 13. Fulop T et al. Aging, immunity, and cancer. Discovery Medicine. 2011;11(61):537-550
  14. 14. Urruticoechea A et al. Recent advances in cancer therapy: An overview. Current Pharmaceutical Design. 2010;16(1):3-10
  15. 15. Nabel GJ. Designing tomorrow's vaccines. The New England Journal of Medicine. 2013;368(6):551-560
  16. 16. Gravekamp C. The importance of the age factor in cancer vaccination at older age. Cancer Immunology, Immunotherapy. 2009;58(12):1969-1977
  17. 17. Tormo A et al. Interleukin-21 promotes thymopoiesis recovery following hematopoietic stem cell transplantation. Journal of Hematology & Oncology. 2017;10(1):120
  18. 18. Al-Chami E et al. Interleukin-21 administration to aged mice rejuvenates their peripheral T-cell pool by triggering de novo thymopoiesis. Aging Cell. 2016;15(2):349-360
  19. 19. Al-Chami E et al. Therapeutic utility of the newly discovered properties of interleukin-21. Cytokine. 2016;82:33-37
  20. 20. Murphy K et al. Janeway's Immunobiology. 7th ed. New York: Garland Science; 2008. xxi, 887 p
  21. 21. Steinmann GG, Klaus B, MÜLler-Hermelink HK. The involution of the ageing human thymic epithelium is independent of puberty. Scandinavian Journal of Immunology. 1985;22(5):563-575
  22. 22. Blackburn CC, Manley NR. Developing a new paradigm for thymus organogenesis. Nature Reviews. Immunology. 2004;4(4):278-289
  23. 23. Haynes BF et al. The role of the thymus in immune reconstitution in aging, bone marrow transplantation, and HIV-1 infection. Annual Review of Immunology. 2000;18:529-560
  24. 24. Pearse G. Normal structure, function and histology of the thymus. Toxicologic Pathology. 2006;34(5):504-514
  25. 25. Griffith AV et al. Spatial mapping of thymic stromal microenvironments reveals unique features influencing T lymphoid differentiation. Immunity. 2009;31(6):999-1009
  26. 26. Takahama Y et al. Generation of diversity in thymic epithelial cells. Nature Reviews. Immunology. 2017;17(5):295-305
  27. 27. Takada K et al. Development and function of cortical thymic epithelial cells. Current Topics in Microbiology and Immunology. 2014;373:1-17
  28. 28. Ohigashi I, Kozai M, Takahama Y. Development and developmental potential of cortical thymic epithelial cells. Immunological Reviews. 2016;271(1):10-22
  29. 29. Ripen AM et al. Ontogeny of thymic cortical epithelial cells expressing the thymoproteasome subunit beta5t. European Journal of Immunology. 2011;41(5):1278-1287
  30. 30. Nakagawa T et al. Cathepsin L: Critical role in Ii degradation and CD4 T cell selection in the thymus. Science. 1998;280(5362):450-453
  31. 31. Gommeaux J et al. Thymus-specific serine protease regulates positive selection of a subset of CD4+ thymocytes. European Journal of Immunology. 2009;39(4):956-964
  32. 32. Meireles C et al. Thymic crosstalk restrains the pool of cortical thymic epithelial cells with progenitor properties. European Journal of Immunology. 2017;47(6):958-969
  33. 33. van Ewijk W et al. Stepwise development of thymic microenvironments in vivo is regulated by thymocyte subsets. Development. 2000;127(8):1583-1591
  34. 34. Alves NL et al. Thymic epithelial cells: The multi-tasking framework of the T cell “cradle”. Trends in Immunology. 2009;30(10):468-474
  35. 35. Schwarz BA, Bhandoola A. Trafficking from the bone marrow to the thymus: A prerequisite for thymopoiesis. Immunological Reviews. 2006;209:47-57
  36. 36. Lind EF et al. Mapping precursor movement through the postnatal thymus reveals specific microenvironments supporting defined stages of early lymphoid development. The Journal of Experimental Medicine. 2001;194(2):127-134
  37. 37. Takada K et al. Development and function of cortical Thymic epithelial cells. In: Boehm T, Takahama Y, editors. Thymic Development and Selection of T Lymphocytes. Berlin, Heidelberg: Springer Berlin Heidelberg; 2014. pp. 1-17
  38. 38. Anderson G et al. Mechanisms of thymus medulla development and function. In: Boehm T, Takahama Y, editors. Thymic Development and Selection of T Lymphocytes. Berlin, Heidelberg: Springer Berlin Heidelberg; 2014. pp. 19-47
  39. 39. Miyasaka M et al. Characterization of lymphatic and venous emigrants from the thymus. Thymus. 1990;16(1):29-43
  40. 40. Petrie HT. Cell migration and the control of post-natal T-cell lymphopoiesis in the thymus. Nature Reviews. Immunology. 2003;3(11):859-866
  41. 41. Toro I, Olah I. Penetration of thymocytes into the blood circulation. Journal of Ultrastructure Research. 1967;17(5):439-451
  42. 42. Miller JF. The discovery of thymus function and of thymus-derived lymphocytes. Immunological Reviews. 2002;185:7-14
  43. 43. Lavini C. The thymus from antiquity to the present day: The history of a mysterious gland. In: Lavini C et al, editors. Thymus Gland Pathology: Clinical, Diagnostic, and Therapeutic Features. Milano: Springer Milan; 2008. pp. 1-12
  44. 44. Solis-Cohen S. The therapeutic uses of the thymus gland. Journal of the American Medical Association. 1900;XXXV(7):421-424
  45. 45. Rosenow EC 3rd, Hurley BT. Disorders of the thymus. A review. Archives of Internal Medicine. 1984;144(4):763–770
  46. 46. Wilson DS. Status thymicolymphaticus; presentation and discussion of a case. Current Researches in Anesthesia & Analgesia. 1950;29(6):356-358
  47. 47. Jacobs MT, Frush DP, Donnelly LF. The right place at the wrong time: Historical perspective of the relation of the thymus gland and pediatric radiology. Radiology. 1999;210(1):11-16
  48. 48. RM S. Poverty?s remains. The Sciences (NY Acad Sci). 1990;31(5):8-10
  49. 49. Chandra RK. Nutritional deficiency and susceptibility to infection. Bulletin of the World Health Organization. 1979;57(2):167-177
  50. 50. Ritterman JB. To Err is Human: Can American Medicine Learn from Past Mistakes? The Permanente Journal. 2017;21:16-181
  51. 51. Duffy BJ Jr, Fitzgerald PJ. Thyroid cancer in childhood and adolescence; a report on 28 cases. Cancer. 1950;3(6):1018-1032
  52. 52. Hildreth NG, Shore RE, Dvoretsky PM. The risk of breast cancer after irradiation of the thymus in infancy. The New England Journal of Medicine. 1989;321(19):1281-1284
  53. 53. Elmore SA. Enhanced histopathology of the thymus. Toxicologic Pathology. 2006;34(5):656-665
  54. 54. Pearse G. Histopathology of the thymus. Toxicologic Pathology. 2006;34(5):515-547
  55. 55. Gruver AL, Sempowski GD. Cytokines, leptin, and stress-induced thymic atrophy. Journal of Leukocyte Biology. 2008;84(4):915-923
  56. 56. Choyke PL et al. Thymic atrophy and regrowth in response to chemotherapy: CT evaluation. AJR. American Journal of Roentgenology. 1987;149(2):269-272
  57. 57. Clarke AG, Kendall MD. The thymus in pregnancy: The interplay of neural, endocrine and immune influences. Immunology Today. 1994;15(11):545-551
  58. 58. Lynch HE et al. Thymic involution and immune reconstitution. Trends in Immunology. 2009;30(7):366-373
  59. 59. de Meis J et al. Thymus atrophy and double-positive escape are common features in infectious diseases. Journal of Parasitology Research. 2012;2012:574020
  60. 60. Billard M, Gruver A, Sempowski G. Acute stress-induced thymic atrophy is mediated by intrathymic inflammatory and wound healing responses (113.25). The Journal of Immunology. 2011;186(1 Supplement):113.25-113.25
  61. 61. Tuckermann JP et al. Molecular mechanisms of glucocorticoids in the control of inflammation and lymphocyte apoptosis. Critical Reviews in Clinical Laboratory Sciences. 2005;42(1):71-104
  62. 62. Distelhorst CW. Recent insights into the mechanism of glucocorticosteroid-induced apoptosis. Cell Death and Differentiation. 2002;9(1):6-19
  63. 63. Perry GA, Jackson JD, Talmadge JE. Effects of a multidrug chemotherapy regimen on the thymus. Thymus. 1994;23(1):39-51
  64. 64. Hakim FT et al. Constraints on CD4 recovery postchemotherapy in adults: Thymic insufficiency and apoptotic decline of expanded peripheral CD4 cells. Blood. 1997;90(9):3789-3798
  65. 65. Randle-Barrett ES, Boyd RL. Thymic microenvironment and lymphoid responses to sublethal irradiation. Developmental Immunology. 1995;4(2):101-116
  66. 66. Daley JP et al. Retarded recovery of functional T cell frequencies in T cell-depleted bone marrow transplant recipients. Blood. 1987;70(4):960-964
  67. 67. Hale LP. Histologic and molecular assessment of human thymus. Annals of Diagnostic Pathology. 2004;8(1):50-60
  68. 68. Boehm T, Swann JB. Thymus involution and regeneration: Two sides of the same coin? Nature Reviews. Immunology. 2013;13(11):831-838
  69. 69. Salam N et al. T cell ageing: Effects of age on development, survival & function. The Indian Journal of Medical Research. 2013;138(5):595-608
  70. 70. Tanchot C, Rocha B. The peripheral T cell repertoire: Independent homeostatic regulation of virgin and activated CD8+ T cell pools. European Journal of Immunology. 1995;25(8):2127-2136
  71. 71. Taub DD, Longo DL. Insights into thymic aging and regeneration. Immunological Reviews. 2005;205:72-93
  72. 72. Gravekamp C, Chandra D. Aging and cancer vaccines. Critical Reviews in Oncogenesis. 2013;18(6):585-595
  73. 73. Sempowski GD et al. Leukemia inhibitory factor, oncostatin M, IL-6, and stem cell factor mRNA expression in human thymus increases with age and is associated with thymic atrophy. Journal of Immunology. 2000;164(4):2180-2187
  74. 74. Valiathan R, Ashman M, Asthana D. Effects of ageing on the immune system: Infants to elderly. Scandinavian Journal of Immunology. 2016;83(4):255-266
  75. 75. Stacy S et al. Immunological memory and late onset autoimmunity. Mechanisms of Ageing and Development. 2002;123(8):975-985
  76. 76. Jones SC et al. Impact of post-thymic cellular longevity on the development of age-associated CD4+ T cell defects. Journal of Immunology. 2008;180(7):4465-4475
  77. 77. Eaton SM et al. Age-related defects in CD4 T cell cognate helper function lead to reductions in humoral responses. The Journal of Experimental Medicine. 2004;200(12):1613-1622
  78. 78. Kay MM. Immunological aspects of aging: Early changes in thymic activity. Mechanisms of Ageing and Development. 1984;28(2–3):193-218
  79. 79. Haynes L, Swain SL. Why aging T cells fail: Implications for vaccination. Immunity. 2006;24(6):663-666
  80. 80. Ansar Ahmed S, Penhale WJ, Talal N. Sex hormones, immune responses, and autoimmune diseases. Mechanisms of sex hormone action. The American Journal of Pathology. 1985;121(3):531-551
  81. 81. Ansar Ahmed S, Dauphinee MJ, Talal N. Effects of short-term administration of sex hormones on normal and autoimmune mice. Journal of Immunology. 1985;134(1):204-210
  82. 82. Herold MJ, McPherson KG, Reichardt HM. Glucocorticoids in T cell apoptosis and function. Cellular and Molecular Life Sciences. 2006;63(1):60-72
  83. 83. Fitzpatrick FT et al. Reappearance of thymus of ageing rats after orchidectomy. The Journal of Endocrinology. 1985;106(3):R17-R19
  84. 84. Min H, Montecino-Rodriguez E, Dorshkind K. Reduction in the developmental potential of intrathymic T cell progenitors with age. Journal of Immunology. 2004;173(1):245-250
  85. 85. Sharp A, Kukulansky T, Globerson A. In vitro analysis of age-related changes in the developmental potential of bone marrow thymocyte progenitors. European Journal of Immunology. 1990;20(12):2541-2546
  86. 86. Eren R et al. Age-related changes in the capacity of bone marrow cells to differentiate in thymic organ cultures. Cellular Immunology. 1988;112(2):449-455
  87. 87. Rossi DJ et al. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(26):9194-9199
  88. 88. Linton PJ, Dorshkind K. Age-related changes in lymphocyte development and function. Nature Immunology. 2004;5(2):133-139
  89. 89. Li L et al. Cellular mechanism of thymic involution. Scandinavian Journal of Immunology. 2003;57(5):410-422
  90. 90. Takahama Y. Journey through the thymus: Stromal guides for T-cell development and selection. Nature Reviews. Immunology. 2006;6(2):127-135
  91. 91. Aw D, Silva AB, Palmer DB. Is thymocyte development functional in the aged? Aging (Albany NY). 2009;1(2):146-153
  92. 92. Hale JS et al. Thymic output in aged mice. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(22):8447-8452
  93. 93. Henson SM, Pido-Lopez J, Aspinall R. Reversal of thymic atrophy. Experimental Gerontology. 2004;39(4):673-678
  94. 94. Ventevogel MS, Sempowski GD. Thymic rejuvenation and aging. Current Opinion in Immunology. 2013;25(4):516-522
  95. 95. Taub DD, Murphy WJ, Longo DL. Rejuvenation of the aging thymus: Growth hormone-mediated and ghrelin-mediated signaling pathways. Current Opinion in Pharmacology. 2010;10(4):408-424
  96. 96. Velardi E, Dudakov JA, van den Brink MR. Sex steroid ablation: An immunoregenerative strategy for immunocompromised patients. Bone Marrow Transplantation. 2015;50(Suppl 2):S77-S81
  97. 97. Evans RM. The steroid and thyroid hormone receptor superfamily. Science. 1988;240(4854):889-895
  98. 98. Marcinkowska E, Wiedlocha A. Steroid signal transduction activated at the cell membrane: From plants to animals. Acta Biochimica Polonica. 2002;49(3):735-745
  99. 99. Luster MI et al. Estrogen immunosuppression is regulated through estrogenic responses in the thymus. Journal of Immunology. 1984;133(1):110-116
  100. 100. Zoller AL, Kersh GJ. Estrogen induces thymic atrophy by eliminating early thymic progenitors and inhibiting proliferation of beta-selected thymocytes. Journal of Immunology. 2006;176(12):7371-7378
  101. 101. Olsen NJ et al. Androgen receptors in thymic epithelium modulate thymus size and thymocyte development. Endocrinology. 2001;142(3):1278-1283
  102. 102. Lai KP et al. Targeting thymic epithelia AR enhances T-cell reconstitution and bone marrow transplant grafting efficacy. Molecular Endocrinology. 2013;27(1):25-37
  103. 103. Olsen NJ, Kovacs WJ. Effects of androgens on T and B lymphocyte development. Immunologic Research. 2001;23(2–3):281-288
  104. 104. Kompella UB et al. Luteinizing hormone-releasing hormone agonist and transferrin functionalizations enhance nanoparticle delivery in a novel bovine ex vivo eye model. Molecular Vision. 2006;12:1185-1198
  105. 105. Goodall A. The post-natal changes in the thymus of guinea-pigs, and the effect of castration on thymus structure. The Journal of Physiology. 1905;32(2):191-198
  106. 106. Henderson J. On the relationship of the thymus to the sexual organs: I. The influence of castration on the thymus. The Journal of Physiology. 1904;31(3–4):222-229
  107. 107. Heng TS et al. Effects of castration on thymocyte development in two different models of thymic involution. Journal of Immunology. 2005;175(5):2982-2993
  108. 108. Heng TS et al. Impact of sex steroid ablation on viral, tumour and vaccine responses in aged mice. PLoS One. 2012;7(8):e42677
  109. 109. Goldberg GL et al. Enhanced immune reconstitution by sex steroid ablation following allogeneic hemopoietic stem cell transplantation. Journal of Immunology. 2007;178(11):7473-7484
  110. 110. Greenstein BD et al. Regeneration of the thymus in old male rats treated with a stable analogue of LHRH. The Journal of Endocrinology. 1987;112(3):345-350
  111. 111. Sutherland JS et al. Activation of thymic regeneration in mice and humans following androgen blockade. Journal of Immunology. 2005;175(4):2741-2753
  112. 112. van Poppel H, Nilsson S. Testosterone surge: Rationale for gonadotropin-releasing hormone blockers? Urology. 2008;71(6):1001-1006
  113. 113. Griffith AV et al. Persistent degenerative changes in thymic organ function revealed by an inducible model of organ regrowth. Aging Cell. 2012;11(1):169-177
  114. 114. Rubin JS et al. Keratinocyte growth factor as a cytokine that mediates mesenchymal-epithelial interaction. EXS. 1995;74:191-214
  115. 115. Rubin JS et al. Purification and characterization of a newly identified growth factor specific for epithelial cells. Proceedings of the National Academy of Sciences of the United States of America. 1989;86(3):802-806
  116. 116. Finch PW, Rubin JS. Keratinocyte growth factor expression and activity in cancer: Implications for use in patients with solid tumors. Journal of the National Cancer Institute. 2006;98(12):812-824
  117. 117. Spielberger R et al. Palifermin for oral mucositis after intensive therapy for hematologic cancers. The New England Journal of Medicine. 2004;351(25):2590-2598
  118. 118. Min D et al. Protection from thymic epithelial cell injury by keratinocyte growth factor: A new approach to improve thymic and peripheral T-cell reconstitution after bone marrow transplantation. Blood. 2002;99(12):4592-4600
  119. 119. Revest JM et al. Development of the thymus requires signaling through the fibroblast growth factor receptor R2-IIIb. Journal of Immunology. 2001;167(4):1954-1961
  120. 120. Zhang X et al. Receptor specificity of the fibroblast growth factor family. The complete mammalian FGF family. Journal of Biological Chemistry. 2006;281(23):15694-15700
  121. 121. Bruinsma M et al. Keratinocyte growth factor improves allogeneic bone marrow engraftment through a CD4+Foxp3+ regulatory T cell-dependent mechanism. Journal of Immunology. 2009;182(12):7364-7369
  122. 122. Rossi S et al. Keratinocyte growth factor preserves normal thymopoiesis and thymic microenvironment during experimental graft-versus-host disease. Blood. 2002;100(2):682-691
  123. 123. Panoskaltsis-Mortari A et al. Keratinocyte growth factor administered before conditioning ameliorates graft-versus-host disease after allogeneic bone marrow transplantation in mice. Blood. 1998;92(10):3960-3967
  124. 124. Krijanovski OI et al. Keratinocyte growth factor separates graft-versus-leukemia effects from graft-versus-host disease. Blood. 1999;94(2):825-831
  125. 125. Vanclee A et al. Keratinocyte growth factor ameliorates acute graft-versus-host disease in a novel nonmyeloablative haploidentical transplantation model. Bone Marrow Transplantation. 2005;36(10):907-915
  126. 126. Hill GR, Ferrara JL. The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: Rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood. 2000;95(9):2754-2759
  127. 127. Panoskaltsis-Mortari A et al. Keratinocyte growth factor facilitates alloengraftment and ameliorates graft-versus-host disease in mice by a mechanism independent of repair of conditioning-induced tissue injury. Blood. 2000;96(13):4350-4356
  128. 128. Berent-Maoz B et al. Fibroblast growth factor-7 partially reverses murine thymocyte progenitor aging by repression of Ink4a. Blood. 2012;119(24):5715-5721
  129. 129. Filicko J, Lazarus HM, Flomenberg N. Mucosal injury in patients undergoing hematopoietic progenitor cell transplantation: New approaches to prophylaxis and treatment. Bone Marrow Transplantation. 2003;31(1):1-10
  130. 130. Freytes CO et al. Phase I/II randomized trial evaluating the safety and clinical effects of repifermin administered to reduce mucositis in patients undergoing autologous hematopoietic stem cell transplantation. Clinical Cancer Research. 2004;10(24):8318-8324
  131. 131. Inui A et al. Ghrelin, appetite, and gastric motility: The emerging role of the stomach as an endocrine organ. The FASEB Journal. 2004;18(3):439-456
  132. 132. Cheng KC et al. The role of ghrelin in energy homeostasis and its potential clinical relevance (review). International Journal of Molecular Medicine. 2010;26(6):771-778
  133. 133. De Vriese C, Delporte C. Influence of ghrelin on food intake and energy homeostasis. Current Opinion in Clinical Nutrition and Metabolic Care. 2007;10(5):615-619
  134. 134. Dixit VD et al. Ghrelin promotes thymopoiesis during aging. The Journal of Clinical Investigation. 2007;117(10):2778-2790
  135. 135. Youm YH et al. Deficient ghrelin receptor-mediated signaling compromises thymic stromal cell microenvironment by accelerating thymic adiposity. The Journal of Biological Chemistry. 2009;284(11):7068-7077
  136. 136. Sun Y et al. Ghrelin stimulation of growth hormone release and appetite is mediated through the growth hormone secretagogue receptor. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(13):4679-4684
  137. 137. Dixit VD, Taub DD. Ghrelin and immunity: A young player in an old field. Experimental Gerontology. 2005;40(11):900-910
  138. 138. Dixit VD et al. Ghrelin inhibits leptin- and activation-induced proinflammatory cytokine expression by human monocytes and T cells. The Journal of Clinical Investigation. 2004;114(1):57-66
  139. 139. Namen AE et al. Stimulation of B-cell progenitors by cloned murine interleukin-7. Nature. 1988;333(6173):571-573
  140. 140. Fry TJ, Mackall CL. Interleukin-7: From bench to clinic. Blood. 2002;99(11):3892-3904
  141. 141. Seddon B, Zamoyska R. TCR and IL-7 receptor signals can operate independently or synergize to promote lymphopenia-induced expansion of naive T cells. Journal of Immunology. 2002;169(7):3752-3759
  142. 142. Wiryana P et al. Augmentation of cell-mediated immunotherapy against herpes simplex virus by interleukins: Comparison of in vivo effects of IL-2 and IL-7 on adoptively transferred T cells. Vaccine. 1997;15(5):561-563
  143. 143. Lynch DH, Namen AE, Miller RE. In vivo evaluation of the effects of interleukins 2, 4 and 7 on enhancing the immunotherapeutic efficacy of anti-tumor cytotoxic T lymphocytes. European Journal of Immunology. 1991;21(12):2977-2985
  144. 144. Khaled AR, Durum SK. Lymphocide: Cytokines and the control of lymphoid homeostasis. Nature Reviews. Immunology. 2002;2(11):817-830
  145. 145. Mackall CL, Fry TJ, Gress RE. Harnessing the biology of IL-7 for therapeutic application. Nature Reviews. Immunology. 2011;11(5):330-342
  146. 146. Lu H et al. Interleukin-7 improves reconstitution of antiviral CD4 T cells. Clinical Immunology. 2005;114(1):30-41
  147. 147. Rosenberg SA et al. IL-7 administration to humans leads to expansion of CD8+ and CD4+ cells but a relative decrease of CD4+ T-regulatory cells. Journal of Immunotherapy. 2006;29(3):313-319
  148. 148. Sportes C et al. Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets. The Journal of Experimental Medicine. 2008;205(7):1701-1714
  149. 149. Spolski R, Leonard WJ. Interleukin-21: A double-edged sword with therapeutic potential. Nature Reviews. Drug Discovery. 2014;13(5):379-395
  150. 150. Spolski R, Leonard WJ. Interleukin-21: Basic biology and implications for cancer and autoimmunity. Annual Review of Immunology. 2008;26:57-79
  151. 151. Zhou L et al. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nature Immunology. 2007;8(9):967-974
  152. 152. Li Y, Bleakley M, Yee C. IL-21 influences the frequency, phenotype, and affinity of the antigen-specific CD8 T cell response. Journal of Immunology. 2005;175(4):2261-2269
  153. 153. Ozaki K et al. A critical role for IL-21 in regulating immunoglobulin production. Science. 2002;298(5598):1630-1634
  154. 154. Parrish-Novak J et al. Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function. Nature. 2000;408(6808):57-63
  155. 155. Ozaki K et al. Overexpression of interleukin 21 induces expansion of hematopoietic progenitor cells. International Journal of Hematology. 2006;84(3):224-230
  156. 156. Zeng R et al. Synergy of IL-21 and IL-15 in regulating CD8+ T cell expansion and function. The Journal of Experimental Medicine. 2005;201(1):139-148
  157. 157. Rafei M et al. Differential effects of gammac cytokines on postselection differentiation of CD8 thymocytes. Blood. 2013;121(1):107-117
  158. 158. Rafei M et al. Development of a novel method for in vitro analysis of γc-cytokine effects on CD8 T cells positive selection (111.15). The Journal of Immunology. 2012;188(1 Supplement):111.15-111.15
  159. 159. Boersma W, Betel I, van der Westen G. Thymic regeneration after dexamethasone treatment as a model for subpopulation development. European Journal of Immunology. 1979;9(1):45-52
  160. 160. Leonard WJ, Wan C-K. IL-21 Signaling in Immunity. F1000Research, 2016;5:p. F1000 Faculty Rev-224
  161. 161. Sureda A et al. Indications for allo- and auto-SCT for haematological diseases, solid tumours and immune disorders: Current practice in Europe, 2015. Bone Marrow Transplantation. 2015;50(8):1037-1056
  162. 162. Auletta JJ, Lazarus HM. Immune restoration following hematopoietic stem cell transplantation: An evolving target. Bone Marrow Transplantation. 2005;35(9):835-857
  163. 163. Drobyski WR. Evolving strategies to address adverse transplant outcomes associated with T cell depletion. Journal of Hematotherapy & Stem Cell Research. 2000;9(3):327-337
  164. 164. Baddley JW et al. Invasive mold infections in allogeneic bone marrow transplant recipients. Clinical Infectious Diseases. 2001;32(9):1319-1324
  165. 165. Marr KA. Delayed opportunistic infections in hematopoietic stem cell transplantation patients: A surmountable challenge. Hematology. American Society of Hematology. Education Program. 2012;2012:265-270
  166. 166. Avigan D, Pirofski LA, Lazarus HM. Vaccination against infectious disease following hematopoietic stem cell transplantation. Biology of Blood and Marrow Transplantation. 2001;7(3):171-183
  167. 167. Takatsuka H et al. Complications after bone marrow transplantation are manifestations of systemic inflammatory response syndrome. Bone Marrow Transplantation. 2000;26(4):419-426
  168. 168. Williams KM, Gress RE. Immune reconstitution and implications for immunotherapy following haematopoietic stem cell transplantation. Best Practice & Research. Clinical Haematology. 2008;21(3):579-596
  169. 169. Chung B et al. Radiosensitivity of thymic interleukin-7 production and thymopoiesis after bone marrow transplantation. Blood. 2001;98(5):1601-1606
  170. 170. Weinberg K et al. Factors affecting thymic function after allogeneic hematopoietic stem cell transplantation. Blood. 2001;97(5):1458-1466
  171. 171. Banfi A et al. Bone marrow stromal damage after chemo/radiotherapy: Occurrence, consequences and possibilities of treatment. Leukemia & Lymphoma. 2001;42(5):863-870
  172. 172. Monroe RJ et al. RAG2:GFP knockin mice reveal novel aspects of RAG2 expression in primary and peripheral lymphoid tissues. Immunity. 1999;11(2):201-212
  173. 173. Okada S et al. Enrichment and characterization of murine hematopoietic stem cells that express c-kit molecule. Blood. 1991;78(7):1706-1712

Written By

Jamilah Abusarah, Fatemeh Khodayarian, Yun Cui, Abed El-Hakim El-Kadiry and Moutih Rafei

Submitted: 09 October 2017 Reviewed: 15 January 2018 Published: 04 July 2018