Hostname: page-component-7c8c6479df-995ml Total loading time: 0 Render date: 2024-03-28T01:05:31.744Z Has data issue: false hasContentIssue false

Nutritional imbalances and infections affect the thymus: consequences on T-cell-mediated immune responses

Published online by Cambridge University Press:  22 September 2010

Wilson Savino*
Affiliation:
Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
Mireille Dardenne
Affiliation:
Hôpital Necker, Université Paris Descartes, CNRS UMR-8147, Paris, France
*
*Corresponding author: Wilson Savino, fax 55 21 38658101, email savino@fiocruz.br; w_savino@hotmail.com
Rights & Permissions [Opens in a new window]

Abstract

The thymus gland, where T lymphocyte development occurs, is targeted in malnutrition secondary to protein energy deficiency. There is a severe thymic atrophy, resulting from massive thymocyte apoptosis (particularly affecting the immature CD4+CD8+ cell subset) and decrease in cell proliferation. The thymic microenvironment (the non-lymphoid compartment that drives intrathymic T-cell development) is also affected in malnutrition: morphological changes in thymic epithelial cells were found, together with a decrease of thymic hormone production, as well as an increase of intrathymic contents of extracellular proteins. Profound changes in the thymus can also be seen in deficiencies of vitamins and trace elements. Taking Zn deficiency as an example, there is a substantial thymic atrophy. Importantly, marginal Zn deficiency in AIDS subjects, children with diarrhoea and elderly persons, significantly impairs the host's immunity, resulting in an increased risk of opportunistic infections and mortality; effects that are reversed by Zn supplementation. Thymic changes also occur in acute infectious diseases, including a severe thymic atrophy, mainly due to the depletion of CD4+CD8+ thymocytes, decrease in thymocyte proliferation, in parallel to densification of the epithelial network and increase in the extracellular matrix contents, with consequent disturbances in thymocyte migration and export. In conclusion, the thymus is targeted in several conditions of malnutrition as well as in acute infections. These changes are related to the impaired peripheral immune response seen in malnourished and infected individuals. Thus, strategies inducing thymus replenishment should be considered as adjuvant therapeutics to improve immunity in malnutrition and/or acute infectious diseases.

Type
3rd International Immunonutrition Workshop
Copyright
Copyright © The Authors 2010

Abbreviations:
ECM

extracellular matrix

TCR

T-cell receptor

TEC

thymic epithelial cells

It has been a long time since scientists noticed that in the context of the malnutrition-related immunodeficiency, the thymus undergoes a variety of alterations, comprising, among others, a consistent severe atrophy (reviewed in(Reference Chandra1)), leading to the notion that the thymus can be considered as a barometer of malnutrition(Reference Prentice2). Interestingly, such a thymic atrophy pattern can also be found in a variety of infectious diseases(Reference Savino3). Importantly, these two pathological situations likely cause profound alterations in the host's immune system, in part as a consequence of targeting the thymus. Thus, the impact of malnutrition plus infection is a relevant issue in health sciences including public health, since in many countries malnutrition frequently parallels infections. Herein, we will review the similarities concerning the changes seen in the thymus of individuals suffering from malnutrition and/or infectious diseases. Yet, before discussing these specific data, it seemed useful to provide a general background of the normal thymus structure and function, comprising both the thymic microenvironment and the process of intrathymic T-cell differentiation.

Fig. 1. Normal intrathymic T-cell differentiation and the thymic microenvironment. In the left panel, we show a simplified view of normal thymocyte differentiation. Bone marrow-derived precursors enter the thymus and migrate from the cortico-medullary junction to the subcapsular cortical region of the thymic lobules. These immature cells do not express the CD3/T-cell receptor (TCR) complex, neither CD4 or CD8 molecules, being referred as CD4CD8 double negative. As shown in the right panel, as developing thymocytes progress in differentiation, they interact with microenvironmental cells, such as cortical thymic epithelial cells and fibroblasts localized in the cortex. At this stage, thymocytes start to express the TCR/CD3 complex and the molecules CD4 and CD8, thus becoming TCRlow/CD3lowCD4+CD8+ double-positive for these molecules. These cells are submitted to the processes of positive and negative selection, as a consequence of the interaction with the thymic microenvironmental cells through MHC/peptide–TCR interactions. Cortical epithelial cells are involved in positive selection, whereas both dendritic cells and epithelial cells can drive negation selection. Negatively selected cells die by apoptosis (most of them being phagocytized by macrophages) and positively selected thymocytes progress in differentiation, migrating through the medulla, ultimately becoming mature TCRhigh/CD3highCD4+CD8 or TCRhigh/CD3highCD8+CD4 single-positive T lymphocytes, which are the cells that normally leave the organ. Based on Savino and Dardenne(Reference Savino and Dardenne66).

The thymic microenvironment and its role in T-cell differentiation

The thymus is a primary lymphoid organ, in which bone marrow-derived T-cell precursors undergo differentiation, ultimately leading to the migration of positively selected thymocytes to the T-cell-dependent areas of peripheral lymphoid organs (see Fig. 1). Such a process involves a sequential expression of various proteins and rearrangements of the T-cell receptor (TCR) genes. Most immature thymocytes express neither the TCR complex nor the CD4 or CD8 accessory molecules; being called double-negative thymocytes, and representing 5% total thymocytes. As maturation progresses thymocytes acquire the membrane expression of the CD4 and CD8 markers, generating the CD4+CD8+ double-positive cells, which comprise 80% of the whole population. In this stage, TCR genes are rearranged, and productive rearrangements yield the membrane expression of the TCR (complexed with the CD3) in low densities (TCRlow). Thymocytes that do not undergo a productive TCR gene rearrangement die by apoptosis, whereas those expressing productive TCR interact with peptides presented by molecules of the MHC, expressed on microenvironmental cells. This interaction determines the positive and negative selection events, crucial for normal thymocyte differentiation. Negative selection results in apoptosis-mediated cell death. Positively selected thymocytes progress to the mature TCRhighCD4+CD8 or TCRhighCD4CD8+ single positive stage, comprising 15% thymocytes that ultimately leave the organ to form the large majority of the peripheral T-cell repertoire (reviewed in(Reference Ciofani and Zúñiga-Pflücker4)).

Thymocyte differentiation occurs as cells migrate within the thymic lobules: TCRCD4CD8 and TCR+CD4+CD8+ cells are cortically located, whereas mature TCR+CD4+CD8 and TCR+CD4CD8+ thymocytes are found in the medulla. Along with this journey, thymocytes interact with various components of the thymic microenvironment, a three-dimensional network formed of thymic epithelial cells (TEC), macrophages, dendritic cells, fibroblasts and extracellular matrix (ECM) components. In addition to the key interaction, involving the TCR/peptide-MHC, in the context of CD8 or CD4 molecules, the thymic microenvironment influences thymocyte maturation via adhesion molecules and ECM; interactions that are relevant to thymocyte migration(Reference Savino, Mendes da Cruz and Silva5, Reference Savino, Mendes-da-Cruz and Smaniotto6). Moreover, microenvironmental cells modulate thymocyte differentiation by soluble polypeptides, comprising: (a) cytokines, such as IL-1, IL-3, IL-6, IL-7, IL-8 and stem cell factor; (b) chemokines and (c) thymic hormones, including thymulin, thymopoietin and thymosin-α1(Reference Savino, Mendes da Cruz and Silva5Reference Petrie and Züniga-Pflucker8).

Thymocyte development is altered in protein–energy malnutrition

As stated earlier, one of the most conspicuous changes in malnutrition is thymic atrophy, which is essentially due to a massive thymocyte death, particularly affecting the immature stage of CD4+CD8+ cells(Reference Chandra1). Yet, it should be pointed out that, in addition to the increase in thymocyte death seen in thymuses from malnourished individuals, there is an intrathymic decrease in cell proliferation, as ascertained by the very low numbers of cells labelled with proliferating cell nuclear antigen, a marker for cell proliferation(Reference Mitsumori, Takegawa and Shimo9). Thus, protein–energy malnutrition-related thymocyte depletion results from enhanced thymocyte death plus decreased thymocyte proliferation. It should be pointed out that the major change in the thymic lymphoid compartment is also observed in human subjects suffering from malnutrition: a severe thymic atrophy with cortical thymocyte depletion is a consistent finding in necropsies of malnourished subjects(Reference Chandra1, Reference Lyra, Madi and Maeda10). Indeed, by means of echography, atrophy of the organ was also observed in vivo in malnourished children, as compared to age-matched healthy individuals(Reference Parent, Chevalier and Zalles11). Importantly, a study conducted in Guinea-Bissau showed that the reduction in thymus size at birth correlated with infant mortality(Reference Aaby, Marx and Trautner12). Fortunately, malnutrition-induced thymic atrophy seems to be reversible if appropriate diet is provided, as it has been demonstrated in a longitudinal study carried out with Bolivian severely malnourished children who were treated for nutrition rehabilitation, and that has been accompanied by ultrasound scanning. In these malnourished children, there was a severe reduction in the thymus volume, paralleled by abnormally high proportions of circulating immature T lymphocytes and lower proportions of mature T cells. Two months after beginning the diet rehabilitation, the thymic area of these children was recovered(Reference Chevalier, Sevilla and Zalles13).

Thymocyte depletion associated with deficiencies in vitamins and trace elements: the zinc-deficiency paradigm

In addition to protein-related malnutrition, trace element as well as vitamin deficiencies result in thymic atrophy, with cortical thymocyte depletion(Reference Kuvibidila, Dardenne and Savino14Reference Nodera, Yanagisawa and Wada17). In Fe-deficient mice, a decrease in mitogen-induced proliferative response of thymocytes has been demonstrated(Reference Kuvibidila, Dardenne and Savino14).

Many of the effects of protein–energy malnutrition on the immune function may be a result of associated alterations in metabolism of metals or vitamins, which in turn affect directly the immune system(Reference Cunningham-Rundles, McNeeley and Moon18). The abnormal incidence of various infections and the existence of lymphopenia and lymphoid organ atrophy in malnourished children have been repeatedly demonstrated and evidence points to Zn insufficiency in this type of immunodeficiency(Reference Shankar and Prasad19).

Zn plays a major role in cell division, differentiation, apoptosis and gene transcription, and strongly influences the immune system, affecting primarily T cells(Reference Chesters, Odel and Sunde20). The studies of the effects of severe Zn deficiency in experimental animals and human subjects showed substantial thymic atrophy as well as accelerated lymphopenia, leading to the reduction in cell- and antibody-mediated responses, thus influencing the susceptibility to infectious diseases(Reference Fraker, King, Garvy and Klurfeld21Reference Fraker, King and Laakko23).

Early observations showed that mice maintained on a Zn-deficient diet develop a progressive thymic involution: after 4 weeks the thymus retains only 25% of its original size and at 6 weeks only a few thymocytes remain in the organ(Reference Fernandes, Nair and Once24). Such changes are observed mostly in the thymic cortex, with a severe loss of CD4+CD8+ thymocytes, and can be reversed by Zn supplementation(Reference Fraker, Depascale-Jardieu and Zwickl25, Reference King, Osati-Ashtiani and Fraker26). Moreover, marginal Zn deficiency, in the early post-natal period, also results in substantial reduction in thymic size(Reference Beach, Gershwin and Hurley27).

The mechanism(s) of heightened apoptosis in Zn deficiency mice remain(s) to be precisely determined. However, glucocorticoid hormones seem to be involved, since Zn deficiency yields a chronic stimulation of corticosterone production(Reference Fraker, Osati-Ashtiani and Wagner28), and adrenalectomy prevents thymic atrophy secondary to Zn deficiency.

These studies raise concern about the impact of intrathymic cell death in human subjects who are deficient in Zn due to suboptimal diet or chronic disease(Reference Fraker29). In this respect, nutritional supplementation should be considered in chronically ill patients, with compromised immune defence, as reported in AIDS patients(Reference Baum, Shor-Posner and Campa30, Reference Baum, Campa and Lai31). Zn supplementation resulted in a significant increase in CD4+ T cells and a decreased mortality. This notion can also be applied in Chagasic patients, since they exhibit a decrease in serum Zn concentrations(Reference Burguera, Burguera and Alarcon32); the same being observed in a variety of haemopoietic organs of infected rats(Reference Matousek de Abel de la Cruz, Burguera and Burguera33). Accordingly, the severity of experimental Chagas disease is much higher in Zn-deficient mice(Reference Fraker, Caruso and Kierszenbaum34).

Acute infections induce thymic atrophy

Severe thymic atrophy is also a common feature in acute infections, reflecting the massive depletion of CD4+CD8+cortical thymocytes (Table 1). This has been shown in a variety of infections, such as AIDS, rabies, malaria, Chagas disease and schistosomiasis, among others (reviewed in(3)). In some cases, thymocyte loss is so severe that the cortical region of thymic lobules virtually disappears, as a consequence of the CD4+CD8+ thymocyte death. Also, similar to what is seen in malnutrition, proliferative response of thymocytes from acutely infected individuals is reduced: we found a significant decrease in both concanavalin A- and anti-CD3-driven proliferative responses in murine Chagas disease. Interestingly, this was paralleled by a decrease in the intrathymic production of IL-2, a major T-cell proliferation cytokine(Reference Leite-de-Moraes, Minoprio and Dy35).

Table 1. Thymic atrophy in human subjects and experimental infectious diseases (modified from(Reference Savino3))

SIV, Simian immunodeficiency virus; ND, not determined.

Thymocyte depletion seen in malnutrition and acute infections is partially under hormonal control

It is now well established that the physiology of the thymus (including both lymphoid and microenvironmental compartments) is influenced by a variety of hormones and neuropeptides(Reference Savino and Dardenne7). It has been shown that glucocorticoid-circulating levels are increased in protein-malnourished mice, as compared to age-matched controls. Additionally, implanted corticosterone-containing pellets, able to generate glucocorticoid serum levels equivalent to those found in malnourished mice, were sufficient to yield thymocyte depletion(Reference Barone, O'Brien and Stevenson36). As discussed later, leptin also seems to be involved. It has been shown that human subjects and rodents lacking proper leptin production or expressing defective leptin receptors, bear a certain degree of immunodeficiency characterized by reduced T-cell proliferative response to various mitogens, impaired production of IL-4 and inappropriate antibody production after immunization(Reference Munzberg and Myers37Reference Farooqi, Matarese and Lord39). Interestingly, leptin/leptin receptor-deficient animals exhibit an atrophy of lymphoid tissues, particularly the thymus, and such a defect can be reversed by the reposition of the hormone(Reference Howard, Lord and Matarese40). Leptin was also able to prevent starvation-induced thymic atrophy(Reference Howard, Lord and Matarese40, Reference Mito, Yoshino and Hosoda41), strongly suggesting that this hormone is one mediator of malnutrition-induced thymic atrophy. It is thus conceivable that in malnutritional states, the imbalance in the production of leptin (which is decreased) and glucocorticoid hormones (which are increased) is at least partially responsible for thymocyte depletion and consequent atrophy of the organ, as we previously proposed(Reference Savino42, Reference Savino, Dardenne and Veloso43).

The precise mechanisms responsible for the thymic atrophy seen in acute infections are not completely elucidated, and may vary in distinct diseases. But similar to malnutrition, one major pathway is related to the rise in glucocorticoid hormone levels in the blood, a classical effect comprised within the stress response of the organism to the infection. In fact, glucocorticoid serum levels are enhanced in Trypanosoma cruzi-infected mice(Reference Leite de Moraes, Hontebeyrie-Joskowicz and Leboulanger44, Reference Corrêa-de-Santana, Paez-Pereda and Theodoropoulou45), and, as discussed later, are likely involved, at least partially, in the T. cruzi-induced thymic atrophy(Reference Perez, Bottasso and Savino46). Thymocyte depletion seen in rabies virus-infected mice(Reference Cardenas-Palomo, de Souza-Matos and Chaves-Leal47) can be prevented by adrenalectomy prior to infection. In murine Chagas disease, adrenalectomy alone did not prevent T. cruzi-induced cortical thymocyte depletion(Reference Leite de Moraes, Hontebeyrie-Joskowicz and Leboulanger44). Nevertheless, more recently it was demonstrated that a complete functional inhibition of glucocorticoid receptors by in vivo injection of RU-486, did prevent thymocyte depletion following acute T. cruzi infection(Reference Roggero, Pérez and Tamae-Kakazu48). Whether leptin levels are down-regulated in acutely infected levels remains to be determined and represents an interesting open field of investigation.

The thymic microenvironment is altered in malnutrition and acute infections

In addition to the lymphoid compartment, the thymic microenvironment is affected in various malnutritional and infectious conditions. Morphological changes in the thymic epithelium from protein-malnourished mice include the decrease in the volume of the epithelial tissue in the cortex and in the medulla of thymuses from malnourished mice, as compared to well-nourished control animals(Reference Mittal, Woodward and Chandra49). By contrast, an increase of intracytoplasmic accumulations of large, circular, homogeneously electron-dense profiles, rich in free and esterified cholesterol was reported in both cortical and medullary TEC of malnourished animals(Reference Mittal and Woodward50). Unfortunately, no data were reported concerning TEC death in this experimental model.

In T. cruzi acutely infected mice, we demonstrated changes in the expression of medullary and cortical-specific markers, as compared to controls, together with a general shrinkage of the thymic epithelial network(Reference Savino, Leite de Moraes and Hontebeyrie-Joskowicz51).

Conceptually, these findings tell us that the thymic epithelium is morphologically altered in malnutrition and infection. As seen later, functional changes of the thymic epithelium are also seen in both these pathological conditions.

Decreased thymic endocrine function in malnourished and acutely infected individuals

One functional parameter that has been largely evaluated in malnutritional conditions is the thymic hormone production by TEC. It was initially found that that protein-malnourished mice exhibited abnormally low levels of circulating thymulin(Reference Chandra1, Reference Mittal, Woodward and Chandra49), and that such a decrease was also observed in protein-malnourished rats and human subjects(Reference Jambon, Ziegler and Maire52). Interestingly, even in human subjects protein malnutrition secondary to anorexia nervosa, low thymulin serum levels were reported(Reference Wade, Bleiberg and Mosse53). Furthermore, decreased thymulin serum levels were reported in mice submitted to diets designed to trigger deficiency in Zn, Fe, or vitamins(Reference Chandra1, Reference Kuvibidila, Dardenne and Savino14, Reference Dardenne, Savino and Wade54). At least regarding Zn deficiency, similar results were found in human subjects(Reference Prasad, Meftah and Abdallah55).

It is noteworthy that the decrease in thymic hormone serum levels seen in malnutrition is not restricted to thymulin, since it was recently reported with regard to thymopoietin production(Reference McDade, Beck and Kuzawa56). In this study, the authors further demonstrated that prenatal undernutrition was significantly associated with reduced thymopoietin production in interaction with the duration of exclusive breast-feeding. These findings provide support for the importance of fetal and early infant programming of thymic function, and long-term implications for the immune system, and consequently adult disease risk.

In severe infection conditions, thymic endocrine function is also affected. We observed in T. cruzi-infected mice a transient decrease in the serum levels of the thymic hormone thymulin(Reference Savino, Leite de Moraes and Hontebeyrie-Joskowicz51). In HIV infection, we and others showed a consistent and long-term diminution of thymulin secretion, in terms of both serum levels and intrathymic contents of the hormone(Reference Dardenne, Bach and Safai57Reference Savino, Dardenne and Marche59).

Increased extracellular matrix in the thymus of malnourished children

In addition to the abnormalities seen in TEC, the thymus from malnourished children presents a further microenvironmental alteration, namely, an increase in the deposition of ECM proteins. We studied by histological, ultrastructural and immunohistochemical means, thymuses obtained in necropsies from malnourished children. There is a consistent increase in the intralobular ECM-containing network, which could be ascertained histologically by the dense reticulin staining and immunohistochemically by the higher contents of fibronectin, laminin and type IV collagen. Importantly, the enhancement of thymic ECM in malnourished individuals positively correlated with the degree of thymocyte depletion(Reference Lyra, Madi and Maeda10). This correlation may represent a cause–effect relationship in which the contact of thymocytes with abnormally high amounts of thymic ECM triggers and/or enhances programmed cell death. However, this notion is still hypothetical, demanding experimental demonstration. Interestingly, similar changes in thymic ECM were observed in glucocorticoid-hormone-treated mice and TEC cultures(Reference Savino and Dardenne7), leading to the hypothesis that the enhanced ECM deposition seen in malnutrition may be also related to high levels of serum glucocorticoid hormones. Such an alteration was also seen in acute infections, as exemplified by experimental Chagas disease(Reference Savino, Leite de Moraes and Hontebeyrie-Joskowicz51, Reference Cotta de Almeida, Mendes da Cruz and Bonomo60). In this infection model, changes in ECM were accompanied by alterations in the migratory response of thymocytes, with an abnormal export of CD4+C8+ immature thymocytes, some of them having bypassed the normal selective selection process(Reference Cotta de Almeida, Mendes da Cruz and Bonomo60Reference Savino, Villa-Verde and Mendes-da-Cruz62). Whether similar cell migration abnormalities exist in malnourished subjects is to be investigated.

Changes in the patterns of thymocyte migratory responses in acute infections

In addition to the thymocyte depletion seen in several infectious diseases, changes in the migratory responses have also been observed. As mentioned earlier, thymocyte depletion parallels T. cruzi-induced alterations of the thymic microenvironment, comprising phenotypic changes and functional changes in the TEC network, with an enhancement in the deposition of cell migration-related molecules such the ECM proteins, fibronectin and laminin, as well as the chemokines CXCL12 and CCL21(Reference Cotta de Almeida, Mendes da Cruz and Bonomo60, Reference Mendes-da-Cruz, Silva and Cotta-de-Almeida61). These changes promote increased migratory responses to the corresponding ligands, and are likely related to the abnormal release of double-positive cells from the thymus into the periphery, resulting in more than 15-fold increase in double-positive cell numbers in subcutaneous lymph nodes. In this vein, it is noteworthy that double-positive cells seen in peripheral lymphoid organs express high densities of ECM and chemokine receptors(Reference Cotta de Almeida, Mendes da Cruz and Bonomo60, Reference Mendes-da-Cruz, Silva and Cotta-de-Almeida61). Among these abnormally released double-positive cells in the periphery, we found lymphocytes expressing potentially autoreactive TCR, which are normally deleted in the thymus of uninfected mice. This suggests that during the infection, immature T lymphocytes escape from the thymic central tolerance process and migrate to the lymph nodes where they eventually differentiate into mature CD4+ or CD8+ cells(Reference Savino3, Reference de Meis, Morrot and Farias-de-Oliveira63).

In a second murine model of parasitic diseases, the thymus was evaluated in mice acutely infected with Plasmodium berghei. Again there is a thymic atrophy, with loss of cortical-medullary limits and the intrathymic presence of parasites(Reference Andrade, Gameiro and Nagib64). We also analysed the thymic expression of ECM ligands and receptors, as well as chemokines and their respective receptors. An increased expression of ECM components was observed in the thymus from infected mice, in parallel to a down-regulation of fibronectin and laminin receptor surface expression in thymocytes from these animals. Moreover, in the thymus from infected mice, we found increased contents of CXCL12 and CXCR4 and decreased expression of CCL25 and CCR9. An altered thymocyte migration towards ECM elements and chemokines was seen when the thymus from infected mice were analysed. The evaluation of ex vivo migration patterns of CD4/CD8-defined thymocyte subpopulations revealed that double-negative and CD4+ and CD8+ single-positive cells from P. berghei-infected mice have higher migratory responses, as compared to controls. Interestingly, increased numbers of these T-cell subpopulations were found in the spleen of infected mice, suggesting an abnormal export of T lymphocytes from the thymus of mice undergoing acute malaria infection(Reference Gameiro, Nagib and Andrade65).

Conclusions

The various issues discussed earlier clearly show that the thymus is a constant target organ in malnutrition as well as in acute infections, being severely affected in both lymphoid and microenvironmental compartments, and resulting in abnormal intrathymic T-cell death, proliferation and migration. These changes likely have consequences, leading to the impaired peripheral immune responses, seen in malnourished and infected individuals. Thus, strategies able to promote thymus replenishment should be considered when designing adjuvant therapeutic approaches, in both malnutrition and acute infectious diseases.

Acknowledgements

This work was partially funded with grants from Fiocruz, CNPq, Faperj (Brazil) and CNRS (France). The authors declare no conflict of interest. Both authors contributed to the writing of the manuscript.

References

1.Chandra, RK (1992) Protein-energy malnutrition and immunological responses. J Nutr 122, Suppl. 3, 597600.CrossRefGoogle ScholarPubMed
2.Prentice, AM (1999) The thymus: a barometer of malnutrition. Br J Nutr 81, 345347.CrossRefGoogle ScholarPubMed
3.Savino, W (2006) The thymus is a common target organ in infectious diseases. PLoS Pathogens 2, 472483.CrossRefGoogle ScholarPubMed
4.Ciofani, M & Zúñiga-Pflücker, JC (2007) The thymus as an inductive site for T lymphopoiesis. Annu Rev Cell Dev Biol 23, 463493.CrossRefGoogle ScholarPubMed
5.Savino, W, Mendes da Cruz, DA, Silva, JS et al. (2002) Intrathymic T cell migration: a combinatorial interplay of extracellular matrix and chemokines? Trends Immunol 23, 305313.CrossRefGoogle ScholarPubMed
6Savino, W, Mendes-da-Cruz, DA, Smaniotto, S et al. . (2004) Control of thymocyte migration: an interplay of distinct cellular interactions. J Leukocyte Biol 75, 951961.CrossRefGoogle Scholar
7.Savino, W & Dardenne, M (2000) Neuroendocrine control of thymus physiology. Endocr Rev 21, 412443.Google ScholarPubMed
8.Petrie, HT & Züniga-Pflucker, JC (2007) Zoned out: functional maing of stromal signaling microenvironments in the thymus. Annu Rev Immunol 25, 649679.CrossRefGoogle Scholar
9.Mitsumori, K, Takegawa, K, Shimo, T et al. . (1996) Morphometric and immunohistochemical studies on atrophic changes in lympho-hematopoietic organs of rats treated with piperonyl butoxide or subjected to dietary restriction. Arch Toxicol 70, 809814.CrossRefGoogle ScholarPubMed
10.Lyra, JS, Madi, K, Maeda, CT et al. . (1993) Thymic extracellular matrix in human malnutrition. J Pathol 171, 231236.CrossRefGoogle ScholarPubMed
11Parent, G, Chevalier, P, Zalles, L et al. . (1994) In vitro lymphocyte-differentiating effects of thymulin (Zn-FTS) on lymphocyte subpopulations of severely malnourished children. Am J Clin Nutr 60, 274278.CrossRefGoogle ScholarPubMed
12.Aaby, P, Marx, C, Trautner, S et al. . (2002) Thymus size at birth is associated with infant mortality: a community study from Guinea Bissau. Acta Pediatr 91, 698703.CrossRefGoogle ScholarPubMed
13.Chevalier, P, Sevilla, R, Zalles, L et al. . (1996) Immuno-nutritional recovery of children with severe malnutrition. Santé 6, 201208.Google ScholarPubMed
14.Kuvibidila, S, Dardenne, M, Savino, W et al. . (1990) Influence of irondeficiency anemia on selected thymus functions in mice: thymulin biological activity, T-cell subsets, and thymocyte proliferation. Am J Clin Nutr 51, 228232.CrossRefGoogle ScholarPubMed
15.Dhur, A, Galan, P, Christides, JP et al. . (1991) Effect of folic acid deficiency upon lymphocyte subsets from lymphoid organs in mice. Comp Biochem Physiol A 98, 235240.CrossRefGoogle ScholarPubMed
16.Malpuech-Brugere, C, Nowacki, W, Gueux, E et al. . (1999) Accelerated thymus involution in magnesium-deficient rats is related to enhanced apoptosis and sensitivity to oxidative stress. Br J Nutr 81, 405411.CrossRefGoogle ScholarPubMed
17.Nodera, M, Yanagisawa, H & Wada, O (2001) Increased apoptosis in a variety of tissues of zinc-deficient rats. Life Sci 69, 16391649.CrossRefGoogle Scholar
18.Cunningham-Rundles, S, McNeeley, DF & Moon, A (2005) Mechanisms of nutrient modulation of the immune response. J Allergy Clin Immunol 115, 11191128.CrossRefGoogle ScholarPubMed
19Shankar, AH & Prasad, AS (1998) Zinc and immune function: the biological basis of altered resistance to infection. Am J Clin Nutr 68, 447S463S.CrossRefGoogle ScholarPubMed
20.Chesters, J (1977) Zinc. In Handbook of Nutritionally Essential Mineral Elements, pp. 185230 [Odel, B Sunde, R and EDS, editors]. New York: Marcel Dekker.Google Scholar
21.Fraker, P, King, L, Garvy, B et al. (1993) Immunopathology of zinc deficiency: a role for apoptosis. In Human Nutrition: A Comprehensive Treatise, pp. 267283 [Klurfeld, DM]. New York: Plenum Press.Google Scholar
22.Kuvibidila, S, Yu, L, Ode, D et al. (1993) The immune response in protein-energy malnutrition and single nutrient deficiencies. In Human Nutrition: A Comprehensive Treatise, pp. 121157 [Klurfeld, DM]. New York: Plenum Press.Google Scholar
23.Fraker, P, King, L, Laakko, T et al. . (2000) The dynamic link between the integrity of the immune system and zinc status. J Nutr 130, 13991406.CrossRefGoogle ScholarPubMed
24.Fernandes, G, Nair, N, Once, K et al. . (1979) Impairment of cell mediated immunity function in dietary zinc deficiency in mice. Proc Natl Acad Sci USA 76, 457461.CrossRefGoogle ScholarPubMed
25.Fraker, PJ, Depascale-Jardieu, R, Zwickl, CM et al. . (1978) Regeneration of T-cell helper function in zinc-deficient adult mice. Proc Natl Acad Sci USA 75, 56605664.CrossRefGoogle ScholarPubMed
26.King, LE, Osati-Ashtiani, F & Fraker, PJ (2002) A distinct role for apoptosis in the loss of precursor lymphocytes during zinc deficiency. J Nutr 132, 974979.CrossRefGoogle ScholarPubMed
27.Beach, RS, Gershwin, ME & Hurley, LS (1979) Altered thymic structure and mitogen responsiveness in postnatally zinc-deprived mice. Dev Comp Immunol 3, 725738.CrossRefGoogle ScholarPubMed
28.Fraker, P, Osati-Ashtiani, F, Wagner, MA et al. . (1995) Possible roles for glucocorticoids and apoptosis in the suppression of lymphopoiesis during zinc deficiency: a review. J Am Coll Nutr 14, 1117.CrossRefGoogle ScholarPubMed
29.Fraker, PJ (2004) Roles for cell death in zinc deficiency. J Nutr 135, 359362.CrossRefGoogle Scholar
30.Baum, M, Shor-Posner, G & Campa, A (2000) Zinc status in human immunodeficiency virus infection. J Nutr 130, 1421S1423S.CrossRefGoogle ScholarPubMed
31.Baum, M, Campa, A, Lai, S, et al. . (2003) Zinc status in human immunodeficiency virus type 1 infection and illicit drug use. Clin Infect Dis 37, 117123.CrossRefGoogle ScholarPubMed
32.Burguera, JL, Burguera, M, Alarcon, OM et al. . (1988) Concentration changes of zinc, copper and iron in serum of chronic chagasic myocardiopathic patients. J Trace Elem Electrolytes Health Dis 2, 215219.Google ScholarPubMed
33.Matousek de Abel de la Cruz, AJ, Burguera, AJ, Burguera, M et al. . (1993) Changes in total content of iron, copper and zinc in serum, heart, liver, spleen and skeletal muscle tissues of rats infected with Trypanosoma cruzi. Biol Trace Elem Res 37, 5170.CrossRefGoogle ScholarPubMed
34.Fraker, PJ, Caruso, R & Kierszenbaum, F (1982) Alteration of the immune and nutritional status of mice by synergy between zinc deficiency and infection with Trypanosoma cruzi. J Nutr 112, 12241229.CrossRefGoogle ScholarPubMed
35.Leite-de-Moraes, MC, Minoprio, P, Dy, M et al. . (1994) Endogenous IL-10 and IFN-g production controls thymic cell proliferation in mice acutely infected by Trypanosoma cruzi. Scand J Immunol 39, 5158.CrossRefGoogle Scholar
36.Barone, KS, O'Brien, PC & Stevenson, JR (1993) Characterization and mechanisms of thymic atrophy in protein-malnourished mice: role of corticosterone. Cell Immunol 148, 226233.CrossRefGoogle ScholarPubMed
37.Munzberg, H & Myers, MG Jr.( 2005) Molecular and anatomical determinants of central leptin resistance. Nat Neurosci 8, 566570.CrossRefGoogle ScholarPubMed
38.Lord, GM, Matarese, G, Howard, JK et al. . (1998) Leptin modulates the T-cell immune response and reverses starvationinduced immunosuppression. Nature 394, 897901.CrossRefGoogle ScholarPubMed
39.Farooqi, IS, Matarese, G, Lord, GM et al. . (2002) Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest 110, 10931103.CrossRefGoogle ScholarPubMed
40.Howard, JK, Lord, GM, Matarese, G et al. . (1999) Leptin protects mice from starvation-induced lymphoid atrophy and increases thymic cellularity in ob/ob mice. J Clin Invest 104, 10511059.CrossRefGoogle ScholarPubMed
41.Mito, N, Yoshino, H, Hosoda, T et al. . (2004) Analysis of the effect of leptin on immune function in vivo using diet-induced obese mice. J Endocrinol 180, 167173.CrossRefGoogle ScholarPubMed
42.Savino, W (2002) The thymus gland is a target in malnutrition. Eur J Clin Nutr 56 (Suppl 3), S46S49.CrossRefGoogle ScholarPubMed
43.Savino, W, Dardenne, M, Veloso, LA et al. . (2007) The thymus is a common target in malnutrition and infection. Br J Nutr 98, 1116.CrossRefGoogle Scholar
44.Leite de Moraes, MC, Hontebeyrie-Joskowicz, M, Leboulanger, F et al. . (1991) Studies on the thymus in Chagas’ disease. II. Thymocyte subset fluctuations in Trypanosoma cruzi-infected mice: relationship to stress. Scand J Immunol 33, 267275.CrossRefGoogle ScholarPubMed
45.Corrêa-de-Santana, E, Paez-Pereda, M, Theodoropoulou, M et al. . (2006) Hypothalamus-pituitary-adrenal axis during Trypanosoma cruzi acute infection in mice. J Neuroimmunol 173, 1222.CrossRefGoogle ScholarPubMed
46Perez, AR, Bottasso, O & Savino, W (2009) The impact of infectious diseases upon neuroendocrine circuits. Neuroimmunomodulation 16, 96–105.CrossRefGoogle ScholarPubMed
47.Cardenas-Palomo, LF, de Souza-Matos, DC, Chaves-Leal, E et al. . (1995) Lymphocyte subsets and cell proliferation analysis in rabies-infected mice. J Clin Lab Immunol 46, 4961.Google ScholarPubMed
48.Roggero, E, Pérez, AR, Tamae-Kakazu, M et al. . (2006) Endogenous glucocorticoids cause thymus atrophy but are protective during acute Trypanosoma cruzi infection. J Endocrinol 190, 495503.CrossRefGoogle ScholarPubMed
49.Mittal, A, Woodward, B & Chandra, RK (1988) Involution of thymic epithelium and low serum thymulin bioactivity in weanling mice subjected to severe food intake restriction or severe protein deficiency. Exp Mol Pathol 48, 226235.CrossRefGoogle ScholarPubMed
50.Mittal, A & Woodward, B (1985) Thymic epithelial cells of severely undernourished mice: accumulation of cholesteryl esters and absence of cytoplasmic vacuoles. Proc Soc Exp Biol Med 178, 385391.CrossRefGoogle ScholarPubMed
51.Savino, W, Leite de Moraes, MC, Hontebeyrie-Joskowicz, M et al. . (1989) Studies on the thymus in Chagas’ disease. I. Changes in the thymic microenvironment in mice acutely infected with Trypanosoma cruzi. Eur J Immunol 19, 17271733.CrossRefGoogle ScholarPubMed
52.Jambon, B, Ziegler, O, Maire, B et al. . (1988) Thymulin (facteur thymique serique) and zinc contents of the thymus glands of malnourished children. Am J Clin Nutr 48, 335342.CrossRefGoogle ScholarPubMed
53.Wade, S, Bleiberg, F, Mosse, A et al. . (1985) Thymulin (Zn-facteur thymique serique) activity in anorexia nervosa patients. Am J Clin Nutr 42, 275280.CrossRefGoogle ScholarPubMed
54Dardenne, M, Savino, W, Wade, S et al. (1984) In vivo and in vitro studies of thymulin in marginally zinc-deficient mice. Eur J Immunol 14, 454458.CrossRefGoogle ScholarPubMed
55.Prasad, AS, Meftah, S, Abdallah, J et al. . (1988) Serum thymulin in human zinc deficiency. J Clin Invest 82, 12021210.CrossRefGoogle ScholarPubMed
56.McDade, TW, Beck, MA, Kuzawa, CW et al. . (2001) Prenatal undernutrition and postnatal growth are associated with adolescent thymic function. J Nutr 131, 12251231.CrossRefGoogle ScholarPubMed
57.Dardenne, M, Bach, JF & Safai, B (1983) Low serum thymic hormone levels in patients with acquired immunodeficiency syndrome. N Engl J Med 309, 4849.Google ScholarPubMed
58.Incefy, GS, Pahwa, S, Pahwa, R et al. . (1986) Low circulating thymulin-like activity in children with AIDS and AIDS-related complex. AIDS Res 2, 109116.CrossRefGoogle ScholarPubMed
59Savino, W, Dardenne, M, Marche, C et al. . (1986) Thymic epithelium in AIDS: an immunohistologic study. Am J Pathol 122, 302307.Google ScholarPubMed
60.Cotta de Almeida, V, Mendes da Cruz, DA, Bonomo, A et al. . (2003) Acute Trypanosoma cruzi infection modulates intrathymic contents of extracellular matrix ligands and receptors and alters thymocyte migration. Eur J Immunol 33, 24392448.CrossRefGoogle ScholarPubMed
61.Mendes-da-Cruz, DA, Silva, JS, Cotta-de-Almeida, V et al. . (2006) Altered thymocyte migration during experimental acute Trypanosoma cruzi infection: combined role of fibronectin and the chemokines CXCL12 and CCL4. Eur J Immunol 36, 14861493.CrossRefGoogle ScholarPubMed
62.Savino, W, Villa-Verde, DMS, Mendes-da-Cruz, DA et al. . (2007) Cytokines and cell adhesion receptors in the regulation of immunity to Trypanosoma cruzi. Cytokine Growth Factor Rev 18, 107124.CrossRefGoogle ScholarPubMed
63.de Meis, J, Morrot, A, Farias-de-Oliveira, DA et al. (2009) Differential regional immune response in Chagas disease. PLoS Negl Trop Dis 3, e417.CrossRefGoogle ScholarPubMed
64.Andrade, CF, Gameiro, J, Nagib, PR et al. . (2008) Thymic alterations in Plasmodium berghei-infected mice. Cell Immunol 253, 14.CrossRefGoogle ScholarPubMed
65.Gameiro, J, Nagib, PRA, Andrade, CF et al. . (2010) Changes in cell migration-related molecules expressed by thymic microenvironment during experimental Plasmodium berghei infection: consequences on thymocyte development. Immunology 129, 248256.CrossRefGoogle ScholarPubMed
66Savino, W, Dardenne, M (2010) Pleiotropic modulation of thymic functions by growth hormone: from physiology to therapy. Curr Opinion Pharmacol 10, 434442.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Normal intrathymic T-cell differentiation and the thymic microenvironment. In the left panel, we show a simplified view of normal thymocyte differentiation. Bone marrow-derived precursors enter the thymus and migrate from the cortico-medullary junction to the subcapsular cortical region of the thymic lobules. These immature cells do not express the CD3/T-cell receptor (TCR) complex, neither CD4 or CD8 molecules, being referred as CD4CD8 double negative. As shown in the right panel, as developing thymocytes progress in differentiation, they interact with microenvironmental cells, such as cortical thymic epithelial cells and fibroblasts localized in the cortex. At this stage, thymocytes start to express the TCR/CD3 complex and the molecules CD4 and CD8, thus becoming TCRlow/CD3lowCD4+CD8+ double-positive for these molecules. These cells are submitted to the processes of positive and negative selection, as a consequence of the interaction with the thymic microenvironmental cells through MHC/peptide–TCR interactions. Cortical epithelial cells are involved in positive selection, whereas both dendritic cells and epithelial cells can drive negation selection. Negatively selected cells die by apoptosis (most of them being phagocytized by macrophages) and positively selected thymocytes progress in differentiation, migrating through the medulla, ultimately becoming mature TCRhigh/CD3highCD4+CD8 or TCRhigh/CD3highCD8+CD4 single-positive T lymphocytes, which are the cells that normally leave the organ. Based on Savino and Dardenne(66).

Figure 1

Table 1. Thymic atrophy in human subjects and experimental infectious diseases (modified from(3))