Apoptosis in the Homeostasis of the Immune System and in Human Immune Mediated Diseases

16
Current Pharmaceutical Design, 2008, 14, 253-268 253 1381-6128/08 $55.00+.00 © 2008 Bentham Science Publishers Ltd. Apoptosis in the Homeostasis of the Immune System and in Human Immune Mediated Diseases A. Giovannetti #,1 , M. Pierdominici #,2 , A. Di Iorio 3 , R. Cianci 4 , G. Murdaca 5 , F. Puppo 5 , F. Pandolfi 4 , R. Paganelli 3, * 1 Department of Clinical Medicine, Clinical Immunology and Allergy, University La Sapienza, Rome; 2 Dept. of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome; 3 Dept. of Medicine and Sciences of Aging, University G. d’Annunzio, Chieti, and Ce.S.I., Foundation University G. d’Annunzio, Chieti; 4 Department of Internal Medicine, Catholic University, Rome and 5 Dept. of Internal Medicine, University of Genoa, Italy Abstract: The immune system has evolved sophisticate mechanisms controlling the development of responses to dangerous antigens while avoiding unnecessary attacks to innocuous, commensal or self antigens. The risk of autoimmunity is continuously checked and bal- anced against the risk of succumbing to exogenous infectious agents. It is therefore of paramount importance to understand the molecular events linking the breakdown of tolerance and the development of immunodeficiency. Apoptotic mechanisms are used to regulate the de- velopment of thymocytes, the shaping of T cell repertoire, its selection and the coordinate events leading to immune responses in the pe- riphery. Moreover, they are at the heart of the homeostatic controls restoring T cell numbers and establishing T cell memory. T lympho- cytes shift continuously from survival to death signals to ensure immune responsiveness without incurring in autoimmune damage. In this review we shall consider some key facts on the relationship of lymphopenia to autoreactivity, the mechanisms controlling positive and negative selection in the thymus, the role of apoptosis in selected primary immunodeficiency states and in systemic and organ-specific autoimmunity, with examples from human diseases and their animal models. Key Words: Thymus, immunodeficiency, apoptosis, selection, autoimmunity, homeostasis, cytokines. INTRODUCTION Apoptosis plays an essential role in T cell development, the shaping of the immune repertoire, and in the ordinate initiation and final resolution of immune responses to exogenous dangerous sig- nals [1]. Thymocytes expressing nonfunctional or strongly autore- active T cell receptor (TCR) specificities are eliminated by apopto- sis during development [2], and tolerance is centrally induced through ectopic expression of self antigens allowed by the autoim- mune regulator (AIRE) gene [3]. Apoptosis also leads to the dele- tion of expanded effector T cells during immune responses as well as the preservation of T cell memory and the general homeostasis of immune cells [4]. The dysregulation of apoptosis in the immune system results in immunodeficiency and autoimmunity [5]. IMMUNE HOMEOSTASIS The numbers of T lymphocytes both in secondary lymphoid organs and in the circulation are kept under strict control [6], so that the CD3+ cell number is maintained even in the face of a sudden drop of a particular subset – as shown by the example of primary HIV infection [7, 8]. Homeostatic proliferation is the essence of this mechanism, which is called into action when lymphopenia occurs [9]. Lymphopenia induces proliferation of naïve T cells without CD28 costimulation [10] or antigenic stimulation, and in this condi- tion they acquire memory T cell markers [11]: however, not all lymphoid cells expand in this scenario, opening the door for replen- ishment by self-reactive cells [5, 12]. Interestingly, many autoim- mune diseases present with lymphopenia, and this is also oberved in animal models of human diseases [5, 13]. The reciprocal observa- tion that several activating factors stimulate T cell division and result in the generation of long-lived memory cells which reduce the incidence of autoimmunity, including bacterial antigens (adju- vants), chronic viral infections and mycobacterial species, repre- sents a cornerstone of the hygiene hypothesis of the rapid surge of immune mediated diseases in Western societies [5, 14, 15]. *Address correspondence to this author at the Dept. of Medicine and Sci- ences of Aging c/o Pal. Se. Bi.Via dei Vestini, 66013 Chieti scalo (CH) – Italy; E-mail: [email protected] # These authors contributed equally in the preparation of the manuscript. Cytokines IL-21 has been postulated to contribute to the generation of perturbed T cell homeostasis in autoimmune models of diseases [5, 12]. Part of its effects seems to be mediated by antagonizing the prosurvival effect of IL-15 on memory CD8+ T cells [12], decreas- ing Bcl-2 and Bcl-XL expression [16] thus targeting them for rapid elimination by apoptosis. In this context a temporary depletion of T cells in susceptible hosts would trigger homeostatic repopulation by peripheral tissue T cells when thymopoiesis is impaired and regula- tory function is inadequate [17]. When considered on the opposite side, both lack of IL-15 and IL-2 deficiency would result in devel- opment of autoimmune characteristics [18], as it is indeed observed in KO mice [19]. Also in these models, repopulation in the absence of T regulatory function [20] would give an advantage to a skewed anti-self repertoire. It is unclear at present which signals could drive homeostatic proliferation in the absence of these cytokines [21] essential for T lymphocyte growth. It is interesting to note that mild depletion of T lymphocytes with modified anti-CD3 monoclonal antibodies set the stage for new ways to manipulate the immune system in autoimmune type 1 (insulin-dependent) diabetes [22]. MOLECULAR SWITCHES FOR T CELL APOPTOSIS During adaptive immune responses, as mentioned in the intro- duction, an expansion phase of T cells, differentiating into effector and memory subsets through asymmetric rounds of division [23], must be followed by a contraction phase to reach normal numbers [24]. This goal is reached by induction of apoptosis through activa- tion-induced and activated T cell autonomous cell death [25]. Sev- eral factors control the sensitivity to apoptosis induction during the different phases of the immune response [26], thus acting as mo- lecular switches between T cell survival and death. The engagement of the T cell receptor (TCR) distinguishes the two apoptotic path- ways, which both result in caspase activation. The molecular switches so far identified range from cytokines (mainly chain containing such as IL-2, IL-7, IL-15 and IL-21, but also IL-6 and IFN- ), death receptors (Fas and TNFR-I), Bcl-2 proteins, NF- B, other kinases and specific organelles (mitochondria and lysosomes). The understanding of the regulation and reciprocal interactions of

Transcript of Apoptosis in the Homeostasis of the Immune System and in Human Immune Mediated Diseases

Current Pharmaceutical Design, 2008, 14, 253-268 253

1381-6128/08 $55.00+.00 © 2008 Bentham Science Publishers Ltd.

Apoptosis in the Homeostasis of the Immune System and in Human Immune Mediated Diseases

A. Giovannetti#,1, M. Pierdominici#,2, A. Di Iorio3, R. Cianci4, G. Murdaca5, F. Puppo5, F. Pandolfi4,

R. Paganelli3,*

1Department of Clinical Medicine, Clinical Immunology and Allergy, University La Sapienza, Rome;

2Dept. of Cell Biology and

Neurosciences, Istituto Superiore di Sanità, Rome; 3Dept. of Medicine and Sciences of Aging, University G. d’Annunzio, Chieti, and

Ce.S.I., Foundation University G. d’Annunzio, Chieti; 4Department of Internal Medicine, Catholic University, Rome and

5Dept. of

Internal Medicine, University of Genoa, Italy

Abstract: The immune system has evolved sophisticate mechanisms controlling the development of responses to dangerous antigens while avoiding unnecessary attacks to innocuous, commensal or self antigens. The risk of autoimmunity is continuously checked and bal-

anced against the risk of succumbing to exogenous infectious agents. It is therefore of paramount importance to understand the molecular events linking the breakdown of tolerance and the development of immunodeficiency. Apoptotic mechanisms are used to regulate the de-

velopment of thymocytes, the shaping of T cell repertoire, its selection and the coordinate events leading to immune responses in the pe-riphery. Moreover, they are at the heart of the homeostatic controls restoring T cell numbers and establishing T cell memory. T lympho-

cytes shift continuously from survival to death signals to ensure immune responsiveness without incurring in autoimmune damage. In this review we shall consider some key facts on the relationship of lymphopenia to autoreactivity, the mechanisms controlling positive and

negative selection in the thymus, the role of apoptosis in selected primary immunodeficiency states and in systemic and organ-specific autoimmunity, with examples from human diseases and their animal models.

Key Words: Thymus, immunodeficiency, apoptosis, selection, autoimmunity, homeostasis, cytokines.

INTRODUCTION

Apoptosis plays an essential role in T cell development, the shaping of the immune repertoire, and in the ordinate initiation and final resolution of immune responses to exogenous dangerous sig-nals [1]. Thymocytes expressing nonfunctional or strongly autore-active T cell receptor (TCR) specificities are eliminated by apopto-sis during development [2], and tolerance is centrally induced through ectopic expression of self antigens allowed by the autoim-mune regulator (AIRE) gene [3]. Apoptosis also leads to the dele-tion of expanded effector T cells during immune responses as well as the preservation of T cell memory and the general homeostasis of immune cells [4]. The dysregulation of apoptosis in the immune system results in immunodeficiency and autoimmunity [5].

IMMUNE HOMEOSTASIS

The numbers of T lymphocytes both in secondary lymphoid organs and in the circulation are kept under strict control [6], so that the CD3+ cell number is maintained even in the face of a sudden drop of a particular subset – as shown by the example of primary HIV infection [7, 8]. Homeostatic proliferation is the essence of this mechanism, which is called into action when lymphopenia occurs [9]. Lymphopenia induces proliferation of naïve T cells without CD28 costimulation [10] or antigenic stimulation, and in this condi-tion they acquire memory T cell markers [11]: however, not all lymphoid cells expand in this scenario, opening the door for replen-ishment by self-reactive cells [5, 12]. Interestingly, many autoim-mune diseases present with lymphopenia, and this is also oberved in animal models of human diseases [5, 13]. The reciprocal observa-tion that several activating factors stimulate T cell division and result in the generation of long-lived memory cells which reduce the incidence of autoimmunity, including bacterial antigens (adju-vants), chronic viral infections and mycobacterial species, repre-sents a cornerstone of the hygiene hypothesis of the rapid surge of immune mediated diseases in Western societies [5, 14, 15].

*Address correspondence to this author at the Dept. of Medicine and Sci-

ences of Aging c/o Pal. Se. Bi.Via dei Vestini, 66013 Chieti scalo (CH) –

Italy; E-mail: [email protected] #These authors contributed equally in the preparation of the manuscript.

Cytokines

IL-21 has been postulated to contribute to the generation of perturbed T cell homeostasis in autoimmune models of diseases [5, 12]. Part of its effects seems to be mediated by antagonizing the prosurvival effect of IL-15 on memory CD8+ T cells [12], decreas-ing Bcl-2 and Bcl-XL expression [16] thus targeting them for rapid elimination by apoptosis. In this context a temporary depletion of T cells in susceptible hosts would trigger homeostatic repopulation by peripheral tissue T cells when thymopoiesis is impaired and regula-tory function is inadequate [17]. When considered on the opposite side, both lack of IL-15 and IL-2 deficiency would result in devel-opment of autoimmune characteristics [18], as it is indeed observed in KO mice [19]. Also in these models, repopulation in the absence of T regulatory function [20] would give an advantage to a skewed anti-self repertoire. It is unclear at present which signals could drive homeostatic proliferation in the absence of these cytokines [21] essential for T lymphocyte growth.

It is interesting to note that mild depletion of T lymphocytes with modified anti-CD3 monoclonal antibodies set the stage for new ways to manipulate the immune system in autoimmune type 1 (insulin-dependent) diabetes [22].

MOLECULAR SWITCHES FOR T CELL APOPTOSIS

During adaptive immune responses, as mentioned in the intro-duction, an expansion phase of T cells, differentiating into effector and memory subsets through asymmetric rounds of division [23], must be followed by a contraction phase to reach normal numbers [24]. This goal is reached by induction of apoptosis through activa-tion-induced and activated T cell autonomous cell death [25]. Sev-eral factors control the sensitivity to apoptosis induction during the different phases of the immune response [26], thus acting as mo-lecular switches between T cell survival and death. The engagement of the T cell receptor (TCR) distinguishes the two apoptotic path-ways, which both result in caspase activation. The molecular switches so far identified range from cytokines (mainly chain containing such as IL-2, IL-7, IL-15 and IL-21, but also IL-6 and IFN- ), death receptors (Fas and TNFR-I), Bcl-2 proteins, NF- B, other kinases and specific organelles (mitochondria and lysosomes). The understanding of the regulation and reciprocal interactions of

254 Current Pharmaceutical Design, 2008, Vol. 14, No. 3 Giovannetti et al.

these switches may lead to new ways to control activated T cells that have escaped apoptosis in autoimmune disorders [25].

In the following sections we shall delineate the intrathymic selection of the T cell repertoire with discrete steps enabling central mechanisms of self-tolerance [27], the aberrations of apoptotic mechanisms leading to autoimmune diseases in genetic defects such as autoimmune lymphoproliferative syndrome (ALPS) and other primary immunodeficiencies, and finally we shall offer some ex-amples of the role of apoptotic imbalance in intestinal immuno-mediated disorders such as inflammatory bowel disease (IBD) and celiac disease.

THE THYMUS IN T CELL DEVELOPMENT AND REPER-TOIRE SHAPING.

Thymus Colonization

In order to generate T cells, hemopoietic precursors are required to colonize the thymic microenvironment. Interestingly, the site of production of T cell precursors changes during ontogeny. Fetal liver-derived precursors colonize the early fetal thymus while the postnatal thymus is colonized by precursors coming from the bone marrow [28]. Differently from adult precursors, fetal precursors have less myeloerythroid potential and can differentiate in the ab-sence of IL-7 [29]. The reasons for these differences are not clear. A possible explanation is that fetal and adult precursors receive distinct prethymic maturational signals capable to regulate their lineage potential. In this regard it should be noted that the fetal liver microenvironment contains discrete Notch ligand-bearing stromal cells that could activate Notch signalling (a pathway strongly impli-cated in T/B lineage choice) in fetal precursors prior to thymus colonization [30] (Fig. (1)). Although the mechanisms allowing precursors to exit blood vessels and entry to the thymus are still undefined, several molecules have been implicated in this process including the CD34, MECA79, VCAM-1, ICAM-1, and P-selectin [31]. However, recruitment of precursors to the thymus is also a temporally regulated process, in which a key role might be exerted by changes in expression of adhesion molecules as well as by intra-thymic competition for stromal cell niches [32].

Interleukin-7

The essential role of IL-7 in T cell development is well estab-lished. Its specific receptor consists of two chains, IL-7R and gamma common ( c). The latter is also part of the receptors for fundamental cytokines such as IL-2, IL-4, IL-9, IL-15, and IL-21. Genetic defects involving the genes encoding for c [33], IL-7R [34], or the Janus kinase 3 (Jak3, a component of the IL-7-induced signal transduction pathway) [35] account for the majority of severe combined immune deficiencies (SCID) characterized by strongly reduced numbers or even absence of T cells (see following section). It has been reported that thymocytes from c-deficient infants pos-sess TCR D-J but lack V-DJ rearrangements [36]. This finding may explain the complete lack of T cells in many c- and IL-7R -deficient patients. Since IL-7 is also required for peripheral T cell homeostasis [37], it can be speculated that the absolute T cell defi-ciency in c-deficient patients is caused by a combination of defects in early T cell development and in T cell homeostasis. IL-7, is es-sential for survival through the induction of the anti-apoptotic MCL-1. IL-7R - or common chain -deficient animals exhibit a block of T cell development at the DN 2-3 stages, [38] Similarly, the T-cell-specific MCL-1 deficiency, results in thymic atrophy and enlargement of the DN compartment [39].

T Cell Progenitors (TPs)

TPs enter the thymus at the cortical-medullary junction. A pro-portion of the precursors that seed the human thymus are unques-tionably multipotential whereas is still unclear whether some of the precursors that migrate into the thymus are already lineage re-stricted before their entrance. Likely, CD7 and CD10 define the

CD34+CD45RA

+ precursors biased to lymphoid lineages. An analy-

sis of the TCR rearrangement status indicates that the most imma-ture population in the thymus is CD34

+ CD10+CD38

lo [40], proba-

bly the direct progeny of the CD34+CD45RA

+CD7

+CD10

+CD38

lo

cord blood cells that have T, B, NK and some GM precursor activi-ties. The presence of multipotential precursors in the human thymus indicates that T cell commitment takes place within this organ. Complete TCR and partial TCR (D -J ) rearrangements have been reported in CD34

+CD7

+ cord blood precursors [41] and T

cell–restricted precursors were identified in human bone marrow [42] but whether these cells can migrate to the thymus is still un-clear. ETP have a CD34

+CD1a

- phenotype. Downstream of the ETP

is the CD4 immature single-positive (CD4 ISP) [43] population which can be divided into CD4

+CD1a

+ (CD4 ISP

lo) and CD4

hi

CD1ahi

(CD4 ISPhi

) populations (Fig. (1)). The subsequent subsets are the CD4

+CD8

+ - (early double-positive) and CD4

+CD8

+ +

populations, the latter cells being the precursors of double-positive TCR

+ cells.

Thymic Microenvironment

The pool of self-epitopes available for repertoire selection com-prises intrathymically expressed ubiquitous antigens and antigens specific to various types of thymic APCs such as cortical thymic epithelial cells (cTECs) and medullary TECs (mTECs), thymic dendritic cells, macrophages, and thymic B cells. Each of these cells presenting unique sets of self-peptides, contribute to the diver-sity of self-antigen displayed in the thymus. Self-antigens have access to the thymus also via the circulation or by association with immigrating cells [44]. However these latter routes are unlikely to allow for a comprehensive array of extrathymic tissue antigens. Therefore, mechanisms of peripheral tolerance, i.e. when the first encounter of a T cell with its cognate TSA occurs outside of the thymus, have been suggested in order to explain the absence of auto-reactivity toward tissue-restricted self-constituents.

Recent findings, showing that TSAs are expressed in the thy-mus and participate in selection of the T cell repertoire, have lead to a reconsideration of the capacity of central tolerance. It is now commonly accepted that expression of TSAs, in evident contraven-tion of the rules established for cell type–specific regulation of gene expression, is a natural property of TECs, and particularly of mTECs, a phenomenon known as promiscuous gene expression [45]. By inclusion of self-antigens that represent most if not all parenchymal organs [46], promiscuous gene expression widens the extent of central tolerance in a manner that was not expected by original concepts of self–non-self discrimination. However, with the notable exception of the involvement of the autoimmune regula-tor Aire, both molecular and cellular regulation of this unconven-tional gene expression pattern are still poorly understood.

Development of T Cells ( / -Lineage Choice)

Two major types of mature T cells are generated in the thymus that can be distinguished by the clonotypic subunits contained within their TCR complexes: T cells and T cells. T cells and precursors to the T cell lineage (bearing the pre-TCR) are thought to be derived from a common CD4

-CD8

- DN precursor

[47].

DN thymocytes that have successfully rearranged TCR- and TCR- chains and expressed a mature TCR are capable of differ-entiating along the -lineage pathway remaining in the DN stage [48]. As opposite, cells harboring in frame TCR- rearrangement and expressing a pre-TCR are capable to differentiate along the -lineage pathway, passing from the DN to the CD4

+CD8

+ DP stage.

Most T cells are DN, except for some intestinal epithelial lym-phocytes, which can express CD8 homodimers. Instead, -lineage cells express, with few exceptions, either CD4 or CD8 heterodimers [49].

Apoptosis in the Homeostasis of the Immune System and in Human Current Pharmaceutical Design, 2008, Vol. 14, No. 3 255

An unsolved matter is whether pre-TCR- and TCR-mediated signals dictate versus T cell lineage choice, and if so, how these signals differ. Two models have been initially proposed: the stochastic/selective and instructive models. According to the first the / -lineage commitment is determined independently of TCR signalling whereas the instructive model postulates that commit-ment is directly determined by the TCR isotype [50]. Now evidence exists supporting a model of lineage commitment based on the sig-nal strength whereby stronger signals promote development while weaker signals promote the fate, irrespective of the TCR isotype [51]. During early stages in T cell development, the TCR loci undergo rearrangement in a sequence TCR > > > [52] but it is still unclear in which subset the rearrangement of particular loci occurs. The current opinion is that expression of a TCR pro-tein and the resulting -selection occur within a certain develop-mental window and are not closely coupled to regulation of CD4, CD8 , and CD8 expression. Currently available data indicate the existence of a first limited number of cells that undergo the -selection before CD4 is expressed [52], a second, larger proportion of cells that is -selected after upregulation of CD4 [53], and finally a third group of the pre-T cells that upregulates CD4 and CD8 before initiating and completing TCR rearrangements [54] (Fig. (1)).

The Pre-TCR

The pre-TCR is initially expressed at the CD25+44

- DN3 stage,

following productive rearrangement of the TCR- locus. TCR- pairs with the pre-TCR- and molecules of the CD3 family forming a receptor that is capable to be expressed on the cell surface.

Pre-TCR signalling has the ability to regulate several transcrip-tion factors NF- B, NFAT [55], E2A/Id proteins [56], and -catenin/TCF-1 [57]. The expression of the pre-TCR receptor on the cell surface associates with the diminution of the IL-7R, Notch, and the hedgehog pathways, indicating that the pre-TCR may substitute the biological role of these pathways during T cell development. The pre-TCR differs from 'mature' and TCR in terms of both signalling initiation and biological outcome. The pre-TCR is a ‘weak’ signal transducer and that the commitment of immature DN cells to the lineage seems depends on these weak signals. Since all three receptors use identical signalling pathways and activate similar transcription factors there must be something unique about the pre-TCR that is not shared with the two other mature TCRs. As indicated by studies using transgenic mice expressing tailless pre-TCR forms, the main functions of the pre-TCR receptor are the pre-T cell proliferation and the development of a normally sized thymus [58]. In Pre-TCR-deficient mice thymi are small and the T cell development arrested at the DN3 stage. Pre-TCR-selected cells display a number of characteristics of cell-cycle entry including hyperphosphorylation of pRb, increased expression of cyclins A and B, downregulation of p27, increased cell CDK2 activity, and DNA synthesis. Unfortunately mechanisms of survival and apopto-sis in immature thymocytes, before the expression of antigen recep-tors and MHC recognition, are poorly understood. Probably, early thymic immigrants depend on prosurvival signals deriving from cytokine receptors for the upregulation of the anti-apoptotic Bcl-2 gene expression [59]. In the absence of these survival signals, the pro-apoptotic Bax translocates to mitochondria where it begins a death cascade.

Fig. (1). Developmental stages of intrathymic differentiation. The strong influence of Notch signalling in driving early precursors through DN stages 1-3a and

affecting the choice between or rearrangement is highlighted. After selection is made, only rearranged TCRB+ progress to DP through the immature

single positive (CD4 ISP) stage. Positive/negative selection occurs at the DP stage. Differential expression of several membrane antigens is shown to distin-

guish DN discrete maturative steps. TCR rearrangements are indicated in the lower part. Most extrinsic signals controlling the sequential entry are discussed in

the text, at each checkpoint failure to express more mature phenotype results in apoptotic elimination of thymocytes.

256 Current Pharmaceutical Design, 2008, Vol. 14, No. 3 Giovannetti et al.

Signalling pathways inducing pre-T cell death in the absence of a pre-TCR are largely unknown. The initially proposed requirement of Fas-induced death pathway for pre-T cell death, has been re-cently challenged. Fas belongs to a subgroup of the TNFR family that interacts through their death domains with cytoplasmic adapt-ers like FADD and TNF-R1-ADD [60]. FADD functions as an adapter molecule in apoptosis initiated by Fas, TNFR-I, DR3, and TNF-related TRAIL receptors. On one hand, the demonstration that Rag

-/- FADD

-/- immature T cells can differentiate and reach the DP

stage indicates that FADD could be essential for the transduction of death signals resulting in pre-T cell death. On the other hand, FADD deficiency leads to inhibition of T cell development at the DN stage (with a consequent reduction in the number of mature T cells) and FADD mutation does not affect apoptosis or signalling events triggered by the pre-TCR [61]. Since the introduction of a TCR transgene failed to rescue the mutant phenotype, these obser-vations suggested that FADD is required for the proliferative phase of early T cell development.

The p53 pathway is involved in both cell survival and prolifera-tion. In fact, p53 regulate not only pro-apoptotic genes such as Bax, Bid, caspase-6, Puma, and Noxa but also cell-cycle regulators like p21

cip. It has been reported that DNA breaks resulting from recom-

bination events trigger death in pre-T cells through action of p53 whereas inactivation of p53 promotes the survival and differentia-tion of pre-TCR-deficient thymocytes [62], Therefore it can be speculated that pre-TCR regulates progression through the DNA-damage checkpoint of the DN to DP transition by inactivating p53.

Bcl-2 is expressed in the thymus, and it has been suggested to induce survival of immature lymphocyte progenitors. The central role of the mammalian pro-survival Bcl-2 proteins, like their virally encoded orthologs, is to inhibit Bax and Bak. Thus, BH3-only pro-teins trigger apoptosis indirectly through Bax or Bak by neutraliz-ing all of the relevant prosurvival proteins, allowing the activation of Bax and Bak to proceed.

However, it appears not to be essential at the stage of pre-TCR selection. Pre-TCR signalling induces Ca

+2 mobilization and activa-

tion of PLC kinase and NF- B transcription factor. Through the induction of genes whose products can inhibit cell death NF- B acts as an important regulator of pre-T cell survival. These genes include cellular inhibitors of apoptosis, caspase-8–c-FLIP and two members of the Bcl-2 family, Bcl-xL and BCL2A.

BCL2A1 (A1) is expressed at the DN thymic compartment and appears as an ideal candidate for the regulation of pre-T cell apop-tosis. Similarly to Bcl-2 and Bcl-xL, A1 contains all major homol-ogy domains of the Bcl-2 family [63], is developmentally regulated in the immune system and is induced on cellular activation [64]. When transfected into cell lines A1 slows cell death and heterodi-merizes with the pro-apoptotic Bax [65]. Moreover, A1 sequesters truncated Bid in order to inhibit its collaboration with pro-apoptotic Bak or Bax [66]. Overexpression studies showed that A1 could protect thymocytes from several death stimuli, including pre-TCR deficiency, suggesting an important role for this molecule in pre-T- cell survival [67].

Positive Selection and Negative Selection

A key step during T cell development is when immature CD4

+CD8

+ thymocytes undergo positive and negative selection

events based on the specificities of the TCR complexes they express. Thymocytes which recognize self-peptide/MHC com-plexes with a low avidity undergo a positive selection process re-sulting in rescue from cell death and the induction of a differentia-tion program leading to the generation of MHC class II-restricted helper CD4

+ T cells and MHC class I-restricted cytotoxic CD8

+ T

cells [68] . As opposite, thymocytes bearing potentially autoreactive TCR specificities that recognize peptide/MHC complexes with high avidity, undergo negative selection by the induction of apoptosis [69]. As a consequence of these combined processes, a self-tolerant

T cell pool capable of recognizing foreign antigens in the context of self-MHC molecules is generated.

Every day of postnatal life approximately 95% of the cells that reach the DP stage fail to become functional lymphocytes and die strengthening the concept that positive selection is a wasteful proc-ess. This highlights the critical role that positive selection plays in the establishment of a working immune system. With the positive selection is achieved the fundamental task for the development of T cells to match the large number of TCR specificities generated by gene rearrangement and the independent expression of a subset of MHC molecules from the polymorphic pool of MHC genes.

However, TCR recognition of self-MHC has been demonstrated in additional mechanisms including the regulation of mature naïve T cell survival [69], the homeostatic T cell proliferation [70] and the establishing of the threshold of T cell activation [45]. Therefore one can speculate that the stimulation of mature T cells by self-MHC may serve to maintain the two hallmarks of T cell recognition i.e. the high sensitivity for activation and the highly discriminate recognition of antigen. Positive selection is also a lineage diversifi-cation process, where randomly generated TCR specificities ex-pressed by DP cells are shunted into appropriate functional cell types. Such a process necessitates a TCR-mediated recognition event in order to distinguish expression of class I and class II MHC-specific antigen receptors that differ only in membrane-distal TCR-variable regions.

Negative selection often operates after positive selection and thus on the smaller population of thymocytes that have migrated to the cortico-medullary junction as part of their maturation process. Although the details of negative selection are scarcely understood it seems a process independent from positive selection and the associ-ated lineage commitment.

The first checkpoint, so-called beta-selection, is contingent upon pre-TCR signalling and ensures that only those DN thymo-cytes that have successfully rearranged their TCR locus progress to the CD4

+CD8

+ (DP) compartment.

For many years the removal of strongly self-reactive T cells has been viewed as the only purpose of negative selection. However, recent evidence drives now to reconsider in part this opinion indi-cating for negative selection a more basic role in molding the T cell repertoire. Huseby and colleagues [71], by limiting negative selec-tion, have demonstrated that after TCR rearrangement, most T cells display a very high affinity for MHC. Therefore, in addition to re-move undesirable clones, with the potential to drive autoimmunity, negative selection exerts an essential role in removing the T cells bearing TCR with the default, unmodified, high affinity to MHC. The survived T cells will be able to bind low affinity MHC, thus allowing the specificity of the adaptive immunity.

In spite of intensive investigations, the molecular mechanisms of clonal deletion remain to be fully elucidated [72]. This may be due to the different models developed for testing negative selection often involving different mechanisms.

The most broadly used models for negative selection are those of TCR transgenic mice expressing a receptor specific for a self-antigen along with that self-antigen.

These kinds of models may be divided according to the stage in which clonal deletion occurs. Among the possible explanations for why some transgenics delete early (at the DN stage) and others delete late (at the SP stage) there are:

1) the anatomy of self-antigen expression (DN cells reside in the subcapsular area or outer cortex and SP cells reside in the me-dulla);

2) the type of APC that presents the particular self-antigen;

3) the affinity of the TCR for self-antigen (DP thymocytes, ex-pressing TCR at lower level than SP might require a stronger TCR stimulus to die [73].

Apoptosis in the Homeostasis of the Immune System and in Human Current Pharmaceutical Design, 2008, Vol. 14, No. 3 257

The injection of specific peptide into TCR transgenic animals also results in apoptosis of DP thymocytes [74] eliminating the concern about the artificial nature of cross-linking antibodies as TCR ligands. However, the massive activation of the cells respond-ing to the stimulus might cause nonspecific death of thymocytes [75] as well as other nonspecific effects such as a cytokine-mediated stromal cell–activation process [76], and the collapse of the thymic architecture [77].

Up to date the more physiologic models to study the antigen-induced clonal deletion are those involving TCR transgenic strain [78]. In fact this model does not present the nonspecific activation observed in other models. On the other hand, the reduced frequency makes the responders difficult to detect and study.

In some models clonal deletion does not occur when TCR transgenics are crossed to animals expressing the high-affinity ligand [79] . In these cases thymocytes expressing endogenously rearranged T cell receptors are preferentially generated. Two expla-nations have been proposed for such unusual findings: 1) the pref-erential survival of cells that had undergone secondary rearrange-ment prior to antigen encounter [80] and, 2) the upregulation of gene rearrangement signaled via the TCR [79]. The second hy-pothesis corresponds to the receptor editing process that occurs in developing B cells [81]. The reasons why deletion predominates in some models but not others are still undefined but could involve factors such as the timing and level of TCR expressed in different transgenics. Therefore, a key question is whether antigen-specific receptor editing occurs also in normal animals and whether some of the 30% of peripheral T cells that express two functional TCR rearrangements could be autoreactive.

Generation of T Regulatory Cells in the Thymus

The tolerance to self-antigens is realized in the thymus by mul-tiple mechanisms, including the generation of Treg and clonal dele-tion by apoptosis. Evidence exists indicating bone marrow-derived DCs and thymic epithelial cells as important mediators of thymo-cyte negative selection and Treg development, respectively [82, 83]. Dendritic cells expressing MHC class I and class II molecules are strategically located in thymus at the level of cortico-medullary junction. Here, they are supposed to screen the TCR specificities of positively selected CD4

+CD8

+ thymocytes during their migration

from the cortex to the medulla. However, negative selection may also occur when corticomedullary migration of thymocytes is ab-normal. It has been reported that the recruitment of new T cell pre-cursors to the thymus is accompanied by the migration of DCs, in order to ensure efficient negative selection during each wave of T cellprecursor development. Medullary epithelial cells might play a role in the recruitment and positioning of DCs in the thymus. In fact, mice deficient in Rel-b lack thymic DC cells, which has been shown to be a direct consequence of an absence of an organized thymic medulla [84].

The majority of Treg cells represent a separate, thymus-derived lineage of CD4 T cells [85]. Although different studies using TCR transgenic systems have demonstrated that the thymic epithelium plays an essential role for Treg development [83] in vitro studies [86] support a role for human thymic DCs in inducing Tregs. Data obtained in transgenic mice that likely express MHC class II mole-cules only on cTECs [86] suggest a cortical origin of the Treg line-age. In these mice both the number of CD4 SP thymocytes express-ing the CD25 marker and the suppressive function of peripheral CD4 CD25

+ T cells were similar to that observed in controls [87].

Therefore according to these data the self-antigen encounter in the medulla would be dispensable. However, the lack of rigorous con-trols (e.g. the capacity of isolated mTECs from K14-IA

b mice to

present MHC class II–restricted antigens in vitro) induce us to take with extreme caution these data. The timing and location of Treg commitment within the thymus has recently been analyzed using a mouse that expresses a fusion protein of FoxP3 and green fluores-

cent protein. FoxP3-reporter expression could be detected in thy-mocytes within the medulla but not in the cortex [88]. In support of a role of mTECs in the selection of Tregs are also a number of tar-geted mutations that, altering the medullary architecture and mTEC differentiation, lead to deeply reduced numbers of CD25

+ Tregs

[89]. However, extrathymic antigen encounter can induce the de novo generation of Treg from naive CD4+ T cells [90] qualitatively indistinguishable from that induced by mTECs in the thymus.

PRIMARY IMMUNODEFICIENCIENCY DISEASES AND

APOPTOSIS

Apoptosis, a physiological process of cell suicide, plays a piv-otal role in the maintenance of homeostasis of the immune system eliminating unwanted cells [91]. If decreased apoptosis can cause an overabundance of lymphocytes and possibly cancer as well as autoimmune and allergic reactions, enhanced apoptosis can result in lymphocyte depletion and immunodeficiency. Here, we will focus on the role of apoptosis in the pathogenesis of innate defects of the immune system, i.e. primary immunodeficiency diseases, and dis-cuss the importance of apoptosis modulation in both the pathogene-sis and treatment of these diseases.

Primary immunodeficiency diseases consist of a group of more than 100 inherited conditions which affect one or more components of the immune system [92-94]. Diseases in which lymphocyte func-tion is predominantly affected are commonly divided into three main groups, i.e. antibody deficiencies, cellular deficiencies and combined immunodeficiencies. In the antibody deficiencies, the genetic lesions mainly affects antibody production whereas in the cellular deficiencies cellular effector mechanisms are primarily compromised. In the combined immunodeficiencies both effector arms of specific immunity are impaired. This category contains the subgroup of SCID in which lymphocyte-dependent specific immu-nity is completely absent. Two other classes of primary immunode-ficiencies are also described: defects in phagocyte function and complement deficiencies. The clinical presentation of patients with a primary immunodeficiency reflects the type of defect present in the immune system and depends on the underlying genetic defect. Patients with SCID generally present with opportunistic infections and fail to thrive within a few months of life. Recurrent pyogenic infections are the hallmark of disease in patients with defects in B cells, phagocytic cells or complement, whereas opportunistic infec-tions with viruses or fungi are frequent in patients with T cell defi-ciencies. Primary immunodeficiencies frequently associate with autoimmune diseases, allergy and malignancies [95].

In the last 15 years the genetic basis of most primary immu-nodeficiencies has been identified. Many of these are associated with single gene defects, whereas others may be polygenic or repre-sent interactions of genetically determined characteristics with envi-ronmental factors [96, 97].

In some primary immunodeficiencies abnormalities of apopto-sis reflect a defect of gene(s) involved in the known mechanisms of cell death, e.g. Bcl-2 and Fas. In this category either diseases in which apoptosis is insufficient (e.g. autoimmune lymphoprolifera-tive syndrome) or enhanced (e.g. SCID) are included. In other pri-mary immunodeficiencies, such as Common Variable Immunodefi-ciency and Chromosome 22q11.2 deletion (del22q11.2) syndrome (DiGeorge syndrome/velocardiofacial syndrome) abnormalities of lymphocytes are likely to be an epiphenomenon of the ongoing immune activation operating in these disease.

Autoimmune Lymphoproliferative Syndrome (ALPS)

ALPS is one of the first human inherited disorders of apoptosis to be described and represents a failure of apoptotic mechanisms that help maintain normal lymphocyte homeostasis. It is character-ized by chronic, nonmalignant lymphadenopathy and splenome- galy, autoimmune manifestations, hypergammaglobulinemia, and expansion of circulating T cell receptor (TCR) /

+ CD4

-CD8

- T

258 Current Pharmaceutical Design, 2008, Vol. 14, No. 3 Giovannetti et al.

cells [98-100]. ALPS generally associates with mutations of the Fas encoding gene (ALPS Ia) and rarely with mutations of FasL (ALPS Ib) [101-105], caspase-8 [106] or caspase-10 [107] gene (ALPS II). A number of ALPS cases lacking in any detectable molecular de-fects have been also described (ALPS III) [108]. In mice, the lpr and gld alleles cause similar diseases due to homozygous recessive mutations in the Fas and FasL gene, respectively [109-111]. Defec-tive apoptosis is the hallmark of peripheral blood lymphocytes of patients with ALPS [101]. Once activated, cultured T cells from ALPS patients exhibit reduced rates of apoptosis when stimulated through the T cell receptor CD3 or through the Fas pathway. ALPS patient B-cell lines transformed with Epstein-Barr virus show the same defective apoptosis after anti-Fas exposure, indicating that both B and T lymphocytes express the defect in Fas-mediated pro-grammed cell death [112]. In addition, mutation bearing family members, even those with no clinical abnormalities and no eleva-tions of CD4

-/CD8

- T cells, demonstrate defective apoptosis in vi-

tro. Thus, the impairment in cellular apoptosis associated with Fas mutation is inherited as a dominant trait.

The Fas/FasL pathway has a key role in the control of periph-eral tolerance and this accounts for the presence of autoimmunity in most ALPS patients as in Fas-deficient mice. Chronically stimu-lated T cells down-modulate CD28, a feature observed also in T lymphocytes of aged subjects [113], and therefore become more vulnerable to FasL-induced cell death according to either a suicide or a killing mechanism [114]. Fas-deficient T cells escape cell death, proliferate and can activate Fas-deficient B cells. In physio-logical conditions, FasL-expressing T cells kill autoimmune B-cell clones, likely because chronic exposure to antigens no longer in-duce protecting signals from cell death. Fas-deficient B cells will thus escape this regulatory process and further proliferate. CD40L/ CD40 T/B interaction can induce class-switch and further affinity maturation of immunoglobulins with autoimmune specificity [115].

Development of lymphoma has also been reported in patients with ALPS and their relatives [116-118]. Deregulated expression of Fas pathway may contribute to tumorigenesis and tumor escape from endogenous growth control. Fresh tumor cells and tumor cell lines, in fact, are often resistant to Fas-induced apoptosis, because they down-regulate Fas expression, secrete soluble forms of Fas, or express a Fas molecule with decreased signalling activity [119-123].

Indications for treatment of ALPS patients depend on the type and severity of symptoms [124]. Hypersplenism, anaemia, and/or thrombocytopenia may require therapeutic intervention. Glucocor-ticoids, intravenous immunoglobulin or cytotoxic drugs have been used with no or only transient effect. Van der Werff ten Bosch et al [125] reported reversion of ALPS with pyrimethamine (2;4-diamino-5-p-chlorophenyl-6-ethyl-pyrimidine), a folic acid antago-nist, suggesting a pro-apoptotic activity of this molecule. More recently, our group demonstrated that pyrimethamine, used at phar- macologically relevant concentration, induced per se apoptosis of activated lymphocytes via the activation of the caspase-8- and cas- pase-10-dependent cascade and subsequent mitochondrial depolari-zation, independently from CD95/Fas engagement [126] (Fig. (2)).

An ALPS-like clinical pattern (named autoimmune lymphopro-liferative disease [ALD]) has also been described by Dianzani et al [127] in patients with decreased Fas function, but no Fas gene mu-tation and peripheral expansion of CD4-

/CD8- T cells. ALD patients

display decreased apoptotic response to ceramide, triggering a death pathway partially overlapping the one used by Fas, which suggests that ALD is caused by downstream alterations of the Fas signalling pathway. Also this defect predisposes to the development of auto-immune diseases and cancer [128].

SCID

Mutations that impair survival signals through cytokine recep-tors can also lead to excessive cell death, resulting in SCID. The

common cytokine receptor -chain ( c) is a component of the recep-tors for different cytokines (IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21) [129]. Their actions are simultaneously inactivated both in X-linked SCID [33,130], which results from mutations in the gene encoding c, and in JAK3-deficient SCID [35,131], because JAK3 associates with c. In humans, these forms of SCID are character-ized by a profound reduction of T cells but normal or increased B cell number [132]. T cell development also fails to occur in the absence of the chain of the interleukin-7 receptor (IL-7R ) [133]. Based on the association of c with IL-7R and the similar pheno-type caused by these deficiencies, SCID caused by c or Jak3-deficiency is thought to be the result of a requirement for IL-7 in lymphocyte development and homeostasis. The mechanism by which IL-7 performs these developmental and homeostatic regula-tions is uncertain, but the ability of IL-7 to promote expression of Bcl-2 and cell survival has been suggested to be a critical aspect of IL-7 activity. In fact, transgenic expression of Bcl-2 can partially rescue T cell development in IL-7R -deficient mice [134]. More recently, Rathmell et al [135] also demonstrated that the addition of IL-7 to tissue culture supernatants protects purified T cells from both death by neglect and cellular atrophy. This effect was not unique to IL-7, but also occurred with c cytokines IL-2, IL-4 and IL-15 as well as the gp130 cytokine IL-6. It has been suggested that the trophic effect of IL-7 is not caused by forcing T cells to enter the cell cycle but is rather the result of preventing the decline in glycolytic and respiratory rates that occurs in neglected cells.

Fig. (2). Pathway of pyrimethamine-induced apoptosis in activated periph-

eral blood lymphocytes. Activated lymphocytes are vulnerable to the

“physiological” signal represented by the triggering of CD95/Fas with the

consequent activation of caspases 8 and 10. Similarly, pyrimethamine in-

duces apoptosis via a mechanism bringing into play the upstream caspases 8

and 10, although it does not require CD95/Fas engagement. Pyrimethamine-

mediated apoptosis is significantly inhibited by caspase-8 (Z-IETD-fmk), -

10 (Z-AEVD-fmk), and -3 (Z-DEVD-fmk) inhibitors. A direct activity on

mitochondria can also be taken into account. In fact, pyrimethamine leads to

the loss of mitochondrial membrane polarization ( ) and cytochrome c

release from mitochondria, that are considered hallmarks of apoptosis-

associated mitochondrial modification. In addition, the expression of BCL-

2, a negative regulator of mitochondrial apoptotic activity, is down-

regulated by pyrimethamine.

Apoptosis in the Homeostasis of the Immune System and in Human Current Pharmaceutical Design, 2008, Vol. 14, No. 3 259

Common Variable Immunodeficiency (CVID)

CVID is an heterogeneous disorder characterized by impaired secretion of immunoglobulins, reduced antibody production, and recurrent infections. Subgroups of patients have increased risks for splenomegaly, granulomatous lesions, autoimmune diseases and malignancies [136]. Despite intense investigation, the aetiology as well as the pathogenesis of CVID, years after its initial recognition, remain largely unknown. Recently, it has been demonstrated that mutations of ICOS [137], TACI [138, 139] and CD19 [140] can result in CVID, although these defects account for only 5 to 10 % of the cases. A major obstacle in characterizing the molecular events leading to CVID is certainly the great heterogeneity of the disease. However, increasing data suggest that there is an underly-ing T cell disorder in most cases. T cells of CVID patients display several features that can be mimicked by a prolonged stimulation in vitro, including the predominance of a memory phenotype, an up-regulation of activation markers, a restriction of TCR variable (BV) chain repertoires, and an increased apoptosis [141-148]. Various reports suggest that the increased susceptibility to apopto-sis, seen in lymphocytes from CVID patients, could be one of the mechanisms implicated in the significant lymphopenia present in CVID and mainly affecting the CD4

+ naïve subset [142, 143, 145,

148]. A positive role of the Fas/FasL system in the increased T cell apoptosis has been suggested by some studies [142, 143, 148]. However, in one study [145] no differences between Fas and FasL expression on CVID and normal T cells were detected. These con-tradictory results may possibly be due to the different phenotypic and clinical characteristics of the patients studied. Although T cells from CVID patients show a persistent immune activation in vivo, attempts to induce Fas-mediated apoptosis, in the presence or ab-sence of activation with mitogenic stimuli, were unsuccessful [143,149]. The impossibility of inducing Fas-mediated apoptosis was hypothesized to be related to the increased serum levels of soluble Fas resulting in the neutralization of the antibody to Fas and/or to the prominent type 1 cytokine profile of CVID patients which was shown to have a protective role in preventing apoptosis [143].

Although most CVID patients show reduced IL-2 production [150], which plays an important role in preventing cell death up-regulating antiapoptotic Bcl-2 family proteins [151], no abnormali-ties in Bcl-2, bax and bcl-xl expression or in the bax/Bcl-2 ratio were demonstrated in both CD4

+ and CD8

+ CVID T cells [143,

145]. Defects of the CD28 receptor pathway, leading to IL-2 depri-vation, have been related, by Di Renzo et al [144], to increased apoptosis in CVID. Costimulation of the CD28 receptor which normally provides, following engagement of the TCR/CD3 recep-tor, a second signal necessary for IL-2 secretion, in CVID did not significantly increase IL-2 production, [152]. Several reports have also shown an activation of the TNF- system in CVID [153, 154]. TNF- is a well known inducer of apoptosis which exerts its effects by binding to two cell surface receptors: TNF-RI and TNF-RII [155]. Di Renzo et al. [145] showed that both TNF-RI and TNF-RII mRNA expressions were significantly increased on CVID CD4

+ T

cells and suggested that this overexpression could contribute to the accelerated T cell death enhancing T cell suceptibility to increased and/or normal levels of circulating TNF- . Increased serum level of TNF- accompanied by glutathione depletion in CD4

+ lympho-

cytes, particularly in the naive subset, have also been reported by Aukrust et al. [156]. Defective glutathione levels were more pro-nounced in those CVID patients characterized by chronic immune activation, low numbers of CD4

+ lymphocytes in peripheral blood

(< 400 cells/μl) and splenomegaly. It has been found that TNF- stimulation of T cell lines and endothelial cells decreases total glu-tathione levels in these cells by causing enhanced ROS generation, which in turn leads to the consumption of reduced glutathione [157, 158]. Thus, increased TNF- activation, may, at least, be responsi-ble for the glutathione depletion and increased levels of oxidative

stress [156]. On the other hand, increased oxidative stress has been related to enhanced apoptosis [159].

Recently, we performed in 60 CVID patients a wide panel of immunological investigations aiming specifically at a quantitative and qualitative evaluation of the T cell compartment [148]. We demonstrated that the vast majority of CVID patients presented multiple T cell abnormalities intimately related among them, the severity of which reflected in a parallel loss of CD4

+ naive T cells.

A strong correlation between the number of CD4+ naive T cells and

clinical features was observed, supporting the subgrouping of pa-tients according to their number of naive CD4

+ T lymphocytes. A

reduced thymic output and disrupted CD4+ and CD8

+ TCR reper-

toires paralleled the contraction of CD4+ naive T cell pools. The

evaluation of activation markers (i.e. CD95 and HLA-DR mole-cules) and cytokine production (IFN- ) indicated a strong T cell activation that was significantly related to the increased levels of T cell turnover and apoptosis.

In our opinion, CVID represents a model of primary immu-nodeficiency in which, similarly to that observed in chronic viral infections as HIV-infection, prolonged T cell activation, followed by an increased apoptosis results in a progressive exhaustion of the renewal capability of the thymus. Attempting to maintain a normal cell number, peripheral naive T cells intensely proliferate, but to the detriment of the diversity of T cell repertoires. A restricted TCR repertoire might result, in turn, in a decreased ability to counteract infections or cancer. The profound reduction of the CD4 subset, detected in CVID by our and other groups, as compared to the CD8 one, may be due to differences in their regenerative pathway [160]. Differently from the reconstitution of CD8

+ T cells which is rapid

and occurring by peripheral expansion, that of CD4+ T cells is

slower and depends on thymic output [161,162]. Furthermore, the regenerative capability of the CD4

+ T cell compartment is also re-

duced by the proneness of CD4+ T cells to undergo apoptosis

[163,164].

Current therapy for CVID includes regular infusions of intrave-nous immunoglobulin and antibiotics as needed, but significant morbidity and mortality remain [136]. Therapeutic strategies capa-ble of hindering apoptosis of T cells could help in reversing the unbalanced lymphocyte homeostasis, thus leading to an ameliora-tion of clinical status. With regard to this, Cunningham-Rundles et al demonstrated that IL-2 therapy was able to enhance T cell func-tions and reverse T cell anergy in most CVID patients [165-167]. They suggested that CVID patients who might be likely to benefit from this therapy are those patients with defective T helper function as a central abnormality, in which treatment with IL-2 may augment antibody responses as well.

Chromosome 22q11.2 Deletion Syndrome

Chromosome 22q11.2 deletion syndrome (DiGeorge syn-drome/velocardiofacial syndrome) is a disorder classically associ-ated with congenital cardiac defects, immune deficiencies secon-dary to aplasia or hypoplasia of the thymus, and hypocalcemia due to small or absent parathyroid glands [168]. The thymic hypoplasia is seen in more than 80% of the patients with the deletion regardless of the other clinical manifestations of the syndrome. The spectrum of immunodeficiency ranges from absent T cells due to thymic aplasia to normal or near-normal T cell numbers [169-174]. In gen-eral, the T cell deficit improves with age, and many patients pro-duce normal numbers of T cells by the end of the first year of life. T cell function is typically preserved, although standard mitogen pro-liferation assays are usually diminished when the T cell count is very low, corresponding to the diminished numbers of cells compe-tent to respond to the stimulus [171]. Immunoglobulin A (IgA) deficiency, hypogammaglobulinemia, and defects in functional antibody production have occasionally been described and are thought to be secondary to the T cell defect [175-179]. The immu-nodeficiency arises as a consequence of the thymic hypoplasia;

260 Current Pharmaceutical Design, 2008, Vol. 14, No. 3 Giovannetti et al.

residual thymic tissue is comprised of normal thymic components and the orderly sequence of maturation appears to be preserved allowing T cell differentiation [168]. No significantly altered ex-pression of Fas, Bcl-2 and p53 was also detected in hypoplastic thymus [180]. However, our group observed a reduction of both spontaneous and CD3- or Fas-mediated apoptosis in lymphoid cells from thymic tissue of two patients with chromosome 22q11.2 dele-tion syndrome as compared in comparison with a normal thymus [181]. Differently, in peripheral blood Gupta et al [182] observed high levels of apoptosis of both CD4

+ and CD8

+ T cells and in-

creased Fas and FasL expression, suggesting that spontaneous apoptosis in peripheral T lymphocytes, at least in part, may be re-sponsible for T cell deficiency, mainly affecting the CD4 subset. The predominance of a memory phenotype, a peripheral homeo-static proliferation, and a restriction of both CD4

+ and CD8

+

TCRBV repertoires have been demonstrated by our and other groups [183,184]. Also in this case, the increased activation occur-ring in vivo could be predominantly responsible for the enhanced apoptosis.

APOPTOTIC MECHANISMS IN AUTOIMMUNITY DE-

VELOPMENT

Mechanisms which control apoptosis induction and regulation play a key role in the balance between cell proliferation and death [91]. Imbalance of these mechanisms may contribute to the devel-opment of autoimmune diseases as defective apoptosis of immune cells leads to dysregulated autoreactive cell proliferation [109,185].

Several factors are involved in the induction of resistance to apoptosis as follows: 1) interactions among soluble or membrane molecules and their ligands (i.e. Fas/Fas-ligand, CD40/CD40-ligand, CD80(B7.1)-CD86(B7.2)/CTLA-4; TNF-alfa/TNF-R); 2) the expression of anti-apoptotic proteins (Bcl-2, Bcl-XL and “in-hibitory receptors” like CD94/NKG2A and KIR2D); 3) modifica-tions of death-inducing signal complex (DISC) proetins like FADD, TRADD and RAIDD; 4) defects in caspases activation [186-190]. In the present chapter we briefly summarize recent researches deal-ing with the dysregulation of apoptotic mechanisms in systemic and organ-specific autoimmune diseases.

Apoptosis in Rheumatoid Arthritis

In rheumatoid arthritis circulating inflammatory cells and lo-cally proliferating fibroblasts, lymphocytes and endothelial cells infiltrate the inflamed synovium. The dysregulated cell proliferation is attributable to defects in apoptosis activation which are mediated by cytokines, immunecomplexes, toll-like receptors ligands and hormones (estrogens and their metabolites) detectable in the micro-environment as well as by CD40/CD40L cell-cell interactions [191,192]. In monocytes/macrophages of rheumatoid synovium Fas-dependent apoptotic pathways are blocked by FLIP, which inhibits caspase 8 activation, whereas NF-kB and PI3K/AKT-1 are activated leading to Bcl-2 upregulation, mitochondrial membrane stabilization and apoptosis inhibition. The inhibitors of procaspase 3-caspase 3 pathway X-IAP and p21/Waf-1are also involved in the dysregulation of monocyte/macrophage survival [193-195]. Ep-stein-Barr virus (EBV) may infect neutrophils and macrophages triggering their apoptotic death and decreasing their phagocytic activity. EBV infection stimulates TNF-alpha secretion which in-creases CD34+ cells apoptosis but decreases B cell apoptosis and the development of autoreactive cells [196-198]. Several autoanti-bodies are detectable in rheumatoid arthritis serum including anti-fillagrin antibodies. Filaggrin is a protein involved in the aggrega-tion of cytokeratin filaments and is synthesized in epithelial cells as a phosphorylated precursor. During differentiation of epithelial cells, the precursor is dephosphorylated, cleaved and finally deimi-nated. Anti-fillagrin antibodies do not react with the precursor molecule but only with deiminated filaggrin. Deimination is a post translational modification involving the conversion of arginine

residues into citrulline residues. The enzyme responsible for this modification, peptidylarginine deiminase (PAD), has been detected in a wide number of mammalian tissues and four isoforms of the enzyme have been identified. PAD V isoform has been localized in precursors of macrophages and neutrophils found in inflamed sino-via. PAD is a Ca-dependent enzyme inactive at normal intracellular Ca concentrations. Calcium ionophores that trigger apoptosis of various cells also activate the enzyme and induce protein deimina-tion [199].

Apoptosis in Systemic Autoimmunity: Connective Tissue Dis-eases

In systemic sclerosis (scleroderma), although the number of cutaneous fibroblasts is in the normal range their rate of collagen production is strongly increased under the influence of cytokines produced by leukocytes that infiltrate the skin and of the autocrine production of soluble factors including transforming growth factor-beta (TGF-beta) [200]. The signal transduction program down-stream TGF-beta is activated, as a result of elevated levels of its receptor and/or of the deregulation of the Smad3 factor which is responsible of the signals induced by TGF-beta and controls the ability of TGF-beta to induce fibrosis in vivo [201]. The acquired resistance to programmed cell death of the sclerodermic fibroblasts may contribute to the maintenance of the signalling and of the acti-vated phenotype. In parallel, the recognition of apoptotic cells in-duces per se the production of TGF-beta by cells of the innate im-mune system [202]. Defects in the clearance of apoptotic cells may contribute to the autoantigen spreading and to chronic inflammation and to immune-mediated tissue damage

Apoptotic phenomena play a key role also in the development of systemic lupus erythematosus (SLE). An increased production of apoptotic bodies along with a strong reduction of serum DNase activity which is involved in their clearance has been detected in SLE patients. However, a detailed analysis of DNase gene mutations in SLE is lacking and it is unknown whether these mutations may represent a diagnostic marker for SLE development. Moreover, phagocytosis of anti- GPI-antibody coated apoptotic cells by dendritic cells, followed by the presentation of their antigenic peptides to autoreactive T lymphocytes, may contribute to the maintainance of autoimmune responses in SLE [203,204]. At present, it is not known if these new autoantigens can be utilized as diagnostic tools in SLE patients.

Apoptosis and Organ-Specific Autoimmunity

As far as organ-specific autoimmune diseases are concerned, dysregulation of Fas and Fas-L expression on thyroid cells and in infiltrating lymphocytes has been reported. Moreover, the associa-tion between Graves’ disease and CTLA-4 polymorphisms has been described and it has been hypothesized that CTLA-4 alleles may contribute to the severity of thyrotoxicosis, the development of ophthalmopathy, the risk of relapse following anti-thyroid drugs and the presence other co-existing autoimmune disorders [205]. Finally, elevated serum levels of soluble CTLA-4 (sCTLA-4) have been detected in patients with autoimmune thyroid diseases and it has been proposed that sCTLA-4 may compete with membrane CTLA-4 for CD80/CD86 binding leading to a decrease of the in-hibitory signal [206]. These findings suggest the existence of poten-tial correlations between autoimmune and lymphoproliferative dis-eases also taking into account the elevated risk for the development of low grade lymphomas in patients with coeliac disease and Sjögren syndrome.

APOPTOSIS IN GASTROINTESTINAL IMMUNO-

MEDIATED DISEASES

In the GI tract, as well as all other organs, the balance between cell proliferation and cell death plays a critical role in local physiol-ogy and pathology. Cell death is regulated by a highly controlled

Apoptosis in the Homeostasis of the Immune System and in Human Current Pharmaceutical Design, 2008, Vol. 14, No. 3 261

process, known as apoptosis, which also modulates immune and inflammatory responses by inducing leukocyte activation and sur-vival, limiting clonal expansion and cytokine production. Insuffi-cient growth factor production or deprivation induces T cell apopto-sis and the entire mechanism is regulated by a fine balance of dif-ferent molecules acting as anti- and pro-apoptotic factors. As a consequence, diseases may be induced by either a reduction or an increase of apoptosis. Among GI diseases with an immuno-mediated pathogenesis, IBD such as CrD and UC are examples of diseases induced by abnormally decreased apoptosis. On the other hand, increased apoptosis has a role in the pathogenesis of coeliac disease and associated diseases.

Direct and Indirect Mechanisms Leading to Decreased Apopto-sis are Associated with the Pathogenesis of IBD

Apoptosis plays a crucial role in IBD. Immune responses in the mucosa are frequently characterized by major expansions of anti-gen-specific T cells that have potent effector functions. Although this is important for host defense, it might also lead to effector cell populations with substantial autoreactivity and the potential to cause mucosal inflammation. The mucosal immune system has developed several strategies for the control of mucosal immune responses. Among these is the regulation of apoptosis that either occurs via an active mechanism involving death receptors such as Fas or TNFR stimulation or via a passive mechanism such as fol-lowing lymphokine withdrawal.

Direct (Fas-Mediated) Apoptosis

Most LP CD4 T cells express memory cell markers (CD45R0) and constitutively high levels of Fas. In contrast to LP T cells from control patients, those from patients with IBD exhibit defective Fas-induced apoptosis upon stimulation via CD2. This led to the sug-gestion that lamina propria T cells are in a partially anergic state. This defect in inflamed CrD tissue is accompanied by elevated Bcl-2 levels [207]. In addition, normal levels of Fas and Fas ligand are expressed thus suggesting that failure of CrD LP cells to respond to Fas induced apoptosis may be secondary to changes in the expres-sion and/or activity of intracellular molecules which participate in Fas mediated programmed cell death. For instance, Flip (Flice in-hibitor protein), acts conferring resistance to Fas mediated apopto-sis of both CrD and UC lamina propria lymphocytes [208]. Al-though LP T cells in IBD express the same amount of Fas on their surface, they are less sensitive to Fas-mediated apoptosis than con-trol cells. However, resistance of LP T cells against apoptosis in CrD is not restricted to the Fas–FasL system, as recent data show that CrD T cells grow more rapidly in response to IL-2 than do control cells and are significantly more resistant to apoptosis in-duced by IL-2 deprivation. These abnormalities are associated with an imbalance of Bcl-2 positive/ Bax positive mucosal cells. Insuffi-cient T cell apoptosis may interfere with clonal deletion, mainte-nance of tolerance and result in inappropriate T cell accumulation contributing to chronic inflammation [209].

Moreover, recent studies have shown that decoy receptors DcR1, DcR2 and DcR3, that compete with CD95 are highly ex-pressed in T cells from inflamed mucosa in UC patients [210].

In addition, apoptosis related genes such as Bcl-2 antago-nist/killer, B lymphoid tyrosine kinase, caspase 14, Harakiri (a proapoptotic member of the Bcl-2 family of proteins), TNF receptor 2, and TRAF1, were highly expressed in epithelial cells isolated from patients with UC [211].

This broad resistance of T cells to apoptosis involving mito-chondrial activity in inflamed tissue occurs because of increased levels of Bcl-2 [212]. Overall, the picture that emerges is that, in the uninflamed gut, T cells manifest increased spontaneous or activa-tion-induced apoptosis mediated by the Fas pathway that places a limit on the expansion of T cells following direct and bystander

stimulation by specific antigen. By contrast, in inflamed tissue, T cells resistant to apoptosis, exhibit prolonged survival and increased inflammatory cytokines production [213].

Indirect (Cytokine-Mediated) Apoptosis

An important role in regulation of apoptosis has been related to cytokines which can stimulate or down-regulate apoptotic mecha-nisms.

IL-23 is essential for T cell-mediated colitis and promotes in-flammation via IL-7 and IL-6 [214]. A critical target of IL-23 is a unique subset of tissue-homing memory T cells, which are specifi-cally activated by IL-23 to produce the proinflammatory mediators IL-17 and IL-6. This pathway may be responsible for chronic intes-tinal inflammation. IL-23 shares the same p40 subunit with IL-12 and anti-p40 antibodies neutralize both IL-12 and IL-23, reversing intestinal inflammation in both humans and animal models [214].

It has been reported that IL-10 is important in maintaining im-mune homeostasis in the gut, demonstrated by the fact that IL-10 knockout (KO) mice spontaneously develop colitis [215]. In pa-tients with CrD, a decreased IL-10 concentration in the ileum pre-dicted a higher risk of disease recurrence [216]. Systemic IL-10 treatment or IL-10 gene transfer has been successfully used in sev-eral animal models of inflammatory bowel disease, such as IL-10 KO mice and severe combined immunodeficiency transfer colitis [217].

A pathway of T cell activation driven by IL-6/IL-6R contributes to the perpetuation of chronic intestinal inflammation. Specific targeting of this pathway may be a promising new approach for the treatment of CrD [212]. Novel therapeutic strategies aim at restor-ing mucosal T cell susceptibility to apoptosis through targeting of signal transduction pathways that are crucial for augmented resis-tance of T lymphocytes against apoptosis. For instance, a newly developed humanized anti-IL-6R monoclonal antibody that induces intestinal T cell apoptosis showed clinical efficacy in patients with active CrD [218].

IL-10 can block apoptosis of intestinal epithelial cells by inhib-iting COX-2 production [219]. In keeping with these data gene polymorphisms associated with reduced production of IL-10 have been reported as risk factors for developing IBD [220]. Also inhibi-tors of IL-23 and IL-12 are able to reverse colitis [221] and anti-interleukin 12 treatment induces Fas mediated apoptosis of Th1 cells in experimentally induced colitis in mice [222].

Increased Apoptosis is Associated with the Pathogenesis of Coeliac Disease

In contrast to IBD, where the pathogenesis has been associated to a failure in feedback apoptotic mechanisms, as discussed above, increased apoptosis may be linked to the pathogenesis of Coeliac disease (OMIM 212750). This is a gluten-dependent entheropathy occurring in genetically predisposed individuals and resulting from inflammatory injury to the mucosa of small intestine associated to diverse clinical manifestations whose frequency has been reported to be very high in Caucasians. Coeliac disease is associated with a variety of other diseases which share common genetic susceptibility and pathogenetic mechanisms. For instance, dermatitis Herpeti-formis (DH) is very often associated with coeliac disease. In DH, dermal Bax and Bcl-2 proteins are increased in the dermal perivas-cular compartment and apoptosis plays a major role in the patho-genesis of cutaneous lesions [223]. Development of coeliac disease is strongly linked to genetic factors both HLA related [224] and non-HLA related [225]. The effector phases of small bowel tissue destruction in coeliac disease appear to be mediated by cytotoxic T and NK cells through both apoptosis mediated tissue damage and release of proteolytic enzymes such as perforins. Apoptosis is me-diated through ligation of FasL expressed on gliadin-activated T-cells.

262 Current Pharmaceutical Design, 2008, Vol. 14, No. 3 Giovannetti et al.

In untreated coeliac disease, FasL and perforin expression by IEL is increased, and correlates with the level of enterocyte apopto-sis [226]. This phenomenon also occurs at the peripheral level and is possibly responsible for lymphopenia, exposure of membrane phospholipids and subsequent production of autoantibodies [227].

IEL of CD3 negative cells expressing NK markers such as CD16 and CD161 are found in coeliac disease [228] and at the molecular level both the absolute number and the percentage of lymphocytes expressing perforin mRNA are increased [229]. This expanded NK population may account for the increased levels of perforin in the gut.

Cytokines are also involved in coeliac disease activation-induced cell death. Epithelium-derived IL-15 signalling via IL-15R is critical for the development, activation and survival of IEL [230]. IL-15 can also induce CD94 positive IEL to gamma inter-feron production and FasL-mediated cytotoxicity [231].

These data, although presently limited to biological models, may soon became useful for the clinical benefits of patients. In fact, blocking IL-15 may suppress uncontrolled IEL activation and sur-vival, with the potential to prevent both tissue damage and lym-phomagenesis in coeliac disease [230].

CONCLUSION

In conclusion, the interactions among soluble and membrane molecules as well as the balance between pro-apoptotic and anti-apoptotic factors, both in effector and target cells, seem to represent key mechanisms underlying the development of autoimmune dis-eases. Although immune tolerance seems to be achieved mainly through central selection implying apoptotic mechanisms, some of which have been described in the chapter on thymocyte differentia-tion, this process is by far insufficient to delete autoreactive clones, which may survive in the periphery. If normal regulatory systems fail or homeostatic peripheral expansion, a physiologically normal control of lymphopenia, is a recurrent phenomenon, selective ad-vantage for autoimmune T cell growth occurs. Immunodeficiency states are often characterized by the paradoxical occurrence of auto-immunity, so that a more detailed characterization of these interac-tions is required in order to better define the pathophysiology of autoimmune diseases. Finally, the possibility to interfere with regu-latory mechanisms of apoptosis, aiming either to block or to en-hance it at different time points, represents a promising approach for the development of new immunomodulatory therapies.

ACKNOWLEDGMENTS

Studies reported in this paper have been supported by grants Ricerche di Facoltà 2007 to R.P.

ABBREVIATIONS

AIRE = Autoimmune regulator

ALD = Autoimmune lymphoproliferative disease

ALPS = Autoimmune lymphoproliferative syndrome

APC = Antigen presenting cell

GPI = 2 Glycoprotein-I

COX-2 = Ciclooxygenase-2

CrD = Crohn’s disease

CVID = Common Variable Immunodeficiency

DC = Dendritic cell

DH = Dermatitis herpetiformis

DISC = Death-inducing signal complex

DN = Double negative

DP = Double CD4+CD8+ positive

EBV = Epstein-Barr virus

ETP = Early thymic precursor

FADD = Fas associated death domain

FasL = Fas ligand

FLIP = FLICE inhibitory protein

c = Common gamma chain

GI = Gastrointestinal

HIV = Human immunodeficiency virus

HLA = Human leukocyte antigens (histocompatibility complex)

IBD = Inflammatory bowel disease

ICAM-1 = Intercellular adhesion molecule-1

ICOS = Inducible co-stimulator

IEL = Intraepithelial lymphocytes

IFN = Interferon

IL = Interleukin-

ISP = Immature single positive

JAK = Janus kinase

KO = knoçck out

LP = Lamina propria

MCL-1 = Myeloid cell leukemia sequence 1

MHC = Major histocompatibility complex

NFAT = Nuclear factor of activated T cells

NF- B = Nuclear factor - B

NK = Natural killer

PAD = Peptidylarginine deiminase

SCID = Severe combined immunodeficiency

SLE = Systemic lupus erythematosus

TACI = Transmembrane activator and CAML interactor

TCR = T cell receptor

TEC = Thymic epithelial cells

TGF = Transforming growth factor-beta

TNF = Tumor necrosis factor

TP = Thymic precursor

TRAIL = TNF-related apoptosis-inducing ligand

Treg = T regulatory cells

TSA = Tissue specific antigens

UC = Ulcerative colitis

VCAM-1 = Vascular cell adhesion molecule-1

REFERENCES

References 232-234 are related articles recently published in Cur-rent Pharmaceutical Design.

[1] Krammer PH. CD95's deadly mission in the immune system. Na-

ture 2000; 407: 789-95.

[2] Kyewski B, Derbinski J. Self-representation in the thymus: an

extended view. Nat Rev Immunol 2004; 4: 688-98.

[3] Rizzi M, Ferrera F, Filaci G, Indiveri F. Disruption of immunologi-

cal tolerance: role of AIRE gene in autoimmunity. Autoimmun Rev

2006; 5: 145-7.

[4] Marrack P, Bender J, Hildeman D, Jordan M, Mitchell T, Mura-

kami M, et al. Homeostasis of alpha beta TCR+ T cells. Nat Im-

munol 2000; 1: 107-11.

[5] King C, Ilic A, Koelsch K, Sarvetnick N. Homeostatic expansion of

T cells during immune insufficiency generates autoimmunity. Cell

2004; 117: 265-77.

[6] Van Parijs L, Abbas AK. Homeostasis and self-tolerance in the

immune system: turning lymphocytes off. Science 1998; 280: 243-

8.

Apoptosis in the Homeostasis of the Immune System and in Human Current Pharmaceutical Design, 2008, Vol. 14, No. 3 263

[7] Douek DC, McFarland RD, Keiser PH, Gage EA, Massey JM,

Haynes BF, et al. Changes in thymic function with age and during

the treatment of HIV infection. Nature 1998; 396: 690-5.

[8] Munier ML, Kelleher AD. Acutely dysregulated, chronically dis-

abled by the enemy within: T-cell responses to HIV-1 infection.

Immunol Cell Biol 2007; 85: 6-15.

[9] Krupica T, Jr., Fry TJ, Mackall CL. Autoimmunity during lym-

phopenia: a two-hit model. Clin Immunol 2006; 120: 121-8.

[10] Sharpe AH, Abbas AK. T-cell costimulation--biology, therapeutic

potential, and challenges. N Engl J Med 2006; 355: 973-5.

[11] Murali-Krishna K, Ahmed R. Cutting edge: naive T cells masquer-

ading as memory cells. J Immunol 2000; 165: 1733-7.

[12] Baccala R, Theofilopoulos AN. The new paradigm of T-cell ho-

meostatic proliferation-induced autoimmunity. Trends Immunol

2005; 26: 5-8.

[13] Gleeson PA, Toh BH, van Driel IR. Organ-specific autoimmunity

induced by lymphopenia. Immunol Rev 1996; 149: 97-125.

[14] Vercelli D. Mechanisms of the hygiene hypothesis--molecular and

otherwise. Curr Opin Immunol 2006; 18: 733-7.

[15] Schaub B, Lauener R, von Mutius E. The many faces of the hy-

giene hypothesis. J Allergy Clin Immunol 2006; 117: 969-77.

[16] Bennett F, Luxenberg D, Ling V, Wang IM, Marquette K, Lowe D,

et al. Program death-1 engagement upon TCR activation has dis-

tinct effects on costimulation and cytokine-driven proliferation: at-

tenuation of ICOS, IL-4, and IL-21, but not CD28, IL-7, and IL-15

responses. J Immunol 2003; 170: 711-8.

[17] Chatenoud L. Protection from autoimmunity: immunological indif-

ference versus T-cell mediated suppression? Eur J Immunol 2006;

36: 2296-8.

[18] O'Shea JJ, Ma A, Lipsky P. Cytokines and autoimmunity. Nat Rev

Immunol 2002; 2: 37-45.

[19] Schimpl A, Berberich I, Kneitz B, Kramer S, Santner-Nanan B,

Wagner S, et al. IL-2 and autoimmune disease. Cytokine Growth

Factor Rev 2002; 13: 369-78.

[20] Ulmanen I, Halonen M, Ilmarinen T, Peltonen L. Monogenic auto-

immune diseases - lessons of self-tolerance. Curr Opin Immunol

2005; 17: 609-15.

[21] Cho BK, Rao VP, Ge Q, Eisen HN, Chen J. Homeostasis-

stimulated proliferation drives naive T cells to differentiate directly

into memory T cells. J Exp Med 2000; 192: 549-56.

[22] Herold KC, Taylor L. Treatment of Type 1 diabetes with anti-CD3

monoclonal antibody: induction of immune regulation? Immunol

Res 2003; 28: 141-50.

[23] Chang JT, Palanivel VR, Kinjyo I, Schambach F, Intlekofer AM,

Banerjee A, et al. Asymmetric T lymphocyte division in the initia-

tion of adaptive immune responses. Science 2007; 315: 1687-91.

[24] Jameson SC. Maintaining the norm: T-cell homeostasis. Nat Rev

Immunol 2002; 2: 547-56.

[25] Arnold R, Brenner D, Becker M, Frey CR, Krammer PH. How T

lymphocytes switch between life and death. Eur J Immunol 2006;

36: 1654-8.

[26] Schluns KS, Lefrancois L. Cytokine control of memory T-cell

development and survival. Nat Rev Immunol 2003; 3: 269-79.

[27] Hogquist KA, Baldwin TA, Jameson SC. Central tolerance: learn-

ing self-control in the thymus. Nat Rev Immunol 2005; 5: 772-82.

[28] Kincade PW, Owen JJ, Igarashi H, Kouro T, Yokota T, Rossi MI.

Nature or nurture? Steady-state lymphocyte formation in adults

does not recapitulate ontogeny. Immunol Rev 2002; 187: 116-25.

[29] Carvalho TL, Mota-Santos T, Cumano A, Demengeot J, Vieira P

Arrested B. lymphopoiesis and persistence of activated B cells in

adult interleukin 7(-/)- mice. J Exp Med 2001; 194: 1141-50.

[30] Harman BC, Jenkinson WE, Parnell SM, Rossi SW, Jenkinson EJ,

Anderson G. T/B lineage choice occurs prior to intrathymic Notch

signalling. Blood 2005; 106: 886-92.

[31] Lepique AP, Palencia S, Irjala H, Petrie HT. Characterization of

vascular adhesion molecules that may facilitate progenitor homing

in the post-natal mouse thymus. Clin Dev Immunol 2003; 10: 27-

33.

[32] Prockop SE, Petrie HT. Regulation of thymus size by competition

for stromal niches among early T cell progenitors. J Immunol 2004;

173: 1604-11.

[33] Noguchi M, Yi H, Rosenblatt HM, Filipovich AH, Adelstein S,

Modi WS, et al. Interleukin-2 receptor gamma chain mutation re-

sults in X-linked severe combined immunodeficiency in humans.

Cell 1993; 73: 147-57.

[34] Giliani S, Mori L, de Saint BG, Le Deist F, Rodriguez-Perez C,

Forino C, et al. Interleukin-7 receptor alpha (IL-7Ralpha) defi-

ciency: cellular and molecular bases. Analysis of clinical, immu-

nological, and molecular features in 16 novel patients. Immunol

Rev 2005; 203: 110-26.

[35] Russell SM, Tayebi N, Nakajima H, Riedy MC, Roberts JL, Aman

MJ, et al. Mutation of Jak3 in a patient with SCID: essential role of

Jak3 in lymphoid development. Science 1995; 270: 797-800.

[36] Sleasman JW, Harville TO, White GB, George JF, Barrett DJ,

Goodenow MM. Arrested rearrangement of TCR V beta genes in

thymocytes from children with X-linked severe combined immu-

nodeficiency disease. J Immunol 1994; 153: 442-8.

[37] Napolitano LA, Grant RM, Deeks SG, Schmidt D, De Rosa SC,

Herzenberg LA, et al. Increased production of IL-7 accompanies

HIV-1-mediated T-cell depletion: implications for T-cell homeo-

stasis. Nat Med 2001; 7: 73-9.

[38] Di Santo JP, Aifantis I, Rosmaraki E, Garcia C, Feinberg J, Fehling

HJ, et al. The common cytokine receptor gamma chain and the pre-

T cell receptor provide independent but critically overlapping sig-

nals in early alpha/beta T cell development. J Exp Med 1999; 189:

563-74.

[39] Opferman JT, Letai A, Beard C, Sorcinelli MD, Ong CC, Kors-

meyer SJ. Development and maintenance of B and T lymphocytes

requires antiapoptotic MCL-1. Nature 2003; 426: 671-6.

[40] Dik WA, Pike-Overzet K, Weerkamp F, de Ridder D, de Haas EF,

Baert MR, et al. New insights on human T cell development by

quantitative T cell receptor gene rearrangement studies and gene

expression profiling. J Exp Med 2005; 201: 1715-23.

[41] Ktorza S, Blanc C, Laurent C, Sarun S, Verpilleux MP, Debre P, et

al. Complete TCR-delta rearrangements and partial (D-J) recombi-

nation of the TCR-beta locus in CD34+7+ precursors from human

cord blood. J Immunol 1996; 156: 4120-7.

[42] Klein F, Feldhahn N, Lee S, Wang H, Ciuffi F, von Elstermann M,

et al. T lymphoid differentiation in human bone marrow. Proc Natl

Acad Sci USA 2003; 100: 6747-52.

[43] Hori T, Cupp J, Wrighton N, Lee F, Spits H. Identification of a

novel human thymocyte subset with a phenotype of CD3- CD4+

CD8 alpha + beta-1. Possible progeny of the CD3- CD4- CD8-

subset. J Immunol 1991; 146: 4078-84.

[44] Klein L, Kyewski B. Self-antigen presentation by thymic stromal

cells: a subtle division of labor. Curr Opin Immunol 2000; 12: 179-

86.

[45] Derbinski J, Schulte A, Kyewski B, Klein L. Promiscuous gene

expression in medullary thymic epithelial cells mirrors the periph-

eral self. Nat Immunol 2001; 2: 1032-9.

[46] Foss DL, Donskoy E, Goldschneider I. The importation of hemato-

genous precursors by the thymus is a gated phenomenon in normal

adult mice. J Exp Med 2001; 193: 365-74.

[47] Petrie HT, Scollay R, Shortman K. Commitment to the T cell re-

ceptor-alpha beta or -gamma delta lineages can occur just prior to

the onset of CD4 and CD8 expression among immature thymo-

cytes. Eur J Immunol 1992; 22: 2185-8.

[48] Kang J, Coles M, Cado D, Raulet DH. The developmental fate of T

cells is critically influenced by TCRgammadelta expression. Im-

munity 1998; 8: 427-38.

[49] Madakamutil LT, Christen U, Lena CJ, Wang-Zhu Y, Attinger A,

Sundarrajan M, et al. CD8alphaalpha-mediated survival and differ-

entiation of CD8 memory T cell precursors. Science 2004; 304:

590-3.

[50] Pennington DJ, Silva-Santos B, Hayday AC. Gammadelta T cell

development--having the strength to get there. Curr Opin Immunol

2005; 17: 108-15.

[51] Hayes SM, Shores EW, Love PE. An architectural perspective on

signalling by the pre-, alphabeta and gammadelta T cell receptors.

Immunol Rev 2003; 191: 28-37.

[52] Blom B, Verschuren MC, Heemskerk MH, Bakker AQ, Gastel-Mol

EJ, Wolvers-Tettero IL, et al. TCR gene rearrangements and ex-

pression of the pre-T cell receptor complex during human T-cell

differentiation. Blood 1999; 93: 3033-43.

[53] Trigueros C, Ramiro AR, Carrasco YR, de Yebenes VG, Albar JP,

Toribio ML. Identification of a late stage of small noncycling

264 Current Pharmaceutical Design, 2008, Vol. 14, No. 3 Giovannetti et al.

pTalpha- pre-T cells as immediate precursors of T cell receptor al-

pha/beta+ thymocytes. J Exp Med 1998; 188: 1401-12.

[54] Aifantis I, Gounari F, Scorrano L, Borowski C, von Boehmer H.

Constitutive pre-TCR signalling promotes differentiation through

Ca2+ mobilization and activation of NF-kappaB and NFAT. Nat

Immunol 2001; 2: 403-9.

[55] Gounari F, Aifantis I, Khazaie K, Hoeflinger S, Harada N, Taketo

MM, et al. Somatic activation of beta-catenin bypasses pre-TCR

signalling and TCR selection in thymocyte development. Nat Im-

munol 2001; 2: 863-9.

[56] Goux D, Coudert JD, Maurice D, Scarpellino L, Jeannet G, Piccolo

S, et al. Cooperating pre-T-cell receptor and TCF-1-dependent sig-

nals ensure thymocyte survival. Blood 2005; 106: 1726-33.

[57] Aifantis I, Borowski C, Gounari F, Lacorazza HD, Nikolich-Zugich

J, von Boehmer H. A critical role for the cytoplasmic tail of pTal-

pha in T lymphocyte development. Nat Immunol 2002; 3: 483-8.

[58] Khaled AR, Durum SK. The role of cytokines in lymphocyte ho-

meostasis. Biotechniques 2002; Suppl: 40-5.

[59] Peter ME, Krammer PH. The CD95(APO-1/Fas) DISC and be-

yond. Cell Death Differ 2003; 10: 26-35.

[60] Newton K, Harris AW, Strasser A. FADD/MORT1 regulates the

pre-TCR checkpoint and can function as a tumour suppressor.

EMBO J 2000; 19: 931-41.

[61] Guidos CJ, Williams CJ, Grandal I, Knowles G, Huang MT, Dan-

ska JS. V(D)J recombination activates a p53-dependent DNA dam-

age checkpoint in scid lymphocyte precursors. Genes Dev 1996;

10: 2038-54.

[62] Hatakeyama S, Hamasaki A, Negishi I, Loh DY, Sendo F, Naka-

yama K, et al. Multiple gene duplication and expression of mouse

bcl-2-related genes, A1. Int Immunol 1998; 10: 631-7.

[63] Tomayko MM, Punt JA, Bolcavage JM, Levy SL, Allman DM,

Cancro MP. Expression of the Bcl-2 family member A1 is devel-

opmentally regulated in T cells. Int Immunol 1999; 11: 1753-61.

[64] Sedlak TW, Oltvai ZN, Yang E, Wang K, Boise LH, Thompson

CB, et al. Multiple Bcl-2 family members demonstrate selective

dimerizations with Bax. Proc Natl Acad Sci USA 1995; 92: 7834-8.

[65] Werner AB, de Vries E, Tait SW, Bontjer I, Borst J. Bcl-2 family

member Bfl-1/A1 sequesters truncated bid to inhibit is collabora-

tion with pro-apoptotic Bak or Bax. J Biol Chem 2002; 277: 22781-

8.

[66] Mandal M, Borowski C, Palomero T, Ferrando AA, Oberdoerffer

P, Meng F, et al. The BCL2A1 gene as a pre-T cell receptor-

induced regulator of thymocyte survival. J Exp Med 2005; 201:

603-14.

[67] Starr TK, Jameson SC, Hogquist KA. Positive and negative selec-

tion of T cells. Annu Rev Immunol 2003; 21: 139-76.

[68] Palmer E. Negative selection--clearing out the bad apples from the

T-cell repertoire. Nat Rev Immunol 2003; 3: 383-91.

[69] Mathis D, Benoist C. Back to central tolerance. Immunity 2004;

20: 509-16.

[70] Villasenor J, Benoist C, Mathis D. AIRE and APECED: molecular

insights into an autoimmune disease. Immunol Rev 2005; 204: 156-

64.

[71] Huseby ES, White J, Crawford F, Vass T, Becker D, Pinilla C, et

al. How the T cell repertoire becomes peptide and MHC specific.

Cell 2005; 122: 247-60.

[72] Sebzda E, Mariathasan S, Ohteki T, Jones R, Bachmann MF,

Ohashi PS. Selection of the T cell repertoire. Annu Rev Immunol

1999; 17: 829-74.

[73] Sant'Angelo DB, Janeway CA Jr. Negative selection of thymocytes

expressing the D10 TCR. Proc Natl Acad Sci USA 2002; 99: 6931-

6.

[74] Murphy KM, Heimberger AB, Loh DY. Induction by antigen of

intrathymic apoptosis of CD4+CD8+TCRlo thymocytes in vivo.

Science 1990; 250: 1720-3.

[75] Martin S, Bevan MJ. Antigen-specific and nonspecific deletion of

immature cortical thymocytes caused by antigen injection. Eur J

Immunol 1997; 27: 2726-36.

[76] Lerner A, Clayton LK, Mizoguchi E, Ghendler Y, van Ewijk W,

Koyasu S, et al. Cross-linking of T-cell receptors on double-

positive thymocytes induces a cytokine-mediated stromal activation

process linked to cell death. EMBO J 1996; 15: 5876-87.

[77] Brabb T, Huseby ES, Morgan TM, Sant'Angelo DB, Kirchner J,

Farr AG, et al. Thymic stromal organization is regulated by the

specificity of T cell receptor/major histocompatibility complex in-

teractions. Eur J Immunol 1997; 27: 136-46.

[78] Baldwin KK, Trenchak BP, Altman JD, Davis MM. Negative se-

lection of T cells occurs throughout thymic development. J Immu-

nol 1999; 163: 689-98.

[79] McGargill MA, Derbinski JM, Hogquist KA. Receptor editing in

developing T cells. Nat Immunol 2000; 1: 336-41.

[80] Buch T, Rieux-Laucat F, Forster I, Rajewsky K. Failure of HY-

specific thymocytes to escape negative selection by receptor edit-

ing. Immunity 2002; 16: 707-18.

[81] Nemazee D. Receptor selection in B and T lymphocytes. Annu Rev

Immunol 2000; 18: 19-51.

[82] Jordan MS, Boesteanu A, Reed AJ, Petrone AL, Holenbeck AE,

Lerman MA, et al. Thymic selection of CD4+CD25+ regulatory T

cells induced by an agonist self-peptide. Nat Immunol 2001; 2:

301-6.

[83] Apostolou I, Sarukhan A, Klein L, von Boehmer H. Origin of regu-

latory T cells with known specificity for antigen. Nat Immunol

2002; 3: 756-63.

[84] Smith KM, Olson DC, Hirose R, Hanahan D. Pancreatic gene ex-

pression in rare cells of thymic medulla: evidence for functional

contribution to T cell tolerance. Int Immunol 1997; 9: 1355-65.

[85] Fontenot JD, Rasmussen JP, Williams LM, Dooley JL, Farr AG,

Rudensky AY. Regulatory T cell lineage specification by the fork-

head transcription factor foxp3. Immunity 2005; 22: 329-41.

[86] Laufer TM, DeKoning J, Markowitz JS, Lo D, Glimcher LH. Un-

opposed positive selection and autoreactivity in mice expressing

class II MHC only on thymic cortex. Nature 1996; 383: 81-5.

[87] Bensinger SJ, Bandeira A, Jordan MS, Caton AJ, Laufer TM. Ma-

jor histocompatibility complex class II-positive cortical epithelium

mediates the selection of CD4(+)25(+) immunoregulatory T cells. J

Exp Med 2001; 194: 427-38.

[88] Fontenot JD, Dooley JL, Farr AG, Rudensky AY. Developmental

regulation of Foxp3 expression during ontogeny. J Exp Med 2005;

202: 901-6.

[89] Derbinski J, Kyewski B. Linking signalling pathways, thymic

stroma integrity and autoimmunity. Trends Immunol 2005; 26:

503-6.

[90] Kretschmer K, Apostolou I, Hawiger D, Khazaie K, Nussenzweig

MC, von Boehmer H. Inducing and expanding regulatory T cell

populations by foreign antigen. Nat Immunol 2005; 6: 1219-27.

[91] Opferman JT, Korsmeyer SJ. Apoptosis in the development and

maintenance of the immune system. Nat Immunol 2003; 4: 410-5.

[92] Primary immunodeficiency diseases. Report of an IUIS Scientific

Committee. International Union of Immunological Societies. Clin

Exp Immunol 1999; 118 Suppl 1: 1-28.

[93] Chapel H, Geha R, Rosen F. Primary immunodeficiency diseases:

an update. Clin Exp Immunol 2003; 132: 9-15.

[94] Notarangelo L, Casanova JL, Conley ME, Chapel H, Fischer A,

Puck J, et al. Primary immunodeficiency diseases: an update from

the International Union of Immunological Societies Primary Im-

munodeficiency Diseases Classification Committee Meeting in Bu-

dapest, 2005. J Allergy Clin Immunol 2006; 117: 883-96.

[95] Bonilla FA, Geha RS. 2 Update on primary immunodeficiency

diseases. J Allergy Clin Immunol 2006; 117: 435-41.

[96] Fischer A. Human primary immunodeficiency diseases: a perspec-

tive. Nat Immunol 2004; 5: 23-30.

[97] Cunningham-Rundles C, Ponda PP. Molecular defects in T- and B-

cell primary immunodeficiency diseases. Nat Rev Immunol 2005;

5: 880-92.

[98] Puck JM, Straus SE, Le Deist F, Rieux-Laucat F, Fischer A. Inher-

ited disorders with autoimmunity and defective lymphocyte regula-

tion 1999;

[99] Lenardo MJ. Molecular regulation of T lymphocyte homeostasis in

the healthy and diseased immune system. Immunol Res 2003; 27:

387-98.

[100] Rieux-Laucat F, Le Deist F, Fischer A. Autoimmune lymphoprolif-

erative syndromes: genetic defects of apoptosis pathways. Cell

Death Differ 2003; 10: 124-33.

[101] Fisher GH, Rosenberg FJ, Straus SE, Dale JK, Middleton LA, Lin

AY, et al. Dominant interfering Fas gene mutations impair apopto-

sis in a human autoimmune lymphoproliferative syndrome. Cell

1995; 81: 935-46.

Apoptosis in the Homeostasis of the Immune System and in Human Current Pharmaceutical Design, 2008, Vol. 14, No. 3 265

[102] Rieux-Laucat F, Le Deist F, Hivroz C, Roberts IA, Debatin KM,

Fischer A, et al. Mutations in Fas associated with human lymphop-

roliferative syndrome and autoimmunity. Science 1995; 268: 1347-

9.

[103] Wu J, Wilson J, He J, Xiang L, Schur PH, Mountz JD. Fas ligand

mutation in a patient with systemic lupus erythematosus and lym-

phoproliferative disease. J Clin Invest 1996; 98: 1107-13.

[104] Drappa J, Vaishnaw AK, Sullivan KE, Chu JL, Elkon KB. Fas gene

mutations in the Canale-Smith syndrome, an inherited lymphopro-

liferative disorder associated with autoimmunity. N Engl J Med

1996; 335: 1643-9.

[105] Bettinardi A, Brugnoni D, Quiros-Roldan E, Malagoli A, La Grutta

S, Correra A, et al. Missense mutations in the Fas gene resulting in

autoimmune lymphoproliferative syndrome: a molecular and im-

munological analysis. Blood 1997; 89: 902-9.

[106] Chun HJ, Zheng L, Ahmad M, Wang J, Speirs CK, Siegel RM, et

al. Pleiotropic defects in lymphocyte activation caused by caspase-

8 mutations lead to human immunodeficiency. Nature 2002; 419:

395-9.

[107] Wang J, Zheng L, Lobito A, Chan FK, Dale J, Sneller M, et al.

Inherited human Caspase 10 mutations underlie defective lympho-

cyte and dendritic cell apoptosis in autoimmune lymphoprolifera-

tive syndrome type II. Cell 1999; 98: 47-58.

[108] Bidere N, Su HC, Lenardo MJ. Genetic disorders of programmed

cell death in the immune system. Annu Rev Immunol 2006; 24:

321-52.

[109] Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA,

Nagata S. Lymphoproliferation disorder in mice explained by de-

fects in Fas antigen that mediates apoptosis. Nature 1992; 356:

314-7.

[110] Lynch DH, Watson ML, Alderson MR, Baum PR, Miller RE,

Tough T, et al. The mouse Fas-ligand gene is mutated in gld mice

and is part of a TNF family gene cluster. Immunity 1994; 1: 131-6.

[111] Takahashi T, Tanaka M, Brannan CI, Jenkins NA, Copeland NG,

Suda T, et al. Generalized lymphoproliferative disease in mice,

caused by a point mutation in the Fas ligand. Cell 1994; 76: 969-

76.

[112] Sneller MC, Wang J, Dale JK, Strober W, Middelton LA, Choi Y,

et al. Clincal, immunologic, and genetic features of an autoimmune

lymphoproliferative syndrome associated with abnormal lympho-

cyte apoptosis. Blood 1997; 89: 1341-8.

[113] Effros RB, Boucher N, Porter V, Zhu X, Spaulding C, Walford RL,

et al. Decline in CD28+ T cells in centenarians and in long-term T

cell cultures: a possible cause for both in vivo and in vitro immu-

nosenescence. Exp Gerontol 1994; 29: 601-9.

[114] Nagata S, Golstein P. The Fas death factor. Science 1995; 267:

1449-56.

[115] Rathmell JC, Cooke MP, Ho WY, Grein J, Townsend SE, Davis

MM, et al. CD95 (Fas)-dependent elimination of self-reactive B

cells upon interaction with CD4+ T cells. Nature 1995; 376: 181-4.

[116] van den Berg A, Maggio E, Diepstra A, de Jong D, van Krieken J,

Poppema S. Germline FAS gene mutation in a case of ALPS and

NLP Hodgkin lymphoma. Blood 2002; 99: 1492-4.

[117] Boulanger E, Rieux-Laucat F, Picard C, Legall M, Sigaux F, Clau-

vel JP, et al. Diffuse large B-cell non-Hodgkin's lymphoma in a pa-

tient with autoimmune lymphoproliferative syndrome. Br J Haema-

tol 2001; 113: 432-4.

[118] Straus SE, Jaffe ES, Puck JM, Dale JK, Elkon KB, Rosen-Wolff A,

et al. The development of lymphomas in families with autoimmune

lymphoproliferative syndrome with germline Fas mutations and de-

fective lymphocyte apoptosis. Blood 2001; 98: 194-200.

[119] Chouaib S, Asselin-Paturel C, Mami-Chouaib F, Caignard A, Blay

JY. The host-tumor immune conflict: from immunosuppression to

resistance and destruction. Immunol Today 1997; 18: 493-7.

[120] Strand S, Galle PR. Immune evasion by tumours: involvement of

the CD95 (APO-1/Fas) system and its clinical implications. Mol

Med Today 1998; 4: 63-8.

[121] Kim R, Emi M, Tanabe K, Uchida Y, Toge T. The role of Fas

ligand and transforming growth factor beta in tumor progression:

molecular mechanisms of immune privilege via Fas-mediated

apoptosis and potential targets for cancer therapy. Cancer 2004;

100: 2281-91.

[122] Debatin KM, Krammer PH. Death receptors in chemotherapy and

cancer. Oncogene 2004; 23: 2950-66.

[123] Kim R, Emi M, Tanabe K, Arihiro K. Tumor-driven evolution of

immunosuppressive networks during malignant progression. Can-

cer Res 2006; 66: 5527-36.

[124] van der Werff ten Bosch J. Autoimmune lymphoproliferative syn-

drome: etiology, diagnosis, and management. Paediatr Drugs 2003;

5: 185-93.

[125] van der Werff ten Bosch J, Schotte P, Ferster A, Azzi N, Boehler T,

Laurey G, et al. Reversion of autoimmune lymphoproliferative

syndrome with an antimalarial drug: preliminary results of a clini-

cal cohort study and molecular observations. Br J Haematol 2002;

117: 176-88.

[126] Pierdominici M, Giammarioli AM, Gambardella L, De Felice M,

Quinti I, Iacobini M, et al. Pyrimethamine (2,4-diamino-5-p-

chlorophenyl-6-ethylpyrimidine) induces apoptosis of freshly iso-

lated human T lymphocytes, bypassing CD95/Fas molecule but in-

volving its intrinsic pathway. J Pharmacol Exp Ther 2005; 315:

1046-57.

[127] Dianzani U, Bragardo M, DiFranco D, Alliaudi C, Scagni P, Buon-

figlio D, et al. Deficiency of the Fas apoptosis pathway without Fas

gene mutations in pediatric patients with autoimmunity/

lymphoproliferation. Blood 1997; 89: 2871-9.

[128] Ramenghi U, Bonissoni S, Migliaretti G, DeFranco S, Bottarel F,

Gambaruto C, et al. Deficiency of the Fas apoptosis pathway with-

out Fas gene mutations is a familial trait predisposing to develop-

ment of autoimmune diseases and cancer. Blood 2000; 95: 3176-

82.

[129] Kovanen PE, Leonard WJ. Cytokines and immunodeficiency dis-

eases: critical roles of the gamma(c)-dependent cytokines interleu-

kins 2, 4, 7, 9, 15, and 21, and their signalling pathways. Immunol

Rev 2004; 202: 67-83.

[130] Leonard WJ. Cytokines and immunodeficiency diseases. Nat Rev

Immunol 2001; 1: 200-8.

[131] Macchi P, Villa A, Giliani S, Sacco MG, Frattini A, Porta F, et al.

Mutations of Jak-3 gene in patients with autosomal severe com-

bined immune deficiency (SCID). Nature 1995; 377: 65-8.

[132] Buckley RH, Schiff RI, Schiff SE, Markert ML, Williams LW,

Harville TO, et al. Human severe combined immunodeficiency:

genetic, phenotypic, and functional diversity in one hundred eight

infants. J Pediatr 1997; 130: 378-87.

[133] Roifman CM, Zhang J, Chitayat D, Sharfe N. A partial deficiency

of interleukin-7R alpha is sufficient to abrogate T-cell development

and cause severe combined immunodeficiency. Blood 2000; 96:

2803-7.

[134] Akashi K, Kondo M, von Freeden-Jeffry U, Murray R, Weissman

IL. Bcl-2 rescues T lymphopoiesis in interleukin-7 receptor-

deficient mice. Cell 1997; 89: 1033-41.

[135] Rathmell JC, Farkash EA, Gao W, Thompson CB. IL-7 enhances

the survival and maintains the size of naive T cells. J Immunol

2001; 167: 6869-76.

[136] Cunningham-Rundles C, Bodian C. Common variable immunode-

ficiency: clinical and immunological features of 248 patients. Clin

Immunol 1999; 92: 34-48.

[137] Grimbacher B, Hutloff A, Schlesier M, Glocker E, Warnatz K,

Drager R, et al. Homozygous loss of ICOS is associated with adult-

onset common variable immunodeficiency. Nat Immunol 2003; 4:

261-8.

[138] Salzer U, Chapel HM, Webster AD, Pan-Hammarstrom Q,

Schmitt-Graeff A, Schlesier M, et al. Mutations in TNFRSF13B

encoding TACI are associated with common variable immunodefi-

ciency in humans. Nat Genet 2005; 37: 820-8.

[139] Castigli E, Wilson SA, Garibyan L, Rachid R, Bonilla F, Schneider

L, et al. TACI is mutant in common variable immunodeficiency

and IgA deficiency. Nat Genet 2005; 37: 829-34.

[140] Salzer U, Grimbacher B. TACItly changing tunes: farewell to a yin

and yang of BAFF receptor and TACI in humoral immunity? New

genetic defects in common variable immunodeficiency. Curr Opin

Allergy Clin Immunol 2005; 5: 496-503.

[141] North ME, Ivory K, Funauchi M, Webster AD, Lane AC, Farrant J.

Intracellular cytokine production by human CD4+ and CD8+ T

cells from normal and immunodeficient donors using directly con-

jugated anti-cytokine antibodies and three-colour flow cytometry.

Clin Exp Immunol 1996; 105: 517-22.

[142] Saxon A, Keld B, Diaz-Sanchez D, Guo BC, Sidell N. B cells from

a distinct subset of patients with common variable immunodefi-

266 Current Pharmaceutical Design, 2008, Vol. 14, No. 3 Giovannetti et al.

ciency (CVID) have increased CD95 (Apo-1/fas), diminished

CD38 expression, and undergo enhanced apoptosis. Clin Exp Im-

munol 1995; 102: 17-25.

[143] Iglesias J, Matamoros N, Raga S, Ferrer JM, Mila J. CD95 expres-

sion and function on lymphocyte subpopulations in common vari-

able immunodeficiency (CVID); related to increased apoptosis.

Clin Exp Immunol 1999; 117: 138-46.

[144] Di Renzo M, Zhou Z, George I, Becker K, Cunningham-Rundles

C. Enhanced apoptosis of T cells in common variable immunodefi-

ciency (CVID): role of defective CD28 co-stimulation. Clin Exp

Immunol 2000; 120: 503-11.

[145] Di Renzo M, Serrano D, Zhou Z, George I, Becker K, Cunning-

ham-Rundles C. Enhanced T cell apoptosis in common variable

immunodeficiency: negative role of the fas/fasligand system and of

the Bcl-2 family proteins and possible role of TNF-RS. Clin Exp

Immunol 2001; 125: 117-22.

[146] Serrano D, Becker K, Cunningham-Rundles C, Mayer L. Charac-

terization of the T cell receptor repertoire in patients with common

variable immunodeficiency: oligoclonal expansion of CD8(+) T

cells. Clin Immunol 2000; 97: 248-58.

[147] Pirovano S, Mazzolari E, Pasic S, Albertini A, Notarangelo LD,

Imberti L. Impaired thymic output and restricted T-cell repertoire

in two infants with immunodeficiency and early-onset generalized

dermatitis. Immunol Lett 2003; 86: 93-7.

[148] Giovannetti A, Pierdominici M, Mazzetta F, Marziali M, Renzi C,

Mileo AM, et al. Unravelling the complexity of T cell abnormali-

ties in common variable immunodeficiency. J Immunol 2007; 178:

3932-43.

[149] Lin SJ, Chao HC, Chang KW, Yan DC. Effect of interleukin 15

and interleukin 2 on anti-CD3-induced T-cell activation and apop-

tosis in children with common variable immunodeficiency. Ann

Allergy Asthma Immunol 2003; 91: 65-70.

[150] Sneller MC, Strober W. Abnormalities of lymphokine gene expres-

sion in patients with common variable immunodeficiency. J Immu-

nol 1990; 144: 3762-9.

[151] Akbar AN, Borthwick NJ, Wickremasinghe RG, Panayoitidis P,

Pilling D, Bofill M, et al. Interleukin-2 receptor common gamma-

chain signalling cytokines regulate activated T cell apoptosis in re-

sponse to growth factor withdrawal: selective induction of anti-

apoptotic (bcl-2, bcl-xL) but not pro-apoptotic (bax, bcl-xS) gene

expression. Eur J Immunol 1996; 26: 294-9.

[152] Lenardo M, Chan KM, Hornung F, McFarland H, Siegel R, Wang

J, et al. Mature T lymphocyte apoptosis--immune regulation in a

dynamic and unpredictable antigenic environment. Annu Rev Im-

munol 1999; 17: 221-53.

[153] Aukrust P, Svardal AM, Muller F, Lunden B, Berge RK, Froland

SS. Decreased levels of total and reduced glutathione in CD4+

lymphocytes in common variable immunodeficiency are associated

with activation of the tumor necrosis factor system: possible im-

munopathogenic role of oxidative stress. Blood 1995; 86: 1383-91.

[154] Mullighan CG, Fanning GC, Chapel HM, Welsh KI. TNF and

lymphotoxin-alpha polymorphisms associated with common vari-

able immunodeficiency: role in the pathogenesis of granulomatous

disease. J Immunol 1997; 159: 6236-41.

[155] Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF receptor

superfamilies: integrating mammalian biology. Cell 2001; 104:

487-501.

[156] Aukrust P, Muller F, Froland SS. Enhanced generation of reactive

oxygen species in monocytes from patients with common variable

immunodeficiency. Clin Exp Immunol 1994; 97: 232-8.

[157] Ishii Y, Partridge CA, Del Vecchio PJ, Malik AB. Tumor necrosis

factor-alpha-mediated decrease in glutathione increases the sensi-

tivity of pulmonary vascular endothelial cells to H2O2. J Clin In-

vest 1992; 89: 794-802.

[158] Staal FJ, Roederer M, Herzenberg LA. Intracellular thiols regulate

activation of nuclear factor kappa B and transcription of human

immunodeficiency virus. Proc Natl Acad Sci USA 1990; 87: 9943-

7.

[159] Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J.

Free radicals and antioxidants in normal physiological functions

and human disease. Int J Biochem Cell Biol 2007; 39: 44-84.

[160] Douek DC, Picker LJ, Koup RA. T cell dynamics in HIV-1 infec-

tion. Annu Rev Immunol 2003; 21: 265-304.

[161] Atkinson K, Hansen JA, Storb R, Goehle S, Goldstein G, Thomas

ED. T-cell subpopulations identified by monoclonal antibodies af-

ter human marrow transplantation. I. Helper-inducer and cytotoxic-

suppressor subsets. Blood 1982; 59: 1292-8.

[162] Watanabe N, De Rosa SC, Cmelak A, Hoppe R, Herzenberg LA,

Roederer M. Long-term depletion of naive T cells in patients

treated for Hodgkin's disease. Blood 1997; 90: 3662-72.

[163] Hakim FT, Cepeda R, Kaimei S, Mackall CL, McAtee N, Zujewski

J, et al. Constraints on CD4 recovery postchemotherapy in adults:

thymic insufficiency and apoptotic decline of expanded peripheral

CD4 cells. Blood 1997; 90: 3789-98.

[164] Foulds KE, Zenewicz LA, Shedlock DJ, Jiang J, Troy AE, Shen H.

Cutting edge: CD4 and CD8 T cells are intrinsically different in

their proliferative responses. J Immunol 2002; 168: 1528-32.

[165] Cunningham-Rundles C, Kazbay K, Hassett J, Zhou Z, Mayer L.

Brief report: enhanced humoral immunity in common variable im-

munodeficiency after long-term treatment with polyethylene gly-

col-conjugated interleukin-2. N Engl J Med 1994; 331: 918-21.

[166] Cunningham-Rundles C, Kazbay K, Zhou Z, Mayer L. Immu-

nologic effects of low-dose polyethylene glycol-conjugated recom-

binant human interleukin-2 in common variable immunodeficiency.

J Interferon Cytokine Res 1995; 15: 269-76.

[167] Cunningham-Rundles C, Bodian C, Ochs HD, Martin S, Reiter-

Wong M, Zhuo Z. Long-term low-dose IL-2 enhances immune

function in common variable immunodeficiency. Clin Immunol

2001; 100: 181-90.

[168] Driscoll D, Sullivan KE. DiGeorge Syndrome: A Chromosome

22q11.2 Deletion syndrome 1999; 198-208.

[169] Jawad AF, McDonald-McGinn DM, Zackai E, Sullivan KE. Im-

munologic features of chromosome 22q11.2 deletion syndrome

(DiGeorge syndrome/velocardiofacial syndrome). J Pediatr 2001;

139: 715-23.

[170] Conley ME, Beckwith JB, Mancer JF, Tenckhoff L. The spectrum

of the DiGeorge syndrome. J Pediatr 1979; 94: 883-90.

[171] Bastian J, Law S, Vogler L, Lawton A, Herrod H, Anderson S, et

al. Prediction of persistent immunodeficiency in the DiGeorge

anomaly. J Pediatr 1989; 115: 391-6.

[172] Barrett DJ, Ammann AJ, Wara DW, Cowan MJ, Fisher TJ, Stiehm

ER. Clinical and immunologic spectrum of the DiGeorge syn-

drome. J Clin Lab Immunol 1981; 6: 1-6.

[173] Ryan AK, Goodship JA, Wilson DI, Philip N, Levy A, Seidel H, et

al. Spectrum of clinical features associated with interstitial chro-

mosome 22q11 deletions: a European collaborative study. J Med

Genet 1997; 34: 798-804.

[174] Muller W, Peter HH, Kallfelz HC, Franz A, Rieger CH. The DiGe-

orge sequence. II. Immunologic findings in partial and complete

forms of the disorder. Eur J Pediatr 1989; 149: 96-103.

[175] Junker AK, Driscoll DA. Humoral immunity in DiGeorge syn-

drome. J Pediatr 1995; 127: 231-7.

[176] Etzioni A, Pollack S. Hypogammaglobulinaemia in DiGeorge

sequence. Eur J Pediatr 1989; 149: 143-4.

[177] Schubert R, Reichenbach J, Zielen S. Deficiencies in CD4+ and

CD8+ T cell subsets in ataxia telangiectasia. Clin Exp Immunol

2002; 129: 125-32.

[178] Smith CA, Driscoll DA, Emanuel BS, McDonald-McGinn DM,

Zackai EH, Sullivan KE. Increased prevalence of immunoglobulin

A deficiency in patients with the chromosome 22q11.2 deletion

syndrome (DiGeorge syndrome/velocardiofacial syndrome). Clin

Diagn Lab Immunol 1998; 5: 415-7.

[179] Mayumi M, Kimata H, Suehiro Y, Hosoi S, Ito S, Kuge Y, et al.

DiGeorge syndrome with hypogammaglobulinaemia: a patient with

excess suppressor T cell activity treated with fetal thymus trans-

plantation. Eur J Pediatr 1989; 148: 518-22.

[180] Zhou H, Perkins SL, Tripp S, Hussong JW, Coffin CM. Expression

of apoptosis-related antigens, Fas, bcl-2, and p53, and Mib-1 pro-

liferation index in the hypoplastic thymus of DiGeorge syndrome.

Pediatr Dev Pathol 2002; 5: 465-71.

[181] Pierdominici M, Marziali M, Giovannetti A, Oliva A, Rosso R,

Marino B, et al. T cell receptor repertoire and function in patients

with DiGeorge syndrome and velocardiofacial syndrome. Clin Exp

Immunol 2000; 121: 127-32.

[182] Gupta S, Aggarwal S, Nguyen T. Increased spontaneous apoptosis

in T lymphocytes in DiGeorge anomaly. Clin Exp Immunol 1998;

1: 65-71.

Apoptosis in the Homeostasis of the Immune System and in Human Current Pharmaceutical Design, 2008, Vol. 14, No. 3 267

[183] Pierdominici M, Mazzetta F, Caprini E, Marziali M, Digilio MC,

Marino B, et al. Biased T-cell receptor repertoires in patients with

chromosome 22q11.2 deletion syndrome (DiGeorge syn-

drome/velocardiofacial syndrome). Clin Exp Immunol 2003; 132:

323-31.

[184] Piliero LM, Sanford AN, McDonald-McGinn DM, Zackai EH,

Sullivan KE. T-cell homeostasis in humans with thymic hypoplasia

due to chromosome 22q11.2 deletion syndrome. Blood 2004; 103:

1020-5.

[185] Cohen PL, Eisenberg RA. Lpr and gld: single gene models of sys-

temic autoimmunity and lymphoproliferative disease. Annu Rev

Immunol 1991; 9: 243-69.

[186] Quezada SA, Jarvinen LZ, Lind EF, Noelle RJ. CD40/CD154

interactions at the interface of tolerance and immunity. Annu Rev

Immunol 2004; 22: 307-28.

[187] Carreno BM, Collins M. The B7 family of ligands and its recep-

tors: new pathways for costimulation and inhibition of immune re-

sponses. Annu Rev Immunol 2002; 20: 29-53.

[188] Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V,

Ross AJ, et al. Proapoptotic BAX and BAK: a requisite gateway to

mitochondrial dysfunction and death. Science 2001; 292: 727-30.

[189] Moretta A, Bottino C, Vitale M, Pende D, Biassoni R, Mingari

MC, et al. Receptors for HLA class-I molecules in human natural

killer cells. Annu Rev Immunol 1996; 14: 619-48.

[190] Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation dur-

ing apoptosis. Nat Rev Mol Cell Biol 2004; 5: 897-907.

[191] Kinne RW, Brauer R, Stuhlmuller B, Palombo-Kinne E, Burmester

GR. Macrophages in rheumatoid arthritis. Arthritis Res 2000; 2:

189-202.

[192] Buckley CD, Pilling D, Lord JM, Akbar AN, Scheel-Toellner D,

Salmon M. Fibroblasts regulate the switch from acute resolving to

chronic persistent inflammation. Trends Immunol 2001; 22: 199-

204.

[193] Choy EH, Panayi GS. Cytokine pathways and joint inflammation in

rheumatoid arthritis. N Engl J Med 2001; 344: 907-16.

[194] Cutolo M. Estrogen metabolites: increasing evidence for their role

in rheumatoid arthritis and systemic lupus erythematosus. J Rheu-

matol 2004; 31: 419-21.

[195] Liu H, Pope RM. The role of apoptosis in rheumatoid arthritis. Curr

Opin Pharmacol 2003; 3: 317-22.

[196] Larochelle B, Flamand L, Gourde P, Beauchamp D, Gosselin J.

Epstein-Barr virus infects and induces apoptosis in human neutro-

phils. Blood 1998; 92: 291-9.

[197] Papadaki HA, Kritikos HD, Gemetzi C, Koutala H, Marsh JC,

Boumpas DT, et al. Bone marrow progenitor cell reserve and func-

tion and stromal cell function are defective in rheumatoid arthritis:

evidence for a tumor necrosis factor alpha-mediated effect. Blood

2002; 99: 1610-9.

[198] Tosato G, Steinberg AD, Yarchoan R, Heilman CA, Pike SE, De

SV, et al. Abnormally elevated frequency of Epstein-Barr virus-

infected B cells in the blood of patients with rheumatoid arthritis. J

Clin Invest 1984; 73: 1789-95.

[199] Girbal-Neuhauser E, Durieux JJ, Arnaud M, Dalbon P, Sebbag M,

Vincent C, et al. The epitopes targeted by the rheumatoid arthritis-

associated antifilaggrin autoantibodies are posttranslationally gen-

erated on various sites of (pro)filaggrin by deimination of arginine

residues. J Immunol 1999; 162: 585-94.

[200] Kahari VM, Sandberg M, Kalimo H, Vuorio T, Vuorio E. Identifi-

cation of fibroblasts responsible for increased collagen production

in localized scleroderma by in situ hybridization. J Invest Dermatol

1988; 90: 664-70.

[201] Flanders KC. Smad3 as a mediator of the fibrotic response. Int J

Exp Pathol 2004; 85: 47-64.

[202] Fadok VA, McDonald PP, Bratton DL, Henson PM. Regulation of

macrophage cytokine production by phagocytosis of apoptotic and

post-apoptotic cells. Biochem Soc Trans 1998; 26: 653-6.

[203] Manfredi AA, Rovere P, Galati G, Heltai S, Bozzolo E, Soldini L,

et al. Apoptotic cell clearance in systemic lupus erythematosus. I.

Opsonization by antiphospholipid antibodies. Arthritis Rheum

1998; 41: 205-14.

[204] Manfredi AA, Rovere P, Heltai S, Galati G, Nebbia G, Tincani A,

et al. Apoptotic cell clearance in systemic lupus erythematosus. II.

Role of beta2-glycoprotein I. Arthritis Rheum 1998; 41: 215-23.

[205] Yanagawa T, Hidaka Y, Guimaraes V, Soliman M, DeGroot LJ.

CTLA-4 gene polymorphism associated with Graves' disease in a

Caucasian population. J Clin Endocrinol Metab 1995; 80: 41-5.

[206] Ueda H, Howson JM, Esposito L, Heward J, Snook H, Chamber-

lain G, et al. Association of the T-cell regulatory gene CTLA4 with

susceptibility to autoimmune disease. Nature 2003; 423: 506-11.

[207] Boirivant M, Marini M, Di Felice G, Pronio AM, Montesani C,

Tersigni R, et al. Lamina propria T cells in Crohn's disease and

other gastrointestinal inflammation show defective CD2 pathway-

induced apoptosis. Gastroenterology 1999; 116: 557-65.

[208] Monteleone I, Monteleone G, Fina D, Caruso R, Petruzziello C,

Calabrese E, et al. A functional role of flip in conferring resistance

of Crohn's disease lamina propria lymphocytes to FAS-mediated

apoptosis. Gastroenterology 2006; 130: 389-97.

[209] Ina K, Itoh J, Fukushima K, Kusugami K, Yamaguchi T, Kyokane

K, et al. Resistance of Crohn's disease T cells to multiple apoptotic

signals is associated with a Bcl-2/Bax mucosal imbalance. J Immu-

nol 1999; 163: 1081-90.

[210] Fayad R, Brand MI, Stone D, Keshavarzian A, Qiao L. Apoptosis

resistance in ulcerative colitis: high expression of decoy receptors

by lamina propria T cells. Eur J Immunol 2006; 36: 2215-22.

[211] Seidelin JB, Nielsen OH. Expression profiling of apoptosis-related

genes in enterocytes isolated from patients with ulcerative colitis.

APMIS 2006; 114: 508-17.

[212] Atreya R, Mudter J, Finotto S, Mullberg J, Jostock T, Wirtz S, et

al. Blockade of interleukin 6 trans signalling suppresses T-cell re-

sistance against apoptosis in chronic intestinal inflammation: evi-

dence in crohn disease and experimental colitis in vivo. Nat Med

2000; 6: 583-8.

[213] Neurath MF, Finotto S, Fuss I, Boirivant M, Galle PR, Strober W.

Regulation of T-cell apoptosis in inflammatory bowel disease: to

die or not to die, that is the mucosal question. Trends Immunol

2001; 22: 21-6.

[214] Yen D, Cheung J, Scheerens H, Poulet F, McClanahan T,

McKenzie B et al. IL-23 is essential for T cell-mediated colitis and

promotes inflammation via IL-17 and IL-6. J Clin Invest 2006;

116: 1310-6.

[215] Berg DJ, Davidson N, Kuhn R, Muller W, Menon S, Holland G, et

al. Enterocolitis and colon cancer in interleukin-10-deficient mice

are associated with aberrant cytokine production and CD4(+) TH1-

like responses. J Clin Invest 1996; 98: 1010-20.

[216] Meresse B, Rutgeerts P, Malchow H, Dubucquoi S, Dessaint JP,

Cohard M, et al. Low ileal interleukin 10 concentrations are predic-

tive of endoscopic recurrence in patients with Crohn's disease. Gut

2002; 50: 25-8.

[217] Barbara G, Xing Z, Hogaboam CM, Gauldie J, Collins SM. Inter-

leukin 10 gene transfer prevents experimental colitis in rats. Gut

2000; 46: 344-9.

[218] Atreya R, Atreya I, Neurath MF. Novel signal transduction path-

ways: analysis of STAT-3 and Rac-1 signalling in inflammatory

bowel disease. Ann N Y Acad Sci 2006; 1072: 98-113.

[219] Al Ashy R, Chakroun I, El Sabban ME, Homaidan FR. The role of

NF-kappaB in mediating the anti-inflammatory effects of IL-10 in

intestinal epithelial cells. Cytokine 2006; 36: 1-8.

[220] Castro-Santos P, Suarez A, Lopez-Rivas L, Mozo L, Gutierrez C.

TNFalpha and IL-10 gene polymorphisms in inflammatory bowel

disease. Association of -1082 AA low producer IL-10 genotype

with steroid dependency. Am J Gastroenterol 2006; 101: 1039-47.

[221] Burakoff R, Barish CF, Riff D, Pruitt R, Chey WY, Farraye FA, et

al. A phase 1/2A trial of STA 5326, an oral interleukin-12/23 in-

hibitor, in patients with active moderate to severe Crohn's disease.

Inflamm Bowel Dis 2006; 12: 558-65.

[222] Fuss IJ, Marth T, Neurath MF, Pearlstein GR, Jain A, Strober W.

Anti-interleukin 12 treatment regulates apoptosis of Th1 T cells in

experimental colitis in mice. Gastroenterology 1999; 117: 1078-88.

[223] Caproni M, Torchia D, Antiga E, Degl'Innocenti D, Barletta E,

Baroni G, et al. The role of apoptosis in the pathogenesis of derma-

titis herpetiformis. Int J Immunopathol Pharmacol 2005; 18: 691-9.

[224] Sollid LM, Thorsby E. HLA susceptibility genes in celiac disease:

genetic mapping and role in pathogenesis. Gastroenterology 1993;

105: 910-22.

[225] Frezza D, Giambra V, Cianci R, Fruscalzo A, Giufre M, Cammaro-

ta G, et al. Increased frequency of the immunoglobulin enhancer

268 Current Pharmaceutical Design, 2008, Vol. 14, No. 3 Giovannetti et al.

HS1,2 allele 2 in coeliac disease. Scand J Gastroenterol 2004; 39:

1083-7.

[226] Ciccocioppo R, Di Sabatino A, Parroni R, D'alo S, Pistoia MA,

Doglioni C, et al. Cytolytic mechanisms of intraepithelial lympho-

cytes in coeliac disease (CoD). Clin Exp Immunol 2000; 120: 235-

40.

[227] Di Sabatino A, D'alo S, Millimaggi D, Ciccocioppo R, Parroni R,

Sciarra G, et al. Apoptosis and peripheral blood lymphocyte deple-

tion in coeliac disease. Immunology 2001; 103: 435-40.

[228] Eiras P, Leon F, Camarero C, Lombardia M, Roldan E, Bootello A,

et al. Intestinal intraepithelial lymphocytes contain a CD3- CD7+

subset expressing natural killer markers and a singular pattern of

adhesion molecules. Scand J Immunol 2000; 52: 1-6.

[229] Buri C, Burri P, Bahler P, Straumann A, Muller-Schenker B, Birrer

S, et al. Cytotoxic T cells are preferentially activated in the duode-

nal epithelium from patients with florid coeliac disease. J Pathol

2005; 206: 178-85.

[230] Di Sabatino A, Ciccocioppo R, Cupelli F, Cinque B, Millimaggi D,

Clarkson MM, et al. Epithelium derived interleukin 15 regulates in-

traepithelial lymphocyte Th1 cytokine production, cytotoxicity, and

survival in coeliac disease. Gut 2006; 55: 469-77.

[231] Ebert EC. IL-15 converts human intestinal intraepithelial lympho-

cytes to CD94 producers of IFN-gamma and IL-10, the latter pro-

moting Fas ligand-mediated cytotoxicity. Immunology 2005; 115:

118-26.

[232] Gonzalez-Rey E, Varela N, Chorny A, Delgado M. Therapeutical

approaches of vasoactive intestinal peptide as a pleiotropic immu-

nomodulator. Curr Pharm Des 2007; 13(11): 1113-39.

[233] Klumpp K, Mirzadegan T. Recent progress in the design of small

molecule inhibitors of HIV RNase H. Curr Pharm Des 2006;

12(15): 1909-22.

[234] Paganelli R, Di Iorio A, Cherubini A, Lauretani F, Mussi C, Vol-

pato S, et al. Frailty of older age: the role of the endocrine--

immune interaction. Curr Pharm Des 2006; 12(24): 3147-59.