Sie sind auf Seite 1von 11

REVIEW ARTICLE

Histologic and Molecular Assessment of Human Thymus


Laura P. Hale, MD, PhD
Most work describing the histopathology of normal human thymus has focused on
pediatric thymus because of tissue availability and high thymopoietic activity.
However, pathologic examination of the thymus can provide information about
immune status that is relevant to the clinical care of patients of all ages. Under-
standing age-related changes in the relative abundance and composition of anatomic
compartments within the thymus is critical for evaluation of the thymus in normal
adults and patients with diseases that affect the thymus. The purpose of this review
is to acquaint diagnostic pathologists with some of the newer histologic, flow
cytometric, and molecular techniques for assessment of non-neoplastic thymus.
Diagnostic criteria are presented for assessment of thymic function and for deter-
mining the mechanisms underlying thymic hyperplasia. Accurate assessment of
thymic function is critical for the diagnosis and treatment of patients with complete
DiGeorge syndrome and can complement the clinical care of patients with a variety
of disorders that affect the immune system.
Ann Diagn Pathol 8: 50-60, 2004. © 2004 Elsevier Inc. All rights reserved.

E XAMINATION of the thymus is an important


part of pediatric autopsies, particularly those
with a history suggestive of immunodeficiency. Al-
parathyroid, to ensure appropriate surgical mar-
gins.
It is of paramount importance to rule out the
though the thymus is rarely examined in adult presence of neoplasia in thymic specimens. Thymic
autopsies, it could potentially provide important epithelial neoplasms (thymoma and thymic carci-
information regarding the patient’s immune status, noma), lymphomas, and neuroendocrine or germ
which might help improve the clinical care of fu- cell tumors make up the majority of thymic neo-
ture patients. Thymus specimens are also submit- plasms. Metastatic tumors are extremely rare. Ex-
ted to the surgical pathology service. Patients with cellent reviews are available regarding the diagnos-
myasthenia gravis (MG) may undergo therapeutic tic histopathology and clinical outcome of thymic
thymectomy, a procedure that can lead to remis- neoplasms.1-7 However, beyond the presence or
absence of neoplasia, thymus tissues provide infor-
sion of their autoimmune disease or to decreased
mation about immune status that is relevant to the
need for treatment with immunosuppressive
clinical care of patients of all ages. The purpose of
agents. Thymus biopsies may be obtained to rule
this review is to acquaint diagnostic pathologists
out malignancy in patients with symptomatic or
with some of the newer histologic and molecular
radiographically identified thymic enlargement or techniques for assessment of non-neoplastic thy-
masses. Portions of the thymus may be removed at mus tissues. Such techniques are critical for the
the time of cardiovascular surgery, either to expose diagnosis and treatment of patients with complete
the operative site or to diagnose perceived discor- DiGeorge syndrome,8 and can complement the
dances in age and thymus cellularity. Thymus tis- clinical care of patients with a variety of disorders
sues may also be removed at the time of surgery for that affect the immune system.
benign and malignant conditions of the thyroid or
Age-Related Changes in Thymus Histology
and Function
From the Department of Pathology, Duke University Medical Center, The thymus has traditionally has been consid-
Durham, NC. ered to be composed of a cortex and a medulla,
Address reprint requests to Laura P. Hale, MD, PhD, Box 3712, which contain developing thymocytes within a net-
Duke University Medical Center, Durham, NC 27710.
© 2004 Elsevier Inc. All rights reserved. work of thymic epithelial and stromal cells. Hassall
1092-9134/04/0801-0010$30.00/0 bodies composed of swirls of terminally differenti-
doi:10.1016/j.anndiagpath.2003.11.006 ated thymic epithelial cells9-12 are present in the

50 Annals of Diagnostic Pathology, Vol 8, No 1 (February), 2004: pp 50-60


Assessment of Human Thymus 51

medulla and are a universally recognized histologic Although total thymic weight and volume remain
indicator of the thymus. Because they both contain relatively constant,13 the portion of the thymus that
thymic epithelial cells, the cortex and medulla can contains thymic epithelium and participates in pro-
also be referred to together as the thymic epithelial duction of new T cells decreases progressively with
space. The perivascular space is defined as the age (Fig 1).13,14,17,18 The thymic perivascular space
portion of tissue that is within the thymic capsule increases correspondingly, with prominent infil-
but outside of the thymic epithelial network.13,14 In trates of peripheral lymphocytes and adipose tissue.
infant thymus, the perivascular space is essentially a The density of lymphocytes in the perivascular
virtual space containing only the thymic blood ves- space is typically very similar to that of the adjacent
sels and thus has largely been ignored. However, medulla. However, changes in proportion of thy-
the perivascular space becomes more prominent mic epithelial and perivascular spaces can be high-
with aging. Understanding the role of thymic epi- lighted using an immunostain (eg, cytokeratin) to
thelial and perivascular compartments in T-cell identify thymic epithelium. Use of a hematoxylin-
production (thymopoiesis) and immune reactions eosin counterstain (rather than hematoxylin
is critical for accurate pathologic evaluation of non- alone) with the cytokeratin immunostain is partic-
neoplastic adult thymus. ularly useful for identifying changes in thymic com-
The thymic epithelial compartment is the site of partments (Fig 1). Many of the lymphocytes
T-lymphocyte maturation within the thymus. Stem present within the thymic perivascular space have a
cells migrate from the bone marrow to the thymus, phenotype consistent with cytotoxic or memory T
where they undergo rearrangement of their T-cell cells (CD4 or CD8⫹, CD45RO [memory T cell]⫹,
receptor genes to create functional T-cell receptors TIA-1 [cytotoxic granule]⫹, CD38⫺)14 and thus
with unique specificities. Developing T cells inter- have migrated to the thymic perivascular space
act with thymic epithelium and/or stromal cells to from the periphery. A subset of B cells present in
achieve positive selection of cells that recognize the perivascular space have somatic mutations in
antigen in the context of self-major histocompati- immunoglobulin genes, showing their previous
bility complex antigens and negative selection to participation in germinal center reactions.19 High
delete cells with inappropriate autoreactivity. Fi- endothelial venules that express the ligand for the
nally, the thymocytes acquire cell surface markers CD62L cell surface molecule used by lymphocytes
typical of mature T cells and are released into the to home to lymph nodes are present in thymic
peripheral T-cell pool as new naive T cells. perivascular spaces that contain prominent infil-
The thymus is absolutely required to establish a trates of T and B lymphocytes.14 Therefore, the
functional T-cell repertoire. Animal models of athy- adult thymus can be viewed as a chimeric organ,
mia (eg, athymic “nude” mice with mutation of the with a thymic epithelial compartment that pro-
whn, winged helix-nude gene15) have been exten- duces new T lymphocytes and a perivascular com-
sively used in medical research because their pro- partment that participates in recirculation of pe-
found immunodeficiency prevents rejection of xe- ripheral lymphocytes similar to a lymph node.20 As
nogeneic cells and tissue. Human children with aging progresses, lymphocyte infiltrates into the
complete DiGeorge syndrome have a developmen- perivascular space decrease and adipose tissue in-
tal defect that results in total absence of thymus filtration becomes prominent (Fig 1). However,
tissue. These children are also profoundly immu- despite these age-related changes in thymic com-
nodeficient and typically die of infection before 2 position, we and others have shown that thymopoi-
years of age unless immune reconstituted by thymic esis continues to occur in the thymus of human
transplantation is achieved.16 Consistent with its adults into late in life.14,21-23 Foci of immature
role in establishing the initial T-cell repertoire, the thymocytes undergoing development can be iden-
human thymus is most active in T-cell production tified even in thymus tissues that grossly and micro-
early in life. Most work describing the histopathol- scopically consist primarily of adipose tissue (Fig 2,
ogy and function of normal human thymus has see below). Understanding age-related changes in
focused on pediatric thymus because of tissue avail- relative abundance and composition of thymic
ability and high thymopoietic activity. However, the compartments is critical to evaluating thymus his-
thymus rapidly begins to undergo age-related de- topathology of normal adults and patients with
creases in activity, beginning in early childhood. diseases that affect the thymus.
52 Laura P. Hale

Figure 1. Age-related changes in the


human thymus. Total thymic size re-
mains relatively constant with aging,
but the fraction that contains thymic
epithelium and participates in thymo-
cyte production (pink) decreases pro-
gressively as shown. Thymus sections
stained with cytokeratin immunoperox-
idase plus hematoxylin-eosin demon-
strate corresponding changes in rela-
tive amounts of cortex (C), medulla (M),
and perivascular space (P) with aging.
(Low-power view.)

Figure 2. Aged thymus. The perivascular space contains abun-


dant adipose tissue and sparse infiltrates of mature lymphocytes
in this thymus from a 78-year-old woman. The oval region indi-
cated by the arrow contains a small focus of thymopoiesis, with
phenotypically immature thymocytes located within a light, lacy
network of thymic epithelial cells. (Scanning power.)

Figure 3. Histologic evaluation of thy-


mic function. Thymus from a 6-month-
old male infant shows robust thymopoi-
esis, with a light lacy cytokeratin
network, abundant CD1a⫹ mib-1⫹
small lymphocytes, and non-calcified,
non-cystic Hassall bodies (indicated by
arrowheads). (A) Hematoxylin-eosin.
(Scanning power.) (B) Cytokeratin im-
munoperoxidase (mAb clones AE1 ⫹
AE3). (Medium-power magnification.)
(C) CD1a immunoperoxidase (mAb
clone O10). (Medium-power magnifica-
tion.) (D) Ki-67 immunoperoxidase
(mAb clone mib-1). (Medium-power
magnification.) C, cortex; M, medulla.
Assessment of Human Thymus 53

Histologic and Flow Cytometric Assessment of


Thymic Function

A large number of research studies in both ani-


mals and humans have determined the phenotype
of immature and mature thymocytes and periph-
eral lymphocytes and have established appropriate
cell surface markers that distinguish between them.
The most immature thymocytes (“triple negative
thymocytes”) do not express CD3, CD4, or CD8,
although they typically express other lymphocyte
markers such as CD45 and CD7. As these thymo-
cytes begin to undergo T-cell receptor gene rear-
rangement, they acquire cytoplasmic then cell sur-
face expression of CD3 and co-expression of CD4
and CD8. CD4⫹CD8⫹ “double positive” thymocytes
are located within the thymic cortex. Double posi-
tive thymocytes additionally express CD1a on their Figure 4. Flow cytometric determination of thymocyte sub-
surface (Fig 3C). They are undergoing rapid clonal sets. Cells mechanically dissociated from thymus tissue from a
expansion and thus also react with mib-1 mAb that 3-month-old female infant are reacted with fluorescently labeled
CD4 and CD8 mAbs and analyzed by multicolor flow cytometry.
recognizes the Ki-67 nuclear proliferation antigen The relative fluorescence with CD4-phycoerythrin (PE) versus
(Fig 3D). As double positive thymocytes complete CD8-phycoerythrin-cyanin 5.1 (Cy5) antibodies for the lympho-
T-cell receptor gene rearrangement and undergo cytes gated is presented as a dot plot. Immature thymocytes
make up the bulk of the cells in this thymus with high thymo-
positive and negative selection, they lose expres- poietic activity. These cells co-express CD4 and CD8 surface
sion of both CD1a and Ki-67 and either CD4 or molecules and appear in the upper right quadrant of the graph.
CD8. The resulting CD4⫹ or CD8⫹ (“single posi-
tive”) thymocytes migrate into the thymic medulla
where they complete their maturation before they lymphocytes because of the increased numbers of
are released to the periphery as naive mature T thymic B cells (detected as CD4⫺CD8⫺ lympho-
cells. Knowledge of this developmental sequence cytes) and infiltration of CD4⫹ or CD8⫹ single
allows identification of ongoing thymopoiesis ei- positive peripheral T cells into the thymic perivas-
ther by multi-color flow cytometry or by immuno- cular space.14,19). Triple color flow cytometry may
histologic analysis of tissue sections. be used for more precise identification of T-cell
Multicolor flow cytometry is an ideal way to iden- subsets, using reactivity with CD3 mAb to identify
tify developing thymocytes within suspensions of the lymphocyte population that will be analyzed for
cells mechanically dissociated from thymic tissue. reactivity with CD4 and CD8 mAbs.
Lymphocytes can be identified by their character- Thymic tissues actively participating in thymopoi-
istic forward scatter and side scatter properties. esis have a light lacy pattern of cytokeratin-positive
Double positive immature thymocytes can then be thymic epithelial cells that allows epithelial cells to
identified by their reactivity with both CD4 and interact with multiple thymocytes (Fig 3B). The
CD8 mAbs (Fig 4, upper right quadrant). The presence of islands of condensed thymic epithelial
CD4⫺CD8⫺ population (Fig 4, lower left quadrant) cells is characteristic of inactive thymus. Phenotyp-
contains the most immature triple negative thymo- ically immature (eg, CD1a⫹ mib-1⫹) thymocytes
cytes as well as thymic B cells. Cells reactive with are present within the thymic epithelial network in
either CD4 or CD8, but not both (Fig 4, upper left active thymus (Fig 3C and D). In addition, we have
and lower right quadrants), represent the more found that tissues with robust thymopoiesis that is
mature medullary thymocytes as well as any periph- confirmed by flow cytometric or molecular meth-
eral T lymphocytes present. The majority (typically ods also typically contain non-cystic, non-calcified
80% to 85%) of lymphocytes present in pediatric Hassall bodies (Fig 3). Thus thymic tissues with: (1)
thymus have a CD4⫹CD8⫹ phenotype. Adult thy- a light lacy pattern of cytokeratin-positive thymic
mus typically has a lower percentage of CD4⫹CD8⫹ epithelial cells; (2) CD1a⫹ mib-1⫹ small lympho-
54 Laura P. Hale

Figure 5. Ligation-mediated PCR de-


tects DNA breaks because of ongoing
T-cell receptor gene (TCR) rearrange-
ment. Breaks present in DNA isolated
from thymocytes are tagged by ligation
with a linker oligonucleotide (hatched
pattern). Breaks specifically associated
with recombinase-mediated TCR gene
rearrangement are identified by nested
PCR using primers (indicated by small
horizontal arrows) specific for both the
linker and for TCR gene sequences.
Ligation-mediated PCR is specific for
ongoing TCR rearrangement (lanes 1
and 3 represent DNA from two different
thymus donors). No signals are pro-
duced when the DNA lacks TCR loci
(lane 5 from bacterial DNA), contains
TCR loci that are not undergoing rear-
rangement (eg, spleen or tonsil DNA,
not shown), or when the ligation step is
omitted (lanes 2, 4, and 6).

cytes; and (3) non-calcified, non-cystic Hassall bod- ated PCR has been used to directly confirm the
ies, can be said to have histologic evidence for presence of ongoing T-cell receptor gene rear-
thymopoiesis. rangement in adult thymus tissues.14
Surrogate measures of thymopoiesis have been
Molecular Analysis of Thymic Function developed to allow assessment of thymic function
T-cell receptor gene rearrangement is a defining without the need for surgery to procure thymic
event of T-cell development within the thymus. tissue. The DNA excised from the chromosome as
Molecular techniques for assessing thymic function T-cell receptor gene rearrangement occurs within
can be divided into two types: those that analyze the thymus is retained as an episome (DNA not
thymic tissue directly for the presence of ongoing associated with a chromosome) within the develop-
T-cell receptor gene rearrangement and those that ing T cell.24 These episomes, termed T cell recep-
analyze peripheral T cells for evidence of prior tor excised circles or TRECs, cannot replicate and
T-cell rearrangement within the thymus. are lost by dilution when the T cells containing
To create T-cell receptors with unique specifici- them undergo clonal expansion after encountering
ties, concerted DNA breakage and repair occurs at antigen in the periphery. T cells that arise in the
specific recombination signal sequences within the periphery via clonal expansion are thus TREC-
T-cell receptor gene under the direction of a re- negative. TREC-positive peripheral cells are either
combinase enzyme complex (Fig 5). A ligation- newly developed T cells that have recently emi-
mediated polymerase chain reaction (PCR) assay grated from the thymus or they are long-lived naive
can be used to directly quantitate the DNA breaks T cells that have never encountered antigen or
that occur at specific loci within T-cell receptor undergone clonal expansion. TRECs present
genes during thymocyte development. DNA is ex- within DNA derived from specific cell populations
tracted from thymocytes or thymus tissue and a (peripheral blood mononuclear cells [PBMCs],
double-stranded linker oligonucleotide is ligated to CD3⫹ T cells, etc) can be quantitated by quantita-
all DNA breaks present in the sample. Breaks (now tive competitive or real-time PCR assays.25 These
tagged by the ligated linker) located adjacent to assays detect the sequence at the joint where the
T-cell receptor loci are identified by nested PCR two ends of the excised previously linear DNA have
using primers specific for the linker and the T-cell been circularized to form the TREC episome (Fig
receptor locus of interest (Fig 5). Ligation-medi- 6). The number of TRECs per ␮g PBMC DNA has
Assessment of Human Thymus 55

Figure 6. The TREC assay provides


a surrogate measure of thymic func-
tion. (A) T-cell receptor excision circles
are generated when a portion of the
TCR ␦ locus is excised before rear-
rangement of the TCR ␣ gene within
the thymus. A quantitative competitive
or real-time PCR assay that recognizes
unique sequences at the site of circu-
larization (horizontal arrow) can be
used to detect TRECs that are present
in PBMC. (B) The age-related de-
crease in the proportion of TREC⫹
PBMCs shown parallels the decrease
in thymus volume devoted to thymo-
cyte production as measured histologi-
cally.14,18 A portion of this data was
presented previously by Patel et al.31

been validated as a surrogate marker for thymopoi- regions but have differing numbers of amino acids
esis in aging normal patients (Fig 6),18 patients in their antigen recognition sites (complementarity
with MG,18,26 and following immune reconstitution determining regions), corresponding to each
therapy after human immunodeficiency virus group of three added or subtracted nucleotides.
(HIV) infection,21,25,27-29 cancer chemotherapy The diversity of the T-cell repertoire can be deter-
and umbilical cord blood transplantation,30 and mined by PCR measurement of V segment length.
transplant of bone marrow or thymus to treat cDNA derived from peripheral blood T cells is
congenital immunodeficiencies.8,31 The absolute amplified with fluorescently labeled primers that
number and frequency of naive phenotype recognize specific V segments and the constant
(CD3⫹CD45RA⫹ CD62Lhigh) T cells in peripheral region of the T-cell receptor gene. Polymerase
blood is often measured by multicolor flow cytom- chain reaction product length is determined using
etry in conjunction with TREC analysis because DNA sequencing gels with fluorescent detection.
most cells with this phenotype have undergone This assay has been called the “immunoscope”
thymic development. T cells generated by clonal technique.32 T-cell repertoires of normal diversity
expansion in the periphery typically express the have a Gaussian distribution of receptor lengths,
RO isoform of CD45 recognized by the mAb with an average of 6 to 10 peaks for each V segment
UCHL-1. family (Fig 7C). Oligoclonal distributions with ⱕ 4
Because the thymus is responsible for establish- peaks or polyclonal non-Gaussian “skewed” distri-
ing the T-cell repertoire, its function can also be butions (Figure 7A, B) are associated with immu-
assessed by examination of the diversity of T-cell nodeficiency and susceptibility to infection and/or
receptors present in peripheral blood T cells. Ran- autoimmune phenomena.8,32
dom addition or subtraction of nucleotides at the
ends of rearranging T-cell receptor gene segments Thymic Hyperplasia
through the action of terminal deoxynucleotide
transferase or deoxynucleotidases creates families The non-malignant thymic enlargement that has
of T-cell receptors that share the same variable (V) been classically termed “thymic hyperplasia” may
56 Laura P. Hale

topathologic confirmation of ongoing thymopoi-


esis. Adult patients with true thymic hyperplasia
may be at increased risk for autoimmune dis-
ease.28,35 Immunodeficient patients lacking thymo-
poiesis may need additional treatment to effect
immune reconstitution. Thus, it is important that
the anatomic diagnoses rendered on thymus spec-
imens reflect our understanding of the mecha-
nisms for non-neoplastic thymic enlargement.
Myasthenia gravis (MG) is an autoimmune dis-
Figure 7. Molecular (Immunoscope) analysis of T-cell receptor ease that is typically caused by autoantibodies
repertoire diversity. RNA isolated from PBMC is reverse tran- against the acetylcholine receptor that disrupt neu-
scribed to cDNA, then the regions encoding the variable antigen romuscular transmission. For reasons that are still
binding (V␤) regions of the T-cell receptor ␤ chain are amplified
with primers specific for each V␤ family and the ␤ chain constant not well understood, therapeutic thymectomy may
(C␤) region. Polymerase chain reaction products are rendered result in clinical improvement of MG symptoms
fluorescent using a fluorescent C␤ primer. Fluorescence intensity and allow tapering of immunosuppressive medica-
as a function of PCR product size (in base pairs) is determined
using standard DNA sequencing equipment. Abnormal repertoires tions.36 Follicular thymic hyperplasia is the charac-
in patients with deficient thymic activity may be oligoclonal (A) or teristic lesion observed in non-neoplastic thymus
skewed polyclonal (B). Normal repertoires contain a Gaussian from patients with MG. The percentage of the
distribution of receptor lengths (C). A full immunoscope analysis
includes 23 similar peak density histograms using a panel of 23 V␤ thymus that is composed of perivascular space is
primers as described.32 thus increased in MG patients,14 with a correspond-
ing decrease in the thymic volume available for
production of new T cells. The hyperplasia in MG
occur because of two distinct mechanisms, with thymus, including follicles with germinal centers, is
different implications for patient treatment and located totally in the perivascular space (Fig 8).
prognosis. Marked thymic enlargement may be Thus, despite increased thymus cellularity com-
seen in settings of thymic reconstitution after che- pared with age-matched normals, MG patients have
motherapy33 or after institution of highly active increased percentages of thymic perivascular
antiretroviral therapy in HIV-infected patients.27-29 space14 and significantly decreased thymopoiesis as
True thymic hyperplasia with active thymopoiesis is measured by the TREC assay when compared with
characterized by expansion of the thymic epithelial age-matched normal patients.18,26 Myasthenia gra-
network by phenotypically immature thymocytes vis patients whose thymus tissues show histologic
and may be associated with decreased susceptibility evidence of robust thymopoiesis have decreased
to opportunistic infections and subsequent normal- TREC levels in peripheral blood lymphocytes fol-
ization of immune function. In contrast, thymic lowing thymectomy. In contrast, MG patients
enlargement may also occur because of increased whose thymus tissues show minimal to no thymo-
migration of mature T and B lymphocytes to the poiesis have no change in TRECs following thymec-
perivascular space, adjacent to but outside of the tomy.26 Serial measurements of TREC levels in MG
thymic epithelial network. If present, follicles with patients at time points following thymectomy fur-
germinal centers are also located within the ther suggest that thymic activity also may regulate
perivascular space (Fig 8).14,34 Hyperplasia of the the turnover of peripheral T cells. Following
perivascular space typically reduces thymic produc- thymectomy, naive phenotype T-cell levels remain
tion of new T cells and may result in massive relatively constant, whereas memory phenotype T-
thymus enlargement with minimal thymopoiesis. cell concentrations increase.26
These two distinct pathogenetic conditions may
appear similar on hematoxylin-eosin–stained sec- Evaluation of Thymus from Pediatric Patients
tions, in part because the density of cells is similar With Suspected Immunodeficiency
in the medulla and the adjacent perivascular space;
however, they can be readily distinguished using The thymus from patients with severe combined
cytokeratin immunostains. Additional immuno- immunodeficiency is typically markedly hypoplastic
stains (CD1a, Ki-67) can be used to provide his- (⬍3 g; normal 12 ⫾ 5 g for 12-month-old male
Assessment of Human Thymus 57

Figure 8. Follicular hyperplasia occurs in the perivascular


space. Combined cytokeratin immunoperoxidase (brown color) Figure 10. Stress and corticosteroid-induced thymic involu-
and hematoxylin-eosin stains show that the germinal centers tion. (A) Thymus with mild stress involution due to endogenous
characteristic of thymus from patients with MG occur within the corticosteroids has a characteristic “starry sky” appearance.
perivascular space, outside of the thymic epithelial network. C, The pale areas result from macrophage clearance of apoptotic
cortex; M, medulla; P, perivascular space. (Scanning power.) thymocytes. (Low-power magnification; inset, medium-power
magnification.) (B) Treatment with high-dose corticosteroids
results in depletion of cortical thymocytes and increased
numbers of medullary Hassall bodies. (Medium-power
infant).37,38 Severe combined immunodeficiency magnification.)
thymus consists of compact lobules of tightly op-
posed thymic epithelial cells surrounded by a small
amount of adipose tissue (Fig 9A). Concentri- ciencies such as severe combined immunodefi-
cally arranged thymic epithelial cells (epithelial ciency. However, this term is confusing because
“pseudo-rosettes”) may be present focally (Fig 9B). although the thymus is hypoplastic and has not
CD1a⫹ small lymphocytes (ie, immature thymo- developed normally, it does not represent a pre-
cytes) are absent, although CD3⫹ cells may some- neoplastic condition. The histologic picture of
times be observed. Corticomedullary distinction post-natal thymic atrophy in association with immu-
and Hassall bodies are typically absent. The term nodeficiency differs from that of the primary thy-
“thymic dysplasia” has been used to refer to the mic hypoplasia observed in severe combined immu-
histology of the thymus in primary immunodefi- nodeficiency. In post-natal thymic atrophy, the

Figure 9. Primary thymic hypoplasia


versus post-natal thymic atrophy. The
thymus from patients with severe com-
bined immunodeficiency and primary
failure of thymic development (A, scan-
ning power) is small and composed
predominately of tightly opposed thy-
mic epithelial cells, with minimal adi-
pose tissue infiltration. Epithelial pseu-
dorosettes may be present (arrowhead
in B, medium power). In contrast, thy-
mus from a 2-year-old patient with
post-natal thymic atrophy (C, scanning
power) is large with abundant adipose
tissue and elongated collections of thy-
mic epithelial cells (D, medium power).
58 Laura P. Hale

thymus is typically of near normal size, with prom- studies, biopsies of the thymic allograft are ob-
inent thymic vessels and large amounts of adipose tained 2 to 3 months following transplantation to
tissue (Fig 9C). Epithelial cells are typically ar- assess graft survival and function. The principles
ranged in elongated strands rather than compact for histologic assessment of thymic function de-
lobules, with absence of CD1a⫹ mib-1⫹ developing scribed above have also proved useful for assessing
T cells (Fig 9D). the success of the transplantation procedure, which
The thymus of children without congenital im- has profound implications for prognosis and clini-
munodeficiencies may appear abnormal at autopsy cal care of these patients. Patients whose allograft
because of stress involution or corticosteroid treat- biopsies lack CD1a⫹ mib-1⫹ small lymphocytes and
ment.12,39 Endogenous corticosteroids produced in show condensed thymic epithelium surrounded by
response to the stress of serious illness or therapeu- an infiltrate of CD3⫹ TIA-1⫹ activated cytotoxic T
tic doses of corticosteroids cause apoptosis of im- cells do not develop immunoreconstitution. This
mature thymocytes,40,41 which are cleared by thy- histologic picture is characteristic of impending
mic macrophages. Mild stress involution appears as allograft rejection. In contrast, patients with a light
cleared areas within the normally densely packed lacy cytokeratin network and CD1a⫹ mib-1⫹ small
thymic cortex, the so-called “starry sky” pattern of lymphocytes at biopsy (Fig 11) typically develop
thymic involution (Fig 10A). As involution TREC⫹ peripheral blood cells and subsequent nor-
progresses, the thymic cortex becomes progres- malization of the T-cell repertoire by immunoscope
sively depleted. The number of medullary Hassall analysis.8 Analyses of naive (CD45RA⫹CD62Lhigh)
bodies may also increase in patients treated with T-cell numbers by flow cytometry, TRECs, and
high dose corticosteroids (Fig 10B).12 T-cell repertoire by immunoscope analysis that
show lack of thymus-derived T cells can be used to
Evaluation of Thymic Reconstitution confirm the initial diagnosis of complete DiGeorge
syndrome. Following successful thymus transplan-
Most studies of patients undergoing therapy for tation, thymus-derived T cells appear in the periph-
congenital immunodeficiencies, HIV infection, or eral blood, with later normalization of the T-cell
cancer have used radiographic determination of repertoire.8
thymus size and/or the TREC assay combined with
flow cytometric identification of CD3⫹CD45RA⫹ Summary
CD62Lhigh (naive) T cells to determine thymic
reconstitution status.27,30,33 The immunoscope as- The thymus is absolutely required to initially
say has also been used to monitor normalization of establish a functional T-cell repertoire in humans,
the T-cell receptor repertoire and to identify pa- but begins to undergo age-related decreases in
tients who have been successfully immunoreconsti- activity, beginning in early childhood. Histologic
tuted and thus are no longer at risk for opportu- and molecular methods have been developed to
nistic infections.30,32 Few studies have combined assess thymic activity as a function of age in normal
these assays with histologic examination of thymic humans and in disease states that affect the thymus.
tissue. In cases where thymic biopsies were ob- Although thymic volume and weight remain rela-
tained from two HIV⫹ patients treated with highly tively constant, the portion of the thymus contain-
active antiretroviral therapy, the thymus tissues ing thymic epithelium and participating in produc-
showed histologic evidence for thymopoiesis that tion of new T cells decreases progressively with age.
was concordant with elevated TREC levels and in- The thymic perivascular space, defined as the por-
creased numbers of naive phenotype peripheral tion of the thymus within the thymic capsule but
blood T cells following highly active antiretroviral outside the thymic epithelial network, increases
therapy.28 correspondingly with prominent infiltrates of pe-
The transplantation of allogeneic thymic tissue ripheral lymphocytes and adipose tissue. However,
has been shown to restore normal immune func- despite age-related changes in thymic composition,
tion in some patients with complete DiGeorge syn- thymopoiesis continues in normal human adults
drome and profound immunodeficiency.16 The ef- into at least the eighth decade. Histologic and
ficacy of this treatment is currently being rigorously molecular studies of thymic function have yielded
studied in phase II clinical trials. As part of these insights into the normal regulation of the T-cell
Assessment of Human Thymus 59

Figure 11. Histologic confirmation of


thymopoiesis in a thymic allograft. A
frozen section biopsy of thymus tissue
obtained 3 months after grafting into
the quadriceps muscle of a patient with
DiGeorge syndrome shows robust
thymopoiesis (A). (Hematoxylin-eosin
stain.) Immunoperoxidase staining with
anti-cytokeratin mAbs (B) shows as
light lacy network of thymic epithelial
cells interacting with the CD1a⫹ (C),
mib-1⫹ (D) immature thymocytes that
are abundant in cortical regions of the
graft. (Medium-power magnification.)

repertoire and the pathogenesis of autoimmune 7. Moran CA, Suster S: Germ cell tumors of the mediasti-
diseases. Determination of thymic function can im- num. Adv Anat Pathol 1998;5:1-15
8. Markert ML, Sarzotti M, Ozaki DA, et al: Thymic trans-
prove clinical decision-making in patients undergo-
plantation in complete DiGeorge syndrome: Immunologic and
ing immune reconstitution therapy for congenital safety evaluation in twelve patients. Blood 2003;102:1121-1130
immunodeficiencies, HIV infection, or following 9. Lobach DF, Scearce RM, Haynes BF: The human thymic
cancer chemotherapy. microenvironment: Phenotypic characterization of Hassall’s
bodies with the use of monoclonal antibodies. J Immunol 1985;
Acknowledgment 134:250-257
The author would like to thank Steven Conlon for assistance 10. Lobach DF, Itoh T, Singer KH, et al: The thymic micro-
with figure preparation and Drs Barton F. Haynes and John environment. Characterization of in vitro differentiation of the
Whitesides for providing Figure 4. The data used to prepare IT26R21 rat thymic epithelial cell line. Differentiation 1987;34:
Figure 6B were provided by Drs Dhavel D. Patel, Rebecca H. 50-59
Buckley, Gregory D. Sempowski, and Barton F. Haynes. Dr M. 11. Patel DD, Whichard LP, Radcliff G, et al: Characterization
Louise Markert provided the immunostained sections used to of human thymic epithelial cell surface antigens: Phenotypic
prepare Figure 11. similarity of thymic epithelial cells to epidermal keratinocytes.
J Clin Immunol 1995;15:80-92
References 12. Hale LP, Markert ML: Corticosteroids regulate epithelial
cell differentiation and Hassall body formation in the human
1. Loehrer PJ Sr, Wick MR: Thymic malignancies. Cancer
Treat Res 2001;105:277-302 thymus. J Immunol 2004 (in press)
2. Suster S: Primary large-cell lymphomas of the mediasti- 13. Steinmann GG: Changes in the human thymus during
num. Semin Diagn Pathol 1999;16:51-64 aging. Curr Topics Pathol 1986;75:43-88
3. Weidner N: Germ-cell tumors of the mediastinum. Semin 14. Flores KG, Li J, Sempowski GD, et al: Analysis of the
Diagn Pathol 1999;16:42-50 thymic perivascular space during aging. J Clin Invest 1999;104:
4. Suster S, Moran CA: Primary thymic epithelial neoplasms: 1031-1039
Spectrum of differentiation and histological features. Semin 15. Nehls M, Pfeifer D, Schorpp M, et al: New member of the
Diagn Pathol 1999;16:2-17 winged-helix protein family disrupted in mouse and rat nude
5. Dadmanesh F, Sekihara T, Rosai J: Histologic typing of mutations. Nature 1994;372:103-107
thymoma according to the new World Health Organization 16. Markert ML, Boeck A, Hale LP, et al: Transplantation of
classification. Chest Surg Clin North Am 2001;11:407-420 thymus tissue in complete DiGeorge syndrome. N Engl J Med
6. Moran CA, Suster S. Neuroendocrine carcinomas (carci- 1999;341:1180-1189
noid tumor) of the thymus. A clinicopathologic analysis of 80 17. Steinmann GG, Klaus B, Muller-Hermelink HK: The in-
cases. Am J Clin Pathol 2000;114:100-110 volution of the aging human thymic epithelium is independent
60 Laura P. Hale

of puberty: A morphometric study. Scand J Immunol 1985;22: 30. Klein AK, Patel DD, Gooding ME, et al: T-Cell recovery in
563-575 adults and children following umbilical cord blood transplanta-
18. Sempowski GD, Hale LP, Sundy JS, et al: Leukemia inhib- tion. Biol Blood Marrow Transplant 2001;7:454-4662001
itory factor, oncostatin M, IL-6, and stem cell factor mRNA 31. Patel DD, Gooding ME, Parrott RE, et al: Thymic function
expression in human thymus increases with age and is associated after hematopoietic stem-cell transplantation for the treatment
with thymic atrophy. J Immunol 2000;164:2180-2187 of severe combined immunodeficiency. N Engl J Med 2000;342:
19. Flores KG, Li J, Hale LP: B cells in epithelial and perivas- 1325-1332
cular compartments of human adult thymus. Hum Pathol 2001; 32. Sarzotti M, Patel DD, Li X, et al: T cell repertoire devel-
32:926-934 opment in humans with severe combined immunodeficiency
20. Haynes BF, Hale LP: The human thymus: A chimeric (SCID) after non-ablative allogeneic marrow transplantation.
organ comprised of central and peripheral lymphoid compo- J Immunol 2003;170:2711-2718
nents. Immunol Res 1998;18:175-192 33. Mackall CL, Fleisher TA, Brown MR, et al: Age, thymo-
21. Jamieson BD, Douek DC, Killian S, et al: Generation of poiesis, and CD4⫹ T lymphocyte regeneration after extensive
functional thymocytes in the human adult. Immunity 1999;10: chemotherapy. N Engl J Med 1995;332:143-149
569-575 34. Tamaoki N, Habu S, Kameya T: Thymic lymphoid follicles
22. von Gaudecker B. Ultrastructure of the age-involuted in autoimmune diseases: Histological, histochemical, and elec-
adult human thymus. Cell Tiss Res 1978;186:507-525 tron microscopic studies. Keio J Med 1971;20:57-68
23. Bertho J-M, Demarquay C, Moulian N, et al: Phenotypic 35. Gilquin J, Viard J-P, Jubault V, et al: Delayed occurrence
and immunohistological analysis of the human adult thymus: of Graves’ disease after immune restoration with HAART. Lan-
Evidence for an active thymus during adult life. Cell Immunol cet 1998;352:1907-1908
1997;179:30-40 36. Remes-Troche JM, Tellez-Zenteno JF, Estanol B, et al:
24. Livak E, Schatz D: T-cell receptor ␣ locus V(D) recombi- Thymectomy in myasthenia gravis: Response, complications,
nation byproducts are abundant in thymocytes and mature T and associated conditions. Arch Med Res 2002;33:545-551
cells. Mol Cell Biol 1996;16:609-618 37. Hale LP, Buckley RH, Puck JM, et al: Abnormal develop-
25. Douek DC, McFarland RD, Keiser PH, et al: Changes in ment of thymic dendritic and epithelial cells in human X-linked
thymic function with age and during treatment of HIV infec- severe combined immunodeficiency. Clin Immunol 2004 (in
tion. Nature 1998;396:690-695 press)
26. Sempowski GD, Thomasch JR, Gooding ME, et al: Effect 38. Stocker JT, Dehner LP (eds): Appendix in: Pediatric
of thymectomy in myasthenia gravis on human peripheral blood Pathology (2nd ed). Philadelphia, PA, Lippincott, 2001, pp1435-
T cell pools. J Immunol 2001;166:2808-2817 1447
27. McCune JM, Loftus R, Schmidt DK, et al: High prevalence 39. van Baarlen J, Schuurman H-J, Huber J: Acute thymus
of thymic tissue in adults with human immunodeficiency virus-1 involution in infancy and childhood: A reliable marker for
infection. J Clin Invest 1998;101:2301-2308 duration of acute illness. Hum Pathol 1988;19:1155-1160
28. Markert ML, Alvarez-McLeod AP, Sempowski GD, et al: 40. Wylie AH: Glucocorticoid-induced thymocyte apoptosis is
Thymopoiesis in HIV-infected adults after highly active antiret- associated with endogenous endonuclease activation. Nature
roviral therapy. AIDS Res Hum Retroviruses 2001;17:1635-1643 1980;284:555-556
29. Markert ML, Hicks CB, Bartlett JA, et al: Effect of HAART 41. Wang W, Wykrzykowska J, Johnson T, et al: A NF-␬B/c-
and thymic transplantation on immunoreconstitution in HIV myc-dependent survival pathway is targeted by corticosteroids in
infection. AIDS Res Hum Retroviruses 2000;16:403-413 immature thymocytes. J Immunol 1999;162:314-322

Das könnte Ihnen auch gefallen