Cesta 2006

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Toxicologic Pathology http://tpx.sagepub.

com/

Normal Structure, Function, and Histology of Mucosa-Associated Lymphoid Tissue


Mark F. Cesta
Toxicol Pathol 2006 34: 599
DOI: 10.1080/01926230600865531

The online version of this article can be found at:


http://tpx.sagepub.com/content/34/5/599

Published by:

http://www.sagepublications.com

On behalf of:

Society of Toxicologic Pathology

Additional services and information for Toxicologic Pathology can be found at:

Email Alerts: http://tpx.sagepub.com/cgi/alerts

Subscriptions: http://tpx.sagepub.com/subscriptions

Reprints: http://www.sagepub.com/journalsReprints.nav

Permissions: http://www.sagepub.com/journalsPermissions.nav

>> Version of Record - Aug 1, 2006

What is This?

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


Toxicologic Pathology, 34:599–608, 2006
Copyright 
C by the Society of Toxicologic Pathology

ISSN: 0192-6233 print / 1533-1601 online


DOI: 10.1080/01926230600865531

Normal Structure, Function, and Histology of Mucosa-Associated


Lymphoid Tissue
MARK F. CESTA

Integrated Laboratory Systems, Inc., Durham, North Carolina 27713, USA

ABSTRACT
The mucosa-associated lymphoid tissue (MALT) initiates immune responses to specific antigens encountered along all mucosal surfaces. MALT
inductive sites are secondary immune tissues where antigen sampling occurs and immune responses are initiated. Effector sites, present as diffuse
lymphoid tissue along all mucosal surfaces are the sites of IgA transport across the mucosal epithelium. Though there are many differences between
inductive sites in various organs, they all contain the same basic compartments—follicles, interfollicular regions, subepithelial dome regions, and
follicle-associated epithelium. The morphologic differences between MALT and other secondary lymphoid tissues, between the MALT sites of differing
anatomic locations, and species differences among laboratory animals are described. The morphologic changes in MALT associated with aging, route
of nutrition, and genetic mutation (i.e., the nude and SCID mutations) are also discussed. MALT tissues comprise the mucosal immune system which
can function independently of the systemic immune system and are, therefore, an important and often overlooked aspect of immunopathology.
Keywords. BALT, GALT, immunopathology, lab animal, MALT, morphology, mucosal immune system, NALT.

INTRODUCTION disseminated lymphoid tissue diffusely distributed through-


About half the lymphocytes of the immune system are in out the lamina or substantia propria (Yan et al., 2003). In ef-
the Mucosa-associated lymphoid tissue (MALT) (Croitoru fector sites, secretory IgA, or S-IgA (2 IgA molecules joined
and Bienenstock, 1994). MALT is situated along the surfaces by a J-chain and bound to secretory component, an epithelial
of all mucosal tissues. Its most well-known representatives cell membrane receptor) is secreted across the mucosal ep-
are gut-associated lymphoid tissue (GALT), nasopharynx- ithelium (Pabst, 1987). In rodents, hepatocytes also produce
associated lymphoid tissue (NALT), and bronchus-associated secretory component, so S-IgA also enters the gut lumen via
lymphoid tissue (BALT); however, conjunctiva-associated the bile duct (Pabst, 1987). The cellular composition of effec-
lymphoid tissue (CALT), lacrimal duct-associated (LDALT), tor sites includes T-cells, the majority of which are CD4+ , IgA
larynx-associated (LALT) and salivary duct-associated lym- plasma cells with fewer IgG and IgM plasma cells, and few B-
phoid tissue (DALT) have also been described. The main cells, dendritic cells, and macrophages (Pabst, 1987; Kelsall
function of MALT is to produce and secrete IgA across mu- and Strober, 1999; MacDonald, 2003). Though MALT sites
cosal surfaces in antigen specific, Th 2-dependent reactions, are anatomically separated, they are functionally connected
though Th 1 and cytotoxic T-cell mediated reactions can also in what has been termed the “common mucosal immune sys-
occur, the later resulting in immunotolerance (Gormley et al., tem,” so that antigen presentation and B-cell activation at one
1998; Kiyono and Fukuyama, 2004). mucosal site can result in IgA secretion at mucosal sites of
MALT can be functionally divided into effector sites and different organs (Bienenstock et al., 1999; Hiroi et al., 1998;
inductive sites. GALT, BALT and NALT, CALT in mice, dogs, Kiyono and Fukuyama, 2004; Kuper et al., 2002). Further-
(Giuliano et al., 2002) and baboons (Astley et al., 2003), and more, the mucosal immune system can act independently of
DALT in cynomolgus macaques (Nair and Schroeder, 1986; the systemic immune system making evaluation of MALT an
Sakimoto et al., 2002) are inductive sites. Inductive sites con- important aspect of immunopathology (Kuper et al., 2002).
tain secondary lymphoid tissues in which IgA class switching Intraepithelial lymphocytes, T-lymphocytes found amid
and clonal expansion of B-cells occurs in response to anti- epithelial cells of all mucosal tissues, are another component
gen specific T-cell activation. After activation and IgA class of MALT. They are predominantly CD8+ and are relatively
switching, T- and B-cells migrate from inductive sites to ef- abundant, typically with 1 lymphocyte per 4-6 epithelial cells
fector sites. Effector sites are present in all mucosal tissues as (Kelsall and Strober, 1999; Kuper et al., 2002; MacDonald,
2003; Pabst et al., 2005). They are somewhat unique, par-
ticularly in rodents, in that a large proportion of them ex-
press the γ δ TCR and many express the CD8αα homod-
Address correspondence to: Mark F. Cesta, Integrated Laboratory Sys-
tems, Inc., 601 Keystone Park Drive, Suite 100, Durham, North Carolina imeric form of the CD8 receptor rather than the αβ TCR and
27713, USA; e-mail: mcesta@ils-inc.com CD8αβ heterodimer found in most other CD8+ T-cell popu-
This work was supported by NIEHS contracts N01ES35513 and lations (Eberl and Littman, 2004; Kelsall and Strober, 1999;
N01ES95435. The author wishes to acknowledge the assistance of the staffs Pabst et al., 2005; Saito et al., 1998). They also uniquely ex-
of Integrated Laboratory Systems Inc., Experimental Pathology Laborato- press the αE β7 integrin, which is thought to be important
ries Inc., and the National Institute of Environmental Health Sciences with
preparation of photographs included in this paper. for sequestering these lymphocytes in the epithelium (Kel-
This research was supported in part by the Intramural Research Program sall and Strober, 1999; MacDonald, 2003). Evidence suggests
of the NIH, National Institute of Environmental Health Sciences. that these cells are generated in extrathymic locations in the
599

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


600 CESTA TOXICOLOGIC PATHOLOGY

FIGURE 1.—Peyer’s Patch, small intestine; Sprague–Dawley rat, male, 31 days old. General structure of a Peyer’s patch with a centrally located follicle (F)
containing a germinal center (GC) flanked by parafollicular or interfollicular regions (IFR). The germinal center is composed of a basal dark zone and an apical
light zone with the mantle zone or corona (C) of the follicle superficial to the light zone. The follicle-associated epithelium (FAE) is separated from the follicle by
the subepithelial dome region (SED). 2.—Peyer’s Patch, small intestine; Sprague–Dawley rat, male, 31 days old. The follicle-associated epithelium (FAE) contains
clusters of lymphocytes which may be associated with an M-cell. It is also slightly attenuated and has fewer goblet cells relative to the villous epithelium (VE).
The subepithelial dome region (SED) contains lymphocytes, macrophages, and dendritic cells. C = corona or mantle zone. 3.—Peyer’s patch, small intestine;

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


Vol. 34, No. 5, 2006 MUCOSA-ASSOCIATED LYMPHOID TISSUE 601

intestinal mucosa, but this remains controversial (Saito et al., (Haley, 2003). B-cell areas are larger than T-cell areas, which
1998). is reflected in the T cell:B cell ratio of 0.2 (Sminia and Kraal,
This paper focuses on the normal morphology of the induc- 1999). The size, number, distribution, and composition of
tive sites of BALT, GALT, and NALT, and these terms will Peyer’s patches may vary depending on species or strain. For
be used to refer to these secondary lymphoid tissues. The example, Peyer’s patches are generally smaller in Fischer
functional compartments of these tissues are the lymphoid 344 rats than in Wistar rats (Bruder et al., 1999). The ratio of
follicles, the interfollicular region, the subepithelial dome re- CD4+ to CD8+ T-cells of 5.0 is approximately 2-fold higher
gion, and the overlying follicle-associated epithelium, or lym- in Peyer’s patches than in NALT or BALT (2.4 and 2.6, re-
phoepithelium, which contains M-cells (Figure 1). M-cells spectively) (Sminia and Kraal, 1999). In Lewis rats, however,
are specialized epithelial cells within the follicle-associated the ratios are nearly equal (Kuper et al., 1992).
epithelium that transport microorganisms, and macro- and Isolated lymphoid follicles, also located on the antimesen-
soluble molecules from the intestinal lumen to the subepithe- teric border of the small intestine, have been identified in
lial dome region giving the lymphoid tissues access to lumi- the rat, mouse, rabbit, and guinea pig (Hamada et al., 2002;
nal antigens (Gebert and Pabst, 1999; Kelsall and Strober, Lorenz and Newberry, 2004; Pabst et al., 2005). They are
1999). They are difficult to distinguish light microscopically smaller than Peyer’s patches with an average diameter of 150
but have a distinct ultrastructural appearance. The apical sur- microns and appear as barrel-shaped lymphoid aggregates
face of M cells is characterized by small, irregular, disor- in shortened intestinal villi and are typically associated with
ganized microvilli with a poorly developed brush border or a single dome (Pabst et al., 2005). They are very similar to
microfolds rather than the dense brush border of absorptive Peyer’s patches having 1-2 B-cell follicles that may contain
enterocytes (Gebert and Pabst, 1999; Kelsall and Strober, germinal centers and a small population of CD4+ T-cells,
1999). The basal membrane of M cells is invaginated, form- an overlying follicle-associated epithelium with M-cells, and
ing a pocket that typically contains one or more lymphocytes scattered dendritic cells with few macrophages, (Lorenz and
(T-cells or B-cells) and occasionally macrophages (Gebert Newberry, 2004; Pabst et al., 2005). Up to 200 isolated lym-
and Pabst, 1999; Pabst, 1987). Since there are no specific phoid follicles per mouse have been identified, though this
histochemical or immunohistochemical markers for M cells, number may vary depending on mouse strain. Hamada et al.
clusters of lymphocytes in the follicle-associated epithelium (2002) identified 150–200 per mouse in BALB/cA/Jcl and
may be the only light microscopic clue to the presence of an M 100–150 per mouse in C57BL/6J/Jcl. Isolated lymphoid fol-
cell. Other common features of MALT include the lack of af- licles are not present in neonatal mice, becoming consis-
ferent lymphatics and medullary regions and the presence of tently detectable in the duodenum and proximal jejunum
high endothelial venules (Figure 5) within the interfollicular by light microscopy at 7 days of age in BALB/cA/Jcl and
regions. The cellular components of MALT include B-cells, at 25 days of age in C57BL/6J/Jcl (Hamada et al., 2002).
CD4+ and CD8+ T-cells, antigen-presenting dendritic cells, According to Lorenz and Newberry, C57BL/6 mice have
macrophages, and, occasionally, mast cells and eosinophils fewer and smaller isolated lymphoid follicles that are located
in the interfollicular region. Thus, they contain all the cell predominantly in the distal small intestine and are not con-
types necessary to initiate an immune response. fined to the antimesenteric border (Lorenz and Newberry,
2004).
Cryptopatches, lymphoid aggregates found in the inter-
GUT-ASSOCIATED LYMPHOID TISSUE cryptal lamina propria of the small intestine, are small ag-
Several types of lymphoid nodules have been described gregates of T-cells and dendritic cells with an average diam-
in the intestine, including Peyer’s patches, isolated lymphoid eter of 80 µm (Eberl and Littman, 2004; Mowat and Viney,
follicles, cryptopatches, and, in the large intestine, lymphog- 1997; Pabst et al., 2005). They form after weaning and are far
landular complexes. Peyer’s patches and lymphoglandular more numerous than isolated lymphoid follicles and Peyer’s
complexes are the primary inductive sites in the gut, but the patches with up to 1700 per adult mouse (Eberl and Littman,
functions of the isolated lymphoid follicles and cryptopatches 2004). Studies suggest that cryptopatches are primary lym-
are unclear. phoid tissue in which extrathymic generation of intraepithe-
Peyer’s patches (Figures 1–5) are randomly distributed lial lymphocytes occurs, though this remains controversial
throughout the mucosa and submucosa of the gastrointesti- (Eberl and Littman, 2004; Saito et al., 1998).
nal tract but are of greatest density in the jejunum and are Lymphoglandular complexes (Figures 6 and 7) in the
oriented along the anti-mesenteric border (Schuurman et al., colon resemble Peyer’s patches, however, they are smaller
1994). The Peyer’s patch follicles in rodents typically con- and have fewer follicles with smaller germinal centers
tain 6-12 basally located germinal centers, which are more (Owen et al., 1991). Also, crypts extending into the colonic
numerous in Peyer’s patches than in either NALT or BALT submucosa that are lined by follicle-associated epithelium
←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−
Sprague–Dawley rat, male, 31 days old. Germinal center of a Peyer’s patch follicle with apoptotic lymphocytes and tingible body macrophages. Germinal centers
also contain follicular dendritic cells, B-cell centrocytes and centroblasts, and few CD4+ T-cells. 4.—Peyer’s patch, interfollicular region; Sprague–Dawley rat,
male, 31 days old. The interfollicular region of a Peyer’s patch contains CD4+ T-cells, macrophages, interdigitating dendritic cells, few B-cells and plasma cells,
and high endothelial venules (H). 5.—Peyer’s patch, high endothelial venule; Sprague–Dawley rat, male, 31 days old. High magnification view of a high endothelial
venule, important in lymphocyte trafficking—note the lymphocytes attached to the endothelial cell surface. 6.—Lymphoglandular Complex. The general structure
is similar to a Peyer’s patch in the small intestine, but there is an invaginated crypt (IC) lined by goblet cell-poor follicle-associated epithelium in the interfollicular
region (IFR). FAE = follicle-associated epithelium, SED = subepithelial dome region, F = follicle.

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


602 CESTA TOXICOLOGIC PATHOLOGY

FIGURE 7.—Lymphoglandular complex, colon; Sprague–Dawley rat, male, 31 days old. The follicle-associated epithelium (FAE) of the colon contains even more
lymphocytes than that of the small intestine (Figure 2). Note the decreased numbers of goblet cells relative to the crypt epithelium (CE). Here, the subepithelial
dome region (SED) is difficult to distinguish from the corona (C). 8.—NALT; Sprague–Dawley rat, male, 31 days old. Location of NALT on the lateroventral floor
of the proximal nasopharyngeal duct (ND). NS = nasal septum, P = hard palate, ET = ethmoid turbinates. 9.—NALT; Sprague–Dawley rat, male, 31 days old. A
well-defined NALT follicle (F). The overlying follicle-associated epithelium (FAE) is attenuated and contains no goblet cells and fewer cilia relative to the respiratory
epithelium (RE). Several high endothelial venules (H) are present in the interfollicular region (IFR) at the edge of the follicle. A nerve (N) is present within the IFR.
ND = nasopharyngeal duct, P = hard palate. 10.—NALT; Sprague–Dawley rat, male, 31 days old. Germinal center (GC) within a NALT follicle. The subepithelial

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


Vol. 34, No. 5, 2006 MUCOSA-ASSOCIATED LYMPHOID TISSUE 603

and surrounded by lymphoid tissue may occasionally be passages at the entrance to the nasopharyngeal duct (Figure 8)
evident (Figure 6) (Owen et al., 1991). In the mouse distal (Spit et al., 1989). It is visible in the caudal portions of level
colon, lymphoglandular complexes are randomly distributed II and throughout level III of the nasal cavity as sectioned for
with an average of 1.4 patches per centimeter of colon toxicological studies performed by the National Toxicology
(Owen et al., 1991). Lymphoglandular complexes in the Program (Herbert and Leininger, 1999). NALT is consid-
proximal colon of the mouse and throughout the colon in ered the rodent equivalent to Waldeyer’s ring, the oro- and
the rat are oriented toward the antimesenteric border (Deasy nasopharyngeal lymphoid tissues (tonsils) found in humans
et al., 1983; Morfitt and Pohlenz, 1989; Perry and Sharp, and some other species (Heritage et al., 1997). Though simi-
1988). In mice, at least one lymphoglandular complex, the lar in appearance, there are a number of differences between
rectal patch, can consistently be found within 10 mm of the NALT and Peyer’s patches. There are fewer intraepithelial
anus (Owen et al., 1991). The cecal patches, large aggregates lymphocytes In NALT (Sminia and Kraal, 1999). The rela-
of lymphoid follicles in the proximal cecum opposite the tive sizes of the B- and T-cell areas are roughly equal to each
ileocecal valve, also have a consistent location (Owen et al., other in NALT, which is reflected in the higher T:B cell ratio
1991). In Fisher rats, the “proximal colonic lymphoid tissue” of 0.9 (Sminia and Kraal, 1999). Plasma cells are present pre-
is a lymphoid nodule that is consistently present in the dominantly in the connective tissues deep to the NALT (i.e.,
submucosa of the proximal colon roughly at the distal end away from the nasal passage) (Sminia and Kraal, 1999). The
of the first fifth of the colon (Crouse et al., 1989). efferent lymphatic vessels and high endothelial venules ex-
Dogs have a total of 26–29 Peyer’s patches (HogenEsch tend deep into the interfollicular region to the follicular mar-
and Hahn, 2001). They have two types of Peyer’s patches, as gins, the former confined to the basilar portion (Bienenstock
opposed to rats and mice that have uniform Peyer’s patches and McDermott, 2005; Kuper et al., 2002; Sminia and Kraal,
(Haley, 2003). In the jejunum and upper ileum, the Peyer’s 1999). Though macrophages and dendritic cells are scattered
patches of dogs are smaller and more discrete (similar to those throughout, dendritic cells in the subepithelial dome region
of mice and rats), while in the terminal ileum, there is a 26–30 are less numerous than in Peyer’s patches (Sminia and Kraal,
cm long Peyer’s patch that completely encircles the distal 1999). There are no significant species differences in NALT,
6–10 cm of the ileum and narrows proximally to a 1-cm-wide except that NALT in nonhuman primates is more extensive
band on the antimesenteric border (Haley, 2003; HogenEsch than in rodents, extending to the lateral surface of the nasal
and Hahn, 2001). The ileal Peyer’s patch has small domes and cavity (Haley, 2003).
interfollicular regions and an inconspicuous corona relative
to those of the duodenum and jejunum (HogenEsch and Fels- Bronchus-Associated Lymphoid Tissue
burg, 1992). The duodenal Peyer’s patches differ in that they There is a great deal of species variability in BALT. For
have intrafollicular invaginations of the dome epithelium example, BALT is normally absent in dogs, cats, and Syrian
(HogenEsch and Felsburg, 1992). The dome regions in dogs hamsters (Brownstein et al., 1980; Pabst and Gehrke, 1990).
contain more plasma cells than the dome regions in rats or Rabbits typically have the most BALT in regard to the num-
mice (Haley, 2003). Dogs have pinpoint to >2 mm diameter ber of BALT sites, followed by rats, guinea pigs, and mice
lymphoid nodules throughout the gastric lamina propria, (Pabst and Gehrke, 1990). BALT is absent in germ-free pigs,
most numerous in the fundic region, but these are not asso- while germ-free rats have BALT, though much less than in
ciated with a follicle-associated epithelium (HogenEsch and their conventionally reared counterparts (Bienenstock et al.,
Hahn, 2001; Kolbjornsen et al., 1994). In rhesus macaques, 1973; Pabst and Gehrke, 1990). The situation in mice (and
ileal Peyer’s patches are larger than those in the jejunum, duo- humans) is somewhat more controversial. There are conflict-
denum, or colon (Veazey et al., 1997). In comparison to rat ing reports on the presence of BALT in germ-free mice (i.e.,
Peyer’s patches, those of Baboons are smaller and they lack in the absence of antigenic stimulation). Bienenstock and
the IgA+ centroblasts associated with the high endothelial McDermott have reported the presence of BALT in germ-
venules as described by Spencer et al. (Spencer et al., 1986). free mice (Bienenstock and McDermott, 2005; Bienenstock
Rabbits are unique in that they have an appendix, represented et al., 1999), but others report that BALT is not present in
as a large collection of lymphoid nodules at the ileocecal germ-free in mice (Moyron-Quiroz et al., 2004; Seymour
valve, as well as a sacculus rotundus, which is a large, et al., 2006). Furthermore, Pabst and Gehrke reported the
circumferential Peyer’s patch in the terminal ileum (Haley, presence of BALT in only 43% of 4-month-old SPF mice
2003). (Pabst and Gehrke, 1990), so the presence of BALT in mice
in general appears to be rather variable. Regardless of this is-
Nasopharynx-Associated Lymphoid Tissue sue, BALT can be induced in mice by exposure to pathogens,
NALT (Figures 8–12), found in rats, mice, hamsters, and and this BALT, referred to as inducible BALT, or iBALT, by
non-human primates, is composed of paired lymphoid aggre- some, has characteristics similar to BALT in other animals
gates in the caudoventral portion of the left and right nasal (Moyron-Quiroz et al., 2004).
←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−
dome region (SED) is often inapparent and, as shown here, may contain numerous lymphocytes extending from the corona (C). ND = nasopharyngeal duct, RE =
respiratory epithelium, IFR = interfollicular dome region, H = high endothelial venule, F = follicle. 11.—NALT; Sprague–Dawley rat, male, 31 days old. High
endothelial venules (H) in the interfollicular region (IFR) of NALT at the margin of a follicle (F). NALT high endothelial venules more frequently extend to the
margin of the follicle than those of Peyer’s patches or BALT. N = nerve, P = hard palate. 12.—Comparison of follicle-associated epithelium of NALT (top) with
adjacent respiratory epithelium of the nasopharyngeal duct (bottom); Sprague–Dawley rat, male, 31 days old. The follicle-associated epithelium is attenuated, lacks
goblet cells, and has fewer cilia.

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


604 CESTA TOXICOLOGIC PATHOLOGY

FIGURE 13.—BALT nodules along a primary bronchiole; Sprague–Dawley rat, male, 31 days old. The nodules are randomly distributed along the airways, but
there is a predilection for branching points. The nodules are typically located between the airway and an artery. 14.—BALT nodule; Sprague–Dawley rat, male, 31
days old. Submucosal lymphoid nodule with patchy overlying follicle-associated epithelium (FAE) lacking goblet cells and cilia. The interfollicular region (IFR) is
identified by the presence of high endothelial venules (H) but is otherwise difficult to distinguish from the follicle. A nerve (N) runs through each BALT nodule,
though it may not be present in all sections. SED = subepithelial dome region, A = alveolus, L = bronchiolar lumen, RE = respiratory epithelium, SM = smooth
muscle. 15.—BALT nodule; Sprague–Dawley rat, male, 31 days old. Higher magnification view of high endothelial venules (H) in the interfollicular region (IFR).

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


Vol. 34, No. 5, 2006 MUCOSA-ASSOCIATED LYMPHOID TISSUE 605

BALT (Figures 13–16) is randomly distributed along the epithelium, which separates islands of reticular epithelium,
airways but is most consistently located at sites of bronchial is stratified squamous in the oropharynx (palatine and lin-
tree bifurcation and is usually located between a bronchus gual tonsils) and respiratory in the dorsal and some lateral
and an artery (Figure 13). Of the 3 main MALT sites, surfaces of the nasopharynx (pharyngeal and tubal tonsils).
BALT is the most divergent. The follicle-associated epithe- The palatine tonsil is located in the palatine fossa, partially
lium of BALT contains the fewest intraepithelial lymphocytes covered by the semilunar fold.
(Sminia and Kraal, 1999). Germinal centers and tingible body
macrophages are less common in BALT than either NALT or
Peyer’s patches (Sminia and Kraal, 1999). Follicular dendritic NONPATHOLOGIC FACTORS AFFECTING
cells, identified by ED5 staining, have been detected in GALT MALT MORPHOLOGY
and NALT, but not BALT (Kuper et al., 1992). The divisions Age
between the B- and T-cell areas are less conspicuous than In general, MALT is relatively undeveloped at birth with
in Peyer’s patches and NALT and, in the rat, there does not low cellularity. Upon stimulation after birth by exposure to
appear to be any consistent spatial relationship between the environmental antigens, especially the developing gut mi-
two regions nor to surrounding structures (e.g., bronchus or croflora, T-cell areas expand and become fully populated and
artery) (Breel et al., 1988; Sminia and Kraal, 1999). Similari- follicular germinal centers develop (as previously stated, ger-
ties do exist, however. For example, the relative sizes of the B- minal centers are frequent in GALT, but relatively infrequent
and T-cell areas in BALT are roughly equal, and the T:B cell in NALT and BALT). There are several differences in the
ratio of 0.7 is comparable to that of NALT (Sminia and Kraal, ontogeny of the MALT sites. In fetal animals, BALT is gen-
1999). The general structure and cellular composition of erally absent, but is present by 4 days of age, initially as a
BALT is also similar to NALT (Bienenstock and McDermott, few lymphocytes within a reticular stroma (Hameleers et al.,
2005). 1989; Pabst and Gehrke, 1990; Sminia and Kraal, 1999). In
contrast, Peyer’s patches are present before birth and NALT
Tonsils is present at birth (Sminia and Kraal, 1999). T- and B-cell
Tonsils are secondary lymphoid organs located in the oro- regions do not develop in BALT until 3-4 weeks of age,
and nasopharynx of most species except rodents. Dogs have 4 whereas distinct B- and T-cell areas are discernible at 10
sets of tonsils, the lingual (diffuse lymphoid tissue at the cau- days of age in both NALT and GALT (Hameleers et al.,
dodorsal base of the tongue), palatine (on the lateral wall of 1989; Sminia and Kraal, 1999). Last, Peyer’s patches and
the pharynx just caudal to the palatoglossal arch), pharyngeal NALT are initially composed of T-cells, whereas B-cells are
(on the roof of the nasopharynx), and tubal (at the openings the first lymphocytes to populate BALT (Sminia and Kraal,
of the auditory tubes) tonsils. Primates have at least 3 sets of 1999).
tonsils (in addition to NALT as described above), the lingual, Though much of the work on the effects of aging on
palatine, and pharyngeal, and may have tubal tonsils (as do the mucosal immune system has focused on changes in
humans and all other domestic animals except rodents), but antibody production, evidence suggests that aging also al-
descriptions of tubal tonsils in these species are lacking. Two ters lymphocyte proliferative capacity (Taylor et al., 1992;
types of tonsils have been described, those with crypts (fol- Schmucker et al., 2001; Schmucker, 2002). With age, the
licular tonsils) and those without. Tonsilar crypts are blind, number of Peyer’s patches does not change significantly,
often branched invaginations of the surface epithelium into however, in aged mice, but not rats, the Peyer’s patches do
the submucosal lymphoid tissue. Tonsils without crypts typ- have fewer T-cells in the interfollicular and parafollicular
ically have a slightly folded surface epithelium and bulge regions relative to younger animals (Kawanishi and Kiely,
into the oro-or nasopharynx (Banks, 1993). They are struc- 1989; Schmucker, 2002; Schmucker et al., 2001). The num-
turally similar to Peyer’s patches but differ in the overlying ber of antibody-containing plasma cells in the Peyer’s patches
epithelium. of mice and lamina propria of rats intraduodenally immunized
There are two types of epithelium overlying the tonsils: with cholera toxin decreases with age (Haaijman et al., 1977;
reticular and non-reticular (Belz and Heath, 1995). Reticu- Taylor et al., 1992). There may also be decreased numbers of
lar epithelium is spongy in appearance and is located over B-cells and changes in the distribution of lymphocyte sub-
the apices of lymphoid follicles (Belz and Heath, 1995). It sets in the intestine, but evidence regarding changes in the
contains M-cells, numerous lymphocytes, macrophages and number of dendritic cells is conflicting (Schmucker, 2002;
dendritic cells, and may contain a few granulocytes (Belz Schmucker et al., 2001). None of these studies, however, re-
and Heath, 1995; Bernstein et al., 1999). The non-reticular ported detectable, light microscopic changes, so even though

←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−
L = bronchiolar lumen, SM = smooth muscle, SED = subepithelial dome region. 16.—Comparison of the follicle-associated epithelium of BALT (top) with the
bronchial respiratory epithelium (bottom); Sprague–Dawley rat, male, 31 days old. The attenuated follicle-associated epithelium (FAE) is patchy and contains no
goblet or ciliated cells but does contain M cells, though they are not readily apparent with light microscopy. RE = respiratory epithelium, SED = subepithelial dome
region, SM = smooth muscle. 17.—Peyer’s patch, small intestine; BALB/cA nude mouse, 8 weeks old. The follicle (F) is well developed and contains numerous
B-cells, but high endothelial venules are not apparent in the interfollicular region (IFR) and lymphocytes numbers are decreased in the IFR, the subepithelial dome
region (SED), and the follicle associated epithelium (FAE). 18.—NALT; BALB/cA nude mouse, 8 weeks old. As with the PP in Figure 17, the follicular region (F)
is well developed, but the interfollicular region (IFR) is largely devoid of lymphocytes and high endothelial venules. FAE = follicle associated epithelium, RE =
respiratory epithelium, ND = nasopharyngeal duct, P = hard palate.

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


606 CESTA TOXICOLOGIC PATHOLOGY

FIGURE 19.—NALT; NIH nude rat, 8 weeks old. NALT in nude rats is very similar to NALT in nude mice (Figure 18), but there are slightly more lymphocytes
in the interfollicular region (IFR). As in nude mice, the follicular region (F) and the follicle associated epithelium (FAE) are well developed. RE = respiratory
epithelium, ND = nasopharyngeal duct, P = hard palate. 20.—BALT; NIH nude rat, 8 weeks old. Similar to other MALT structures in nude mice and rats with
a well developed follicle (F) and follicle associated epithelium (FAE) and an interfollicular region (IFR) depleted of lymphocytes and high endothelial venules.
RE = respiratory epithelium. 21.—Peyer’s Patch, small intestine; CB-17 SCID mouse, 8 weeks old. Both the follicular region and interfollicular region are poorly
developed, difficult to distinguish from each other, and contain relatively few lymphocytes. The follicle associated epithelium (FAE) is proportionately decreased
in size. SED = subepithelial dome region. 22.—NALT; CB-17 SCID mouse, 8 weeks old. The follicular region and the interfollicular region are largely devoid of
lymphocytes and are poorly delineated. The follicle associated epithelium (FAE) is decreased in size. RE = respiratory epithelium, ND = nasopharyngeal duct.

cell numbers may be decreased, whether or not this is de- Route of Nutrition
tectable morphologically is unknown. Administration of total parenteral nutrition (TPN) has been
In dogs, Peyer’s patch precursors can be seen as small shown to affect GALT, decreasing lymphocyte numbers in
aggregates of lymphocytes in intestinal villi in 45 day old both rats and mice (Tanaka et al., 1991; King et al., 1997).
fetuses, but lymphoglandular complexes first appear in the In mice, both B and T cells in Peyer’s patches and lamina
cecum 1 week after birth (HogenEsch and Hahn, 2001). In propria and the intraepithelial lymphocytes decreased sig-
50-day-old fetuses, the Peyer’s patches are much more well nificantly after 2 days of receiving TPN with the maximal
developed with domes and submucosal follicles, however, decrease in the lamina propria occurring after 2 days and in
germinal centers do not form until 1 week of age (HogenEsch the Peyer’s patches after 3 days (King et al., 1997). These
and Hahn, 2001). The ileal Peyer’s patch expands rapidly after Peyer’s patches and lamina propria changes were shown to
birth, reaching maximal size by 6 months of age (HogenEsch be reversible in mice after returning to enteral nutrition and
and Hahn, 2001). Upon reaching sexual maturity, the follicles returned to control levels after 2 days (King et al., 1997). The
of the canine ileal Peyer’s patch become markedly reduced intraepithelial lymphocytes responded much more slowly and
in size (HogenEsch and Hahn, 2001). by day 5, had not yet returned to normal levels (King et al.,

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


Vol. 34, No. 5, 2006 MUCOSA-ASSOCIATED LYMPHOID TISSUE 607

1997). Histologic evaluation of rats receiving TPN revealed intrapulmonary immunization with sheep erythrocytes. Am J Pathol 98,
markedly atrophied Peyer’s patches and restriction of intraep- 499–514.
ithelial lymphocytes to the basal third of the intestinal villi Bruder, M. C., Spanhaak, S., Bruijntjes, J. P., Michielsen, C. P., Vos, J. G., and
Kuper, C. F. (1999). Intestinal T lymphocytes of different rat strains in
(some intraepithelial lymphocytes were noted in the distal
immunotoxicity. Toxicol Pathol 27, 171–9.
two-thirds of the villi in control rats) (Tanaka et al., 1991). Croitoru, K., and Bienenstock, J. (1994). Characteristics and Functions of
mucosa-associated lymphoid tissue. In Handbook of Mucosal Immunol-
MALT in Immunodeficient Mutant Mice and Rats ogy (P. L. Ogra, J. Mestecky, M. E. Lamm, W. Strober, J. R. McGhee, and
There are a number of immunodeficient mutant mice and J. Bienenstock, eds.), pp. 141–51. Academic Press, San Diego.
rats, both naturally occurring and genetically engineered, in Crouse, D. A., Perry, G. A., Murphy, B. O., and Sharp, J. G. (1989). Character-
istics of submucosal lymphoid tissue located in the proximal colon of the
which the morphology of MALT is altered. In SCID mice rat. J Anat 162, 53–65.
(Figures 21 and 22), which are deficient in both B- and T- Custer, R. P., Bosma, G. C., and Bosma, M. J. (1985). Severe combined immun-
cells, the lymphoid structures of the gut and lung are very odeficiency (SCID) in the mouse. Pathology, reconstitution, neoplasms.
small (almost nonexistent) and disorganized with sparsely Am J Pathol 120, 464–77.
scattered lymphoid cells of variable size, often appearing im- Deasy, J. M., Steele, G., Jr., Ross, D. S., Lahey, S. J., Wilson, R. E., and
mature (Custer et al., 1985). Effector sites are similarly af- Madara, J. (1983). Gut-associated lymphoid tissue and dimethylhydrazine-
fected, as exemplified by the relative lack of immune cells in induced colorectal carcinoma in the Wistar/Furth rat. J Surg Oncol 24, 36–
the lamina propria of the intestine. This phenotype, however, 40.
may vary with background strain and age, as the SCID mu- Eberl, G., and Littman, D. R. (2004). Thymic origin of intestinal alphabeta T cells
tation is “leaky” and these mice may develop more B- and revealed by fate mapping of RORgammat+ cells. Science 305, 248–51.
Gebert, A., and Pabst, R. (1999). M cells at locations outside the gut. Semin
T-cells later in life. Athymic nude rats and mice (Figures 17– Immunol 11, 165–70.
20), being T-cell deficient, have decreased numbers of in- Giuliano, E. A., Moore, C. P., and Phillips, T. E. (2002). Morphological evidence
traepithelial lymphocytes and depleted T-cell regions (inter- of M cells in healthy canine conjunctiva-associated lymphoid tissue. Grae-
follicular/parafollicular regions) (Hanes, 2005; Rocha et al., fes Arch Clin Exp Ophthalmol 240, 220–6.
1994) which, in nude rats at least, have increased numbers of Gormley, P. D., Powell-Richards, A. O., Azuara-Blanco, A., Donoso, L. A., and
macrophages. Also, since the formation of germinal centers Dua, H. S. (1998). Lymphocyte subsets in conjunctival mucosa-associated-
requires T-cell activity, nude rats and mice are typically de- lymphoid-tissue after exposure to retinal-S-antigen. Int Ophthalmol 22,
void of germinal centers (Hanes, 2005). Other components of 77–80.
the lymphoid structures in these animals are variably present. Haaijman, J. J., Schuit, H. R., and Hijmans, W. (1977). Immunoglobulin-
For example, in both SCID and nude animals, the stromal containing cells in different lymphoid organs of the CBA mouse during its
life-span. Immunology 32, 427–34.
cells of the lymphoid structures are comparable in number Haley, P. J. (2003). Species differences in the structure and function of the
and appearance to those of immunocompetent strains (Custer immune system. Toxicology 188, 49–71.
et al., 1985; Hanes, 2005). RAG1−/− mice, a genetically en- Hamada, H., Hiroi, T., Nishiyama, Y., Takahashi, H., Masunaga, Y., Hachimura,
gineered mutant deficient in B- and T-cells similar to SCID S., Kaminogawa, S., Takahashi-Iwanaga, H., Iwanaga, T., Kiyono, H.,
mice, have limited follicle associated epithelium and fewer M Yamamoto, H., and Ishikawa, H. (2002). Identification of multiple isolated
cells, but both are present and associated with small Peyer’s lymphoid follicles on the antimesenteric wall of the mouse small intestine.
patch anlagen (MacDonald, 2003). J Immunol 168, 57–64.
Hameleers, D. M., van der Ende, M., Biewenga, J., and Sminia, T. (1989). An
immunohistochemical study on the postnatal development of rat nasal-
REFERENCES associated lymphoid tissue (NALT). Cell Tissue Res 256, 431–8.
Astley, R. A., Kennedy, R. C., and Chodosh, J. (2003). Structural and cellular ar- Hanes, M. A. (2005). The nude rat. In The Laboratory Rat (M. A. Suckow, S.
chitecture of conjunctival lymphoid follicles in the baboon (Papio anubis). Weisbroth, and C. L. Franklin, eds.), pp. 733–59. Academic Press, San
Exp Eye Res 76, 685–94. Diego.
Banks, W. J. (1993). Applied Veterinary Histology. Mosby Year Book, St. Louis. Herbert, R. A., and Leininger, J. R. (1999). Nose, Larynx, and Trachea. In
Belz, G. T., and Heath, T. J. (1995). The epithelium of canine palatine tonsils. Pathology of the Mouse (R. R. Maronpot, ed.), pp. 259–92. Cache River
Anat Embryol (Berl) 192, 189–94. Press, Vienna.
Bernstein, J. M., Gorfien, J., and Brandtzaeg, P. (1999). The immunobiology of Heritage, P. L., Underdown, B. J., Arsenault, A. L., Snider, D. P., and McDermott,
the tonsils and adenoids. In Mucosal Immunity (O. L. Pearay, J. Mestecky, M. R. (1997). Comparison of murine nasal-associated lymphoid tissue and
M. E. Lamm, W. Strober, J. Bienenstock, and J. R. McGhee, eds.), pp. Peyer’s patches. Am J Respir Crit Care Med 156, 1256–62.
1339–62. Academic Press, San Diego. Hiroi, T., Iwatani, K., Iijima, H., Kodama, S., Yanagita, M., and Kiyono, H.
Bienenstock, J., Johnston, N., and Perey, D. Y. (1973). Bronchial lymphoid (1998). Nasal immune system: distinctive Th0 and Th1/Th2 type envi-
tissue. I. Morphologic characteristics. Lab Invest 28, 686–92. ronments in murine nasal-associated lymphoid tissues and nasal passage,
Bienenstock, J., and McDermott, M. R. (2005). Bronchus- and nasal-associated respectively. Eur J Immunol 28, 3346–53.
lymphoid tissues. Immunol Rev 206, 22–31. HogenEsch, H., and Felsburg, P. J. (1992). Immunohistology of Peyer’s patches
Bienenstock, J., McDermott, M. R., and Clancy, R. L. (1999). Respiratory tract in the dog. Vet Immunol Immunopathol 30, 147–60.
defenses: role of mucosal lymphoid tissues. In Mucosal Immunity (O. L. HogenEsch, H., and Hahn, F. F. (2001). The lymphoid organs: anatomy, devel-
Pearay, J. Mestecky, M. E. Lamm, W. Strober, J. Bienenstock, and J. R. opment, and age-related changes. In Pathobiology of the Aging Dog. (U.
McGhee, eds.), pp. 283–92. Academic Press, San Diego. Mohr, W. W. Carlton, D. L. Dungworth, S. A. Benjamin, C. C. Capen,
Breel, M., Van der Ende, M., Sminia, T., and Kraal, G. (1988). Subpopulations of and F. F. Hahn, eds.), Vol. 1, pp. 127–35. Iowa State University Press,
lymphoid and non-lymphoid cells in bronchus-associated lymphoid tissue Ames.
(BALT) of the mouse. Immunology 63, 657–62. Kawanishi, H., and Kiely, J. (1989). Immune-related alterations in aged gut-
Brownstein, D. G., Rebar, A. H., Bice, D. E., Muggenburg, B. A., and Hill, associated lymphoid tissues in mice. Dig Dis Sci 34, 175–84.
J. O. (1980). Immunology of the lower respiratory tract. Serial morpho- Kelsall, B., and Strober, W. (1999). Gut-associated lymphoid tissue: antigen
logic changes in the lungs and tracheobronchial lymph nodes of dogs after handling and T-lymphocyte responses. In Mucosal Immunity (O. L. Pearay,

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013


608 CESTA TOXICOLOGIC PATHOLOGY

J. Mestecky, M. E. Lamm, W. Strober, J. Bienenstock, and J. R. McGhee, Perry, G. A., and Sharp, J. G. (1988). Characterization of proximal colonic
eds.), pp. 293–317. Academic Press, San Diego. lymphoid tissue in the mouse. Anat Rec 220, 305–12.
King, B. K., Li, J., and Kudsk, K. A. (1997). A temporal study of TPN-induced Rocha, B., Vassalli, P., and Guy-Grand, D. (1994). Thymic and extrathymic
changes in gut-associated lymphoid tissue and mucosal immunity. Arch origins of gut intraepithelial lymphocyte populations in mice. J Exp Med
Surg 132, 1303–9. 180, 681–6.
Kiyono, H., and Fukuyama, S. (2004). NALT- versus Peyer’s-patch-mediated Saito, H., Kanamori, Y., Takemori, T., Nariuchi, H., Kubota, E., Takahashi-
mucosal immunity. Nat Rev Immunol 4, 699–710. Iwanaga, H., Iwanaga, T., and Ishikawa, H. (1998). Generation of intesti-
Kolbjornsen, O., Press, C. M., Moore, P. F., and Landsverk, T. (1994). Lym- nal T cells from progenitors residing in gut cryptopatches. Science 280,
phoid follicles in the gastric mucosa of dogs. Distribution and lymphocyte 275–8.
phenotypes. Vet Immunol Immunopathol 40, 299–312. Sakimoto, T., Shoji, J., Inada, N., Saito, K., Iwasaki, Y., and Sawa, M. (2002).
Kuper, C. F., De Heer, E., van Loveren, H., and Vos, J. G. (2002). Immune Histological study of conjunctiva-associated lymphoid tissue in mouse.
System. In Handbook of Toxicologic Pathology (W. M. Hascheck-Hock, Jpn J Ophthalmol 46, 364–69.
C. G. Rousseaux, and M. A. Wallig, eds.), Vol. 2, pp. 585–644. Academic Schmucker, D. L. (2002). Intestinal mucosal immunosenescence in rats. Exp
Press, New York. Gerontol 37, 197–203.
Kuper, C. F., Koornstra, P. J., Hameleers, D. M., Biewenga, J., Spit, B. J., Dui- Schmucker, D. L., Thoreux, K., and Owen, R. L. (2001). Aging impairs intestinal
jvestijn, A. M., van Breda Vriesman, P. J., and Sminia, T. (1992). The role immunity. Mech Ageing Dev 122, 1397–411.
of nasopharyngeal lymphoid tissue. Immunol Today 13, 219–24. Schuurman, H. J., Kuper, C. F., and Vos, J. G. (1994). Histopathology of the
Lorenz, R. G., and Newberry, R. D. (2004). Isolated lymphoid follicles can immune system as a tool to assess immunotoxicity. Toxicology 86, 187–
function as sites for induction of mucosal immune responses. Ann N Y 212.
Acad Sci 1029, 44–57. Seymour, R., Sundberg, J. P., and HogenEsch, H. (2006). Abnormal lymphoid
MacDonald, T. T. (2003). The mucosal immune system. Parasite Immunol 25, organ development in immunodeficient mutant mice. Vet Pathol 43, 401–
235–46. 23.
Morfitt, D. C., and Pohlenz, J. F. (1989). Porcine colonic lymphoglandular com- Sminia, T., and Kraal, G. (1999). Nasal-associated lymphoid tissue. In Mu-
plex: distribution, structure, and epithelium. Am J Anat 184, 41–51. cosal Immunity (O. L. Pearay, J. Mestecky, M. E. Lamm, W. Strober, J.
Mowat, A. M., and Viney, J. L. (1997). The anatomical basis of intestinal im- Bienenstock, and J. R. McGhee, eds.), pp. 357–64. Academic Press, San
munity. Immunol Rev 156, 145–66. Diego.
Moyron-Quiroz, J. E., Rangel-Moreno, J., Kusser, K., Hartson, L., Sprague, F., Spencer, J., Finn, T., and Isaacson, P. G. (1986). A comparative study of the
Goodrich, S., Woodland, D. L., Lund, F. E., and Randall, T. D. (2004). Role gut-associated lymphoid tissue of primates and rodents. Virchows Arch B
of inducible bronchus associated lymphoid tissue (iBALT) in respiratory Cell Pathol Incl Mol Pathol 51, 509–19.
immunity. Nat Med 10, 927–34. Spit, B. J., Hendriksen, E. G., Bruijntjes, J. P., and Kuper, C. F. (1989). Nasal
Nair, P. N., and Schroeder, H. E. (1986). Duct-associated lymphoid tissue lymphoid tissue in the rat. Cell Tissue Res 255, 193–8.
(DALT) of minor salivary glands and mucosal immunity. Immunology Tanaka, S., Miura, S., Tashiro, H., Serizawa, H., Hamada, Y., Yoshioka, M., and
57, 171–80. Tsuchiya, M. (1991). Morphological alteration of gut-associated lymphoid
Owen, R. L., Piazza, A. J., and Ermak, T. H. (1991). Ultrastructural and cytoar- tissue after long-term total parenteral nutrition in rats. Cell Tissue Res 266,
chitectural features of lymphoreticular organs in the colon and rectum of 29–36.
adult BALB/c mice. Am J Anat 190, 10–8. Taylor, L. D., Daniels, C. K., and Schmucker, D. L. (1992). Ageing compro-
Pabst, O., Herbrand, H., Worbs, T., Friedrichsen, M., Yan, S., Hoffmann, M. W., mises gastrointestinal mucosal immune response in the rhesus monkey.
Korner, H., Bernhardt, G., Pabst, R., and Forster, R. (2005). Cryptopatches Immunology 75, 614–8.
and isolated lymphoid follicles: dynamic lymphoid tissues dispensable for Veazey, R. S., Rosenzweig, M., Shvetz, D. E., Pauley, D. R., DeMaria, M.,
the generation of intraepithelial lymphocytes. Eur J Immunol 35, 98–107. Chalifoux, L. V., Johnson, R. P., and Lackner, A. A. (1997). Characteriza-
Pabst, R. (1987). The anatomical basis for the immune function of the gut. Anat tion of gut-associated lymphoid tissue (GALT) of normal rhesus macaques.
Embryol (Berl) 176, 135–44. Clin Immunol Immunopathol 82, 230–42.
Pabst, R., and Gehrke, I. (1990). Is the bronchus-associated lymphoid tissue Yan, Z., Wang, J. B., Gong, S. S., and Huang, X. (2003). Cell proliferation
(BALT) an integral structure of the lung in normal mammals, including in the endolymphatic sac in situ after the rat Waldeyer ring equivalent
humans? Am J Respir Cell Mol Biol 3, 131–5. immunostimulation. Laryngoscope 113, 1609–14.

Downloaded from tpx.sagepub.com at YALE UNIV on February 26, 2013

You might also like