You are on page 1of 7

Pathophysiology 17 (2010) 337–343

Review

Lung lymphatic anatomy and correlates


Dean E. Schraufnagel ∗
Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine M/C 719, University of Illinois at Chicago,
840 S. Wood St., Chicago, IL 60612-7323, United States
Received 10 March 2009; received in revised form 12 June 2009; accepted 21 October 2009

Abstract
The pulmonary lymphatics do much more than keep the lung dry. They defend the lungs from airborne particles and microbes and allow a
local influx of liquid to clear and clean inflamed or damaged tissue. Lymphatic morphology, especially the three-dimensional structure, is best
demonstrated by casting the lymphatics, corroding the tissue, and viewing the casts by scanning electron microscopy. With this technique,
different lymphatic forms exist. On the pleural surface prelymphatics are simply tissue planes that connect with lymphatic channels. Reservoir
lymphatics are another form of initial lymphatics that have blind-ending pouches. They empty into conduit lymphatics. Lymphatics around
blood vessels and airways are generally tubular and saccular. In the last decade, lymphatic markers have been discovered that allow the study
of lymphatic development in health and disease. Modulators of these pathways could be potential therapeutics for diverse pulmonary problems
such as cancer and lung transplantation. The size of the lymphatic system expands manifold in response to an excess fluid load, cancer, or
inflammation. Immune cells move through the lymphatics, mature, and become activated there. The lymphatics enable the immune defense
system by allowing a sequestered place and close proximity for antigen presentation and lymphocyte maturation.
© 2009 Elsevier Ireland Ltd. All rights reserved.

Keywords: Pulmonary lymphatic system; Corrosion casting; Anatomy and histology

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 337
2. Cast structures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 338
3. Pleural surface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 338
3.1. Initial lymphatics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 338
3.2. Conduit lymphatics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 338
4. Lymphatics within the lung . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 339
4.1. Perivascular and airway lymphatics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 339
5. Markers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341
6. Lymphatic expansion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341
7. Lymph flow . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342
8. Lymph nodes and immune cell trafficking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343

1. Introduction in thickness at its thinnest part. On the tissue side of this


barrier are blood capillaries and scant interstitial space that
The pulmonary alveoli in the normal lung are relatively contains several types of cells mostly of fibroblast and
dry. Alveolar fluid would reduce or prevent efficient gas immune lineage. Each day the human lung absorbs oxygen
exchange across the air–blood barrier, which is about 0.2 ␮m from about 10,000 L of air breathed in. Air contains particu-
lates and other inert and noxious matter. Fine particles may
∗ Tel.: +1 312 996 8039; fax: +1 312 996 4665. react with or pierce through the epithelium. The air also con-
E-mail address: schrauf@uic.edu. tains bacteria and viruses, which may gain a foothold in the

0928-4680/$ – see front matter © 2009 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.pathophys.2009.10.008
338 D.E. Schraufnagel / Pathophysiology 17 (2010) 337–343

lung. The body must rally it cellular defense against infection lie near airways. Pulmonary arteries greater than 100 ␮m in
and move a variety of immune cells into and out of the lung. diameter usually have short branches before giving off cap-
It must present antigens to lymphocytes and allow those cells illaries, which arise at acute angles. Systemic arteries such
to mature into effector cells. The lungs regularly encounter as those of the bronchial circulation, have deeper nuclear
pharyngeal fluid, which may emanate from the stomach with impressions and thicker walls than pulmonary arteries [3].
its acid or from the mouth with its microorganisms. The lung Although the pulmonary blood and lymphatic vascula-
responds to many forms of injury from local or systemic tures have been cast in many species, most of the following
insults by an outpouring of fluid—focal or diffuse pulmonary discussion and all images shows are derived from casts of rat
edema. The interstitial space and thin epithelium are exposed lung. The lymphatic cast structure has not been well studied
to osmotic, hydrostatic, and hydrodynamic forces of the tis- in other species. To expand the lymphatics, pulmonary edema
sues and blood stream, and must adapt to a constant shifting was induced in the rats by exposure to a toxic level of oxygen
of fluid, protein, and cells. All of these challenges are met at or increased lung volume.
least in part by the lung lymphatics.
It is the job of the lung lymphatics to keep the lungs dry
even in the face of a massive fluid challenge, to clear inert 3. Pleural surface
and toxic substances that penetrate the epithelium, and to
quarter, control, and regulate the pulmonary immune system. 3.1. Initial lymphatics
With all this, it is amazing that the alveoli themselves do
not have lymphatics. Lymphatics around the alveoli would The lung interstitial space and the lymphatics that emanate
impede gas exchange and would make gas exchange worse from it can be cast and visualized by scanning electron
in flood states where it would be most needed. When not microscopy. The tissue space leading into the lymphatics
needed, the lymphatics are collapsed and hardly detectable. is sometimes called prelymphatics. The space lies within
When called upon, they swell to many times their resting and just below the visceral pleura fills with fluid in edema
size. conditions. It is exterior to pleural blood capillaries but is
contained by the outer layer of pleura. The prelymphatic tis-
sue planes are marked by connective tissue strands indenting
2. Cast structures a textured surface (Fig. 1). There is no sign of tubularity.
This structural form allows access of the fluid bathing all
Lymphatics are delicate and complex structures that arise tissues to lymphatics. Any substance in the interstitial space
from the tissue spaces. In the absence of disease or stimulus, can be swept into lymphatics; antigens can be trapped, bro-
and until the discovery of specific markers about a decade ken down, and presented to lymphocytes in lymph nodes.
ago, they were nearly unseen by light microscopy and dif- Prelymphatics blend into areas with specific architectural
ficult to detect even with transmission electron microscopy. lace-like structures termed reservoir lymphatics. Reservoir
The clearest visualization of the structure of lung lymphatics lymphatics have a hierarchal shape. They are thin flat struc-
was attained by casting them and viewing the casts with the tures within and beneath the visceral pleura. In contrast to
scanning electron microscopy [1]. Even with this method, it prelymphatic casts, they are not sheets of plastic but have
was necessary to induce a state of lymphatic expansion or open areas and quasi-tubular segments with frond-like, short
edema to visualize them well, although in the absence of an blind-ending side branches. Reservoir lymphatics are found
edematous state they could still be seen under conditions of with prelymphatics and the two intermingle at times making
microvascular leakage, such as the blood vessel angiogenesis it difficult to distinguish them. The junctions of prelymphat-
that occurs with neoplasia. ics with conduit lymphatics and reservoir lymphatics with
With such techniques, it is possible to delineate differ- conduit lymphatics are structurally identical. Because of the
ent forms of lymphatics in the lungs. However, it is first difficulty in distinguishing them at times, both structures have
important to distinguish blood from lymphatic vessels [2]. been called initial lymphatics [4].
Blood vessels are more cylindrical and generally have dis- The pleural surface of normal animals seldom has lym-
tinct endothelial cell nuclear impressions. Cast pulmonary phatic casts. (The experience with cast human material is
veins have round impressions caused by their endothelial insufficient to draw conclusions.) In an experiment with
cell nuclei, regular ring-like constrictions, and do not run hyperoxic edema, almost two-thirds of the pleural surfaces
with airways once they are away from the hila. Even rather of rats had cast initial lymphatics [4].
large pulmonary veins (>200 ␮m in diameter) may have cap-
illaries joining them at right angles. On the other hand, the 3.2. Conduit lymphatics
casts of pulmonary arteries have flattened, elliptic endothelial
cell nuclear impressions running in the same direction as the Initial lymphatics are boundary structures from which
blood stream. The pulmonary artery casts lack narrow ring- conduit lymphatics arise. Conduit lymphatics occupy lit-
like impressions and have greater space between their surface tle space compared to initial lymphatics, but are a more
and adjacent capillaries than pulmonary veins. Arteries often consistent finding on normal pleural surfaces, although it
D.E. Schraufnagel / Pathophysiology 17 (2010) 337–343 339

Fig. 1. Lymphatics on the pleural surface. (A) A cast of prelymphatics (P), or tissue planes crossed by collagen and other connective tissue. A conduit lymphatic
(C) arises from the prelymphatic. The free access to the lymphatics from the tissue space allows the immune system to sample the entire lung. (B) Casts of
reservoir lymphatics (R), which are in close association with prelymphatics (P). Reservoir lymphatics are characteristically flat structures with budding blind
pouches and (C) shows them joining with conduit lymphatics (C). (D) A conduit lymphatic (C) on the pleural surface. The lack of strict cylindrical shape is a
feature that distinguished conduit lymphatics from the pleural blood capillaries beneath it. A small patch of reservoir lymphatics (R) is present. These scanning
electron microscopic images are of vascular and lymphatic casts on the pleural surface of rats exposed to a hyperoxic environment.

is still necessary to survey the whole pleural surface at mainly in the adventitial space in lymphatic structures with-
low magnification to find them in normal animals. Cast out complete endothelialization [5].
conduit lymphatics are tubular and have endothelium and In edema states, saccular lymphatic casts are found
valves, but they are not as rigidly cylindrical as blood ves- surrounding digested bronchioles. These have the same
sels. They vary in diameter irrespective of their course; their characteristics as the periarterial and perivenous saccular
diameter can be twice what it was just upstream or down- lymphatics (Fig. 2C). Conduit lymphatic casts in the bron-
stream. Nor are their surfaces as smooth as those of blood chovascular bundle are rarely seen if perivascular saccular
vessels. They may arise from either reservoir lymphatics lymphatics are absent (Fig. 3A). In conditions of lymphatic
or prelymphatics on the pleural surface or saccular lym- profusion, conduit casts can be identified along the bron-
phatics of the perivascular, peribronchial or peribronchiolar chovascular bundle and in the interlobular septa. When these
space [1]. vessels are in the bronchovascular bundle they are round or
oval. When they are away from the bronchovascular bundle
(usually in the interlobular septa) they may be thin, tortuous
4. Lymphatics within the lung and spiraling (Fig. 3C). The cast lymphatics of the inter-
lobular septa often have a medial notch. In rats exposed to
4.1. Perivascular and airway lymphatics hyperoxia, the conduit lymphatic vessels in the bronchovas-
cular bundle are larger than the accompanying bronchial
Saccular and tubulo-saccular lymphatics surround non- artery and up to about 40% of the size of the pulmonary
capillary blood vessels and airways within the lung (Fig. 2A artery. The surface of the lymphatic casts is textured with
and B). These large, thin-walled sac-like structures are often diagonal and longitudinal indentations and oval outpouch-
more than 100 ␮m in width. Lymphatic filling varies greatly ings. The diameter of the conduit lymphatics may vary greatly
depending on how edemogenic the condition is. They range within short lengths of the long axis [1], although this is more
from surrounding the blood vessel wall with a thick double typical of pleural conduit than parenchymal lymphatics and
layer to being only a scant patchwork on its surface. Aharine- in conditions of more moderate edema. Larger (more proxi-
jad et al. showed that the lymphatics were increased around mal) conduit lymphatics accept tributaries less frequently. At
pulmonary venous muscular constrictions (sphincters) more times, the flat intrapulmonary lymphatics join in star-shape
than other areas of the vein. The lymphatic enlargement is figures with multiple tributaries coming together at one place.
340 D.E. Schraufnagel / Pathophysiology 17 (2010) 337–343

Fig. 2. Lymphatics within the lung occur around blood vessels and airways. (A) Saccular and tubulo-saccular lymphatics (S) found around blood vessels in
states of lymphatic expansion. They empty into conduit lymphatics that transport lymph to hilar nodes. (B) Another image of a cast conduit lymphatic (C)
traveling with a pulmonary vein. This conduit lymphatic around the vein is flatter than the one shown in (A). Veins and their lymphatics travel in the interlobular
septa. Pulmonary and bronchial arteries, bronchioles, and lymphatics lie in bronchovascular bundles in the center of a pulmonary lobule. (C) The cast of the
lymphatics around a bronchus (Br) that has been digested. The tubulo-saccular (S) and conduit (C) lymphatics have the same form as those around the artery
in the bronchovascular bundle. These scanning electron microscopic images are of the lymphatic casts of rats exposed to hyperoxia.

Fig. 3. Conduit lymphatics. (A) An enlarged conduit lymphatic (C) adjacent to a blood vessel (BV) and saccular lymphatics (S); (B) An enlarged conduit
lymphatic adjacent to a pulmonary artery (a). The pulmonary artery has oval endothelial nuclear impressions and is more tubular than the conduit lymphatic,
which has a fluctuating diameter. (C) An intrapulmonary conduit lymphatic (IC) not in a bronchovascular bundle. These thin lymphatic vessels have central
grooves, stretch long distances, and often twist. At times, 4 or 5 branches may join at one place.These scanning electron microscopic images are of lymphatic
casts of rats exposed to hyperoxia.
D.E. Schraufnagel / Pathophysiology 17 (2010) 337–343 341

5. Markers cells because these endothelial cells had VEGFR-3 and


podoplanin, markers of lymphatic endothelial cells. However,
Fortunately, several molecular markers have been found it is now known that they infect blood vascular endothelial
on lymphatics to make their identification easier. These dis- cells and upregulate Prox-1, which is the master regulator of
coveries in the last decade have had far-reaching implications the lymphatic lineage. Prox-1 downregulates blood vascular
because they allow the study of lymphatic development in endothelial cell markers in favor of those of the lymphatic
health and disease. For example, since the lymphatics are endothelial cells [11].
a major route for cancer metastases, controlling lymphatic
development might control neoplasia.
Prox-1 (prospero-related homeobox gene) is found only 6. Lymphatic expansion
on lymphatics. It is a transcription factor that has wide rang-
ing control of the development of lymphatic endothelial cells. The lymphatics expand in many ways. After continu-
LYVE-1 (lymphatic vessel endothelial hyaluronan receptor- ous exposure to 85% oxygen for 7 days, experimental rats
1) is a CD44 homologue and hyaluronan receptor. It is found develop advanced pulmonary edema. Injecting a plastic resin
on lymphatic endothelial cells and macrophages but not into the blood vasculature will result in extensive casting of
blood endothelial cells. This marker is useful for measur- the lymphatics [4]. In these animals most blood vessels and
ing lymphatic density, which has been correlated with tumor conducting airways will be extensively flanked by saccular
prognosis in some settings. lymphatic casts. This resolves when the animals are removed
VEGF-C and VEGF-D (vascular endothelial growth fac- from the hyperoxic environment. After 14 days in ambient air,
tors C and D) and their cell surface receptor, VEGFR-3, the lymphatic casts are again uncommon with this injection
are found on lymphatic endothelial cells but not blood method [4].
vascular endothelial cells. VEGF-C and VEGF-D promote Another model of pulmonary edema entails acute air-
lymphangiogenesis by interacting with VEGFR-3, which way injury from excessive volume of mechanical ventilation.
activates an intracellular tyrosine kinase-dependent signal- Rats ventilated at high tidal volume (12–16 mL) for 25 min
ing cascade. VEGF-C and VEGF-D also bind neuropilin-2 showed more perivascular, interlobular septal, and alveolar
(Nrp2), a semaphoring receptor expressed on lymphatic ves- edema than those ventilated with low tidal volume. Most of
sels. With the VEGFs, the semaphorins help define a pattern the lymphatic changes occurred in the perivascular lymphat-
of vessel growth by regulating cell and extracellular matrix ics, with little accumulation in pleural lymphatics. Larger
interactions. This makes the neuropilin receptors for vascu- blood vessels had more edema in their surrounding lymphat-
lar endothelial growth factors important in tumor biology. ics than smaller blood vessels [12]. The pleural lymphatics
Controlling neuropilin may downregulate tumor growth and may be expected to show less filling that perivascular because
metastasis [6]. Knocking out Nrp2 in mice leads to lymphatic the acute insult was greater to the proximal airway and
hypoplasia. alveoli, which are drained by perivascular lymphatics. The
Deletion of Prox-1 or VEGF-C leads to complete absence importance of this model was that it showed the lymphatic
of lymphatics in mice. VEGF-D knock out does not affect expansion can occur quickly—without time for lymphangio-
lymphatic development, although if VEGF-D is given to genesis.
VEGF-C knockout mice, it can allow lymphatic develop- Metastatic lung cancer may spread via lymphatics. Cells
ment. Prox-1, VEGF-C, and VEGF-D are important in the were thought to gain access to existing lymphatic channels.
normal development of lungs and their levels are elevated in Part of the defense against lymphangitic metastases came
late neonatal life [7]. VEGF-C and VEGF-D are ligands for from lymph nodes that occur at regular intervals along the
integrin ␣9, which is required for normal lymph development lymphatic network. Although this is still true, recent work
[8]. has shown a more complex picture. For example, lung can-
Other important markers include desmoplakin and cer can produce high levels of VEGF-C, which can stimulate
plakoglobin found in the adherent junctions of lymphatic expansion of the lymphatics and facilitate spread of the
endothelial cells and LA102, another lymphatic endothelial tumor. Several important molecules associated with lym-
cell specific marker [9]. phatic expansion are now potential targets for antineoplastic
Podoplanin belongs to the family of type-1 transmem- therapy. It appears that patients with cancers with increased
brane sialomucin-like glycoproteins and normally is found lymphatic vessel density and higher levels of VEGF-C,
only on lymphatic vascular endothelium, but it is also VEGF-D, and VEGFR-3 have a shorter survival [13].
expressed in different types of tumor cells. Prox-1 induces the Pulmonary lymphatic expansion occurs in a variety of dis-
expression of podoplanin. Podoplanin/D2-40 in lymphatic eases besides edema states. In asthma and other conditions
endothelial cells is also used to estimate lymphatic microvas- involving chronic inflammation of the airway, mucosal and
cular density, which is greater in more aggressive neoplasms submucosal edema is associated with an influx of inflam-
[10]. matory cells, mucus cell hyperplasia, subepithelial fibrosis,
It is interesting that herpesvirus-8, the cause of Kaposi and thickened basement membranes. McDonald and col-
sarcoma, was first thought to infect lymphatic endothelial leagues infected mice with Mycoplasma pulmonis to mimic
342 D.E. Schraufnagel / Pathophysiology 17 (2010) 337–343

a bronchitis state. Lymphangiogenesis associated with this contraction of muscle or respiratory movements because the
model was dependent of signaling of VEGF-C, VEGF-D, initial lymphatics have incomplete endothelia. Lymphatic
and VEGFR-3. Inhibiting VEGFR-3 resulted in lymphatic valves interestingly do not have moving parts but are eccen-
growth inhibition without affecting blood vessel growth [14]. tric monocuspid, funnel-shaped structures [22]. In addition
In a rat model of lung transplantation rejection, transfer to the pumping action of the lymphatics, the casting of lym-
of VEGF resulted in the doubling of the LYVE-1 positive phatics of dead experimental animals attests to the passive
lymphatic vessels in the airway walls. Giving an inhibitor role of lymphatic flow.
of VEGFR resulted in a 50% reduction [15]. Accompany-
ing this was a reduction in CD4 and CD8 cells in the airway
walls. This highlights that an expanded lymphatic system can 8. Lymph nodes and immune cell trafficking
traffic more lymphocytes and alter immunity. In transplanta-
tion, this may not be good because it could increase the risk If microbes transgress the upper respiratory tract barriers,
of rejection. On the other hand, it could result in a more they come in contact with dendritic cells that extend to the
rapid clearance of infecting microbes in these immunocom- epithelial surface. Dendritic cells sample and capture micro-
promised hosts and be beneficial. bial antigens and then migrate to the lymphatic vessels. The
dendritic cells breakdown protein into peptides, which bind
to major histocompatibility complex (MHC) molecules, a
7. Lymph flow necessary process for presentation to T lymphocytes. While
transporting the antigens to the lymph nodes, the dendritic
Lymph flow in the lung proceeds in opposite directions. cells mature. They lose their adhesiveness to the epithelium
Lymphatics in the lung perimeter flow to the pleura and the and express chemokine receptor 7 (CCR7), which tracks them
rest flows toward the hilum. The pleural flow is more effi- to the T cell zones of the lymph node. Mature dendritic cells
cient than the central flow, which explains the classic chest express toll-like receptors that recognize microbial molecules
radiographic “butterfly pattern” of acute pulmonary edema in and activate cells to produce cytokines. They also produce
human patients [16]. Gravity and respiratory movement also costimulatory molecules that are required to develop T cell
affect lymph flow; the lower lobes clear lymph four times immune competence. Maturation of dendritic cells converts
more than the upper [17]. them from cells that capture foreign protein to cells that can
India ink injected into the pleural cavity rapidly enters the present antigen to naïve T cells and activate lymphocytes
lymphatics. The material is taken up by preformed parietal [23].
pleural stomata that enter directly into the chest wall lym- Antigens also may enter the lymphatics in a free form.
phatics [18]. Although normal pleural fluid, which is present Lymph-borne antigens can be extracted from the lymph by
in small amounts, may be produced and taken up by either dendritic cells and macrophages residing in the lymph nodes.
the visceral or parietal pleura, the parietal pleura is the main Soluble inflammatory mediators produced at the sites of
agent to remove excess fluid in pathologic conditions. infection also enter the lymphatics. Lymph nodes, which are
Inhaled particle distribution is also probably affected by imposed along the lymphatics, filter and delay transport of
lymph flow. With tidal breathing, the greatest air flow goes to immune cells back to the blood. These lung lymph nodes
the lower lung zones but silica, for example, deposits in the are sentinels that prevent bacterial pneumonia from becom-
upper lung zones in humans. Animal experiments have shown ing bacterial sepsis. After lymph enters the node, it goes to
particles less than 1 ␮m in diameter are cleared from the the subcapsular sinus and then the paracortical area. Here
dependent parts and migrate to the upper zones after several the dendritic cells present antigens to naïve T cells and initi-
days [19]. Asbestos particles, which are more often found in ate the adaptive immune response. The lymph channels and
the lower lung zones in humans, pass through the epithelium. nodes provide for other immune functions such as delivering
The fibers that enter the interstitial space can be picked up by chemokines from the inflamed tissues. Chemokines, such as
the lung lymphatics, dumped into the blood stream, and be CCL2 (C–C motif, ligand), contribute to the recruitment of
distributed to all organs. Other asbestos fibers penetrate to the leukocytes into the nodes. Specialized lining within the node
viscera and even parietal pleura. Pleural fluid is preferentially separates molecules by size and chemical composition and
sucked into the parietal pleural stomata. Hard, needle-like shunts them into the T cell zones for processing. The nodes
asbestos fibers accumulate on the parietal pleural and cre- are separated into zones by reticular and collagen fibers and
ate a reaction to this irritation, pleural plaques. Asbestos other tissue that allow the antigen presenting cells to come
fiber translocation takes place over decades in humans with and remain in close contact with the lymphocytes. Different
asbestos-related lung disease [20]. classes of lymphocytes are segregated into different areas.
The heterogeneity of lymph movement suggests that addi- Follicles are the B cell zones, where both mature and naïve B
tional factors, such as surface tension of the fluid, are cells aggregate. Germinal centers develop in response to anti-
involved [21]. Although fluid especially in large lymphat- genic stimuli and are sites of B cell proliferation, selection of
ics can move by muscular contraction of lymphatic walls, B cells producing high-affinity antibodies, and the generation
most lymph movement occurs by external forces, such as of memory B cells. T cells aggregates are located beneath and
D.E. Schraufnagel / Pathophysiology 17 (2010) 337–343 343

more central to the follicles in the paracortical cords. Naïve T [5] S. Aharinejad, P. Böck, W. Firbas, D.E. Schraufnagel, Pulmonary lym-
and B cells enter the node through high endothelial venules in phatics and their spatial relationship to venous sphincters, Anat. Rec.
response to chemokines and their receptors. The chemokines 242 (1995) 531–544.
[6] A. Bagri, M. Tessier-Lavigne, R.J. Watts, Neuropilins in tumor biology,
bind to their cell surface receptors and set off a cascade of Clin. Cancer Res. (2009).
signaling to exert several intracellular changes. [7] S. El-Chemaly, S.J. Levine, J. Moss, Lymphatics in lung disease, Ann.
Another important component in the lymphatic system is N. Y. Acad. Sci. 1131 (2008) 195–202.
the bronchus associated lymphoid follicles. In addition to the [8] N.E. Vlahakis, B.A. Young, A. Atakilit, D. Sheppard, The lymphangio-
defense of the dendritic cells, lung microbial invaders may genic vascular endothelial growth factors VEGF-C and -D are ligands
for the integrin alpha9beta1, J. Biol. Chem. 280 (2005) 4544–4552.
encounter specialized secondary lymphoid tissue situated in [9] T. Ezaki, K. Kuwahara, S. Morikawa, K. Shimizu, N. Sakaguchi, K.
aggregates below the bronchial or bronchiolar epithelium. Matsushima, K. Matsuno, Production of two novel monoclonal anti-
These lymphoid follicles have access to the epithelial sur- bodies that distinguish mouse lymphatic and blood vascular endothelial
face as well as both the blood stream and lymphatics and cells, Anat. Embryol. (Berl). 211 (2006) 379–393.
contain specialized dendritic cells, called follicular dendritic [10] M. Raica, A.M. Cimpean, D. Ribatti, The role of podoplanin in tumor
progression and metastasis, Anticancer Res. 28 (2008) 2997–3006.
cells. The follicles thus can monitor immune stimuli from [11] H.W. Wang, M.W. Trotter, D. Lagos, D. Bourboulia, S. Henderson, T.
all three sources. Follicular dendritic cells can present anti- Makinen, S. Elliman, A.M. Flanagan, K. Alitalo, C. Boshoff, Kaposi
gens to previously sensitized B cells in germinal centers. The sarcoma herpesvirus-induced cellular reprogramming contributes to the
immune stimuli do not have to be bacteria. Dendritic cells can lymphatic endothelial gene expression in Kaposi sarcoma, Nat. Genet.
ingest tumor cells and virus infected host cells and present 36 (2004) 687–693.
[12] D.E. Schraufnagel, N.P. Agaram, A. Faruqui, S. Jain, L. Jain, K.M.
them to CD8 T cells instead of the usual CD4 T cells (cross Ridge, J.I. Sznajder, Pulmonary lymphatics and edema accumulation
presentation). after brief lung injury, Am. J. Physiol. Lung Cell Mol. Physiol 284
Different lymphocytes traffic through the blood stream, (2003) L891–L897.
lymphatics, lymphoid organs, such as the spleen, special- [13] M.G. Achen, S.A. Stacker, Molecular control of lymphatic metastasis,
ized lymphoid tissue, such as the aggregates associated with Ann. N. Y. Acad. Sci. 1131 (2008) 225–234.
[14] D.M. McDonald, Angiogenesis and remodeling of airway vasculature
bronchi, bronchioles, and nonlymphoid tissue at different in chronic inflammation, Am. J. Respir. Crit Care Med. 164 (2001)
frequencies and paces. This allows a constant sampling of S39–S45.
the body for antigens. The lymphocytes’ remarkable ability [15] R. Krebs, J.M. Tikkanen, A.I. Nykanen, J. Wood, M. Jeltsch, S.
to home back to the site of the inflammation results from Yla-Herttuala, P.K. Koskinen, K.B. Lemstrom, Dual role of vascu-
well-orchestrated set of molecular events involving adhesion lar endothelial growth factor in experimental obliterative bronchiolitis,
Am. J. Respir. Crit Care Med. 171 (2005) 1421–1429.
molecules, including selectins and integrins, and lymphocyte [16] F.G. Fleischner, The butterfly pattern of acute pulmonary edema, Am.
homing receptors that bind to endothelial cells. The adhesion J. Cardiol. 20 (1967) 39–46.
and detachment to extra cellular matrix along with chemokine [17] N.C. Staub, “State of the art” review. Pathogenesis of pulmonary edema,
concentrations determines how long the lymphocytes reside Am. Rev. Respir. Dis. 109 (1974) 358–372.
in the tissue before recirculating back through the lymphatics [18] N.S. Wang, The preformed stomas connecting the pleural cavity and
the lymphatics in the parietal pleura, Am. Rev. Respir. Dis. 111 (1975)
[24–27]. 12–20.
The lung lymphatics, thus, do far more that clear fluid [19] P.A. Valberg, R.K. Wolff, J.L. Mauderly, Redistribution of retained
and particulates. The facilitate and orchestrate the immune particles. Effect of hyperpnea, Am. Rev. Respir. Dis. 131 (1985) 273–
response that involves neoplasia and transplantation as well as 280.
infection. The availability of markers and better understand- [20] G. Miserocchi, G. Sancini, F. Mantegazza, G. Chiappino, Translocation
pathways for inhaled asbestos fibers, Environ. Health 7 (2008) 4.
ing of the process heralds a new era of lymphatic research [21] H. Bachofen, S. Schurch, R.P. Michel, E.R. Weibel, Experimental
and potential for medical monitoring and treatments. hydrostatic pulmonary edema in rabbit lungs. Morphology, Am Rev.
Respir. Dis. 147 (1993) 989–996.
[22] J.M. Lauweryns, Morphological studies of the blood and lymphatic
References microcirculation of the lung, Acta Cardiol. 19 (Suppl.) (1974) 157–167.
[23] D.N. Cook, K. Bottomly, Innate immune control of pulmonary dendritic
cell trafficking, Proc. Am. Thorac. Soc. 4 (2007) 234–239.
[1] D.E. Schraufnagel, Forms of lung lymphatics: a scanning electron
[24] A.K. Abbas, A.H. Lichtman, S. Pillai, Cellular and Molecular
microscopic study of casts, Anat. Rec. 233 (1992) 547–554.
Immunology, Elsevier, Philadelphia, 2007.
[2] D.E. Schraufnagel, Microvascular corrosion casting of the lung. A state-
[25] O. Ohtani, Y. Ohtani, Structure and function of rat lymph nodes, Arch.
of-the-art review, Scanning Microsc. 1 (1987) 1733–1747.
Histol. Cytol. 71 (2008) 69–76.
[3] D.E. Schraufnagel, D.B. Pearse, W.A. Mitzner, E.M. Wagner, Three-
[26] L.M. Ebert, P. Schaerli, B. Moser, Chemokine-mediated control of T
dimensional structure of the bronchial microcirculation in sheep, Anat.
cell traffic in lymphoid and peripheral tissues, Mol. Immunol. 42 (2005)
Rec. 243 (1995) 357–366.
799–809.
[4] D.E. Schraufnagel, J. Llopart Basterra, K. Hainis, J.I. Sznajder, Lung
[27] J. Bienenstock, M.R. McDermott, Bronchus- and nasal-associated lym-
lymphatics increase after hyperoxic lung injury: an ultrastructural study
phoid tissues, Immunol. Rev. 206 (2005) 22–31.
of casts, Am. J. Pathol. 144 (1994) 1393–1402.

You might also like