Download Apoptotic cell removal

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Extracellular matrix wikipedia , lookup

Tissue engineering wikipedia , lookup

Cell cycle wikipedia , lookup

Endomembrane system wikipedia , lookup

Cell growth wikipedia , lookup

Cytokinesis wikipedia , lookup

Mitosis wikipedia , lookup

Cell culture wikipedia , lookup

Cell encapsulation wikipedia , lookup

SULF1 wikipedia , lookup

Apoptosis wikipedia , lookup

Cellular differentiation wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

Programmed cell death wikipedia , lookup

Signal transduction wikipedia , lookup

Amitosis wikipedia , lookup

List of types of proteins wikipedia , lookup

Transcript
Review
R795
Apoptotic cell removal
Peter M. Henson, Donna L. Bratton and Valerie A. Fadok
Ingestion by professional or amateur phagocytes is the
fate of most cells that undergo apoptosis. Studies in
both Caenorhabditis elegans and mammals are now
converging to reveal some of the key mechanisms and
consequences of this removal process. At least seven
corpse removal genes in nematodes have mammalian
equivalents, and represent elements of signaling
pathways involved in uptake. In mammals, a wide
variety of apoptotic cell recognition receptors has been
implicated and appears to be divided into two
categories, involved in tethering the apoptotic cell or
triggering an uptake mechanism related to
macropinocytosis. Apoptotic cell removal is normally
efficient and non-inflammatory. By contrast, the process
may become subverted by parasites to yield a more
favorable growth environment, or in other cases lead to
fibrosis. Removal may also clinch the apoptotic process
itself in cells not yet completely committed to death.
Address: Program in Cell Biology and Department of Pediatrics,
National Jewish Medical and Research Center, 1400 Jackson Street,
Denver, Colorado 80206, USA.
Correspondence: Peter M. Henson
E-mail:[email protected]
Introduction
Cell recognition, engulfment and digestion are the final
stages in the process of apoptosis. The outcome of these
actions is the efficient deletion of unwanted or damaged
cells, often considered a counterpoint to mitosis. That
apoptosis is accompanied by rapid removal of the affected
cells in situ has been recognized since the phenomenon
was first characterized [1,2]. As discussed recently by Jack
Cohen [3], a newer view of biology suggests significant
overproduction, at the level of the organism, sperm, egg
and embryo, of individual cells during development. In
each case, excess material is removed and thus removal is
an important element of overall control. Even in the adult
organism, significant cell turnover seems to be the rule
rather than the exception, including systems such as the
brain that were long thought not to exhibit such plasticity.
Turnover of cells in metazoa means apoptosis and, critically, apoptotic cell removal — often called corpse removal.
However the mechanisms of apoptotic cell recognition and
removal are only now becoming apparent, an understanding that represents a fascinating investigative interplay
between genetic approaches in the nematode, Caenorhabditis elegans, and analyses of uptake mechanisms in mammalian systems (Figure 1).
Current Biology 2001, 11:R795–R805
0960-9822/01/$ – see front matter
© 2001 Elsevier Science Ltd. All rights reserved.
Combining studies in mammals and the worm
Seven genes have recently been identified in C. elegans that
appear to play a role in apoptotic corpse removal during
larval development or in corpse digestion after engulfment
[4–8]. One of the many strengths of the C. elegans system
goes back to the original cell mapping studies that led to the
identification of 131 specific somatic cells, as well as some
300 germ cells, that are destined for deletion (see [4,7]).
Until recently, the genes involved in removal each appeared
to correspond to steps in the signal pathways leading to
engulfment, rather than to cell recognition steps per se. In
contrast, studies in mammalian systems generated a host of
potential recognition receptors (see [9–13]) but made little
progress towards identifying uptake signaling molecules.
Recent reports provide a crucial synthesis between these
two approaches and highlight a number of important
points, not the least of which is the broad commonality in
mechanisms employed for apoptotic cell removal. It seems
reasonable to suggest that potential redundancy among
the recognition elements would make their identification
by the genetic screening approaches used for C. elegans less
likely. Such redundancy is certainly implicated in the
studies of mammalian systems in vitro. On the other hand,
the involvement of limited, and presumptively unique,
signaling pathways for uptake could readily explain the
R796
Current Biology Vol 11 No 19
Figure 1
A comparison of elements implicated in
apoptotic cell uptake in (a) mammals and
(b) C. elegans. The electron micrograph of
uptake in the nematode (b) is taken with
permission from [7].
1. Surface changes on the apoptotic cells
Mammal
PS
Oxidized phospholipid
ABC-1
Altered carbohydrates
ICAM3
Nematode
PS?
ced-7
2/3. Tethering and/or stimulation receptors
(a)
Macrophage
Corpse
PSR
PSR?
SR-A
CD36/SR-B
CD36?
SREC?
ced-1
CD68
αvβ3/αvβ5 integrins
β1 integrins
CD14
CR3
β2GP receptor
Lectin(s)
Collectin receptors (calreticulin/CD91)
4. Signaling molecules
CrkII
DOCK180
rac-1
ced-6
ABC-1
ELMO
(b)
Sheath cell
Corpse
ced-2
ced-5
ced-10
ced-6
ced-7
ced-12
5. Digestion
Acid DNAase?
nuc-1
Current Biology
identification of genes within such pathways in the nematodes. These mechanisms for cell removal may also extend
to necrotic cells and cell debris, even if the consequences
of such clearance may sometimes be different [5,14,15].
An example of this synergism is the recent, elegant paper
by Zhou et al. [7]. In this work, the last but one of the
seven C. elegans genes to be characterized, ced-1, does seem
to encode a potential recognition and signaling molecule
that may initiate cell uptake. ced-1 was originally identified
in the genetic screens for persistent cell corpses and encodes
a protein that plays a critical role in engulfment. The protein
is expressed in the membranes of phagocytosing cells and
clusters around corpses during removal. Deletion of potential signaling domains within the intracellular portion of
the molecule leads to defective apoptotic cell clearance,
even in the face of unaltered clustering. These data suggest
that CED-1 both binds to the target apoptotic cell and initiates signaling for uptake.
While the specific ligand that might be recognized by
CED-1 remains unknown, clustering, which is taken to
be equivalent to ligand engagement, appears to be associated with the presence of the product of ced-7. This latter
gene encodes a protein similar to ABC-1 in mammals [16],
a member of the ATP binding cassette transporter family
[17]. Defects in ABC-1 in man are associated with Tangier
disease [18–20] and the molecule appears to be involved
in cholesterol or phospholipid transport. Zhou et al. [7]
suggest the possibility that CED-7 changes phospholipid
distribution on the apoptotic cell (see below) in C. elegans
and that this is what CED-1 recognizes.
It seems reasonable to consider the apoptotic cell removal
process to comprise five steps, namely: surface alterations
and ligand expression on the apoptotic cell; recognition of
these ligands by tethering receptors on the phagocyte; initiation of the appropriate signaling pathways in the phagocyte, which might be mediated by the tethering
interaction but may also involve separate or additional
uptake receptors; the process of engulfment; and finally,
phagosome–endosome interactions leading to digestion. It
is also important to emphasize that uptake is not limited to
professional phagocytes, such as macrophages or immature
dendritic cells in mammals, but may be performed by most
cell types. An example of this is seen in the normal developmental clearance of cells from the interdigital webs in mice
wherein removal occurs even in animals that do not contain
macrophages — PU-1–/– mice — but at a slower rate [21].
Surface changes on apoptotic cells leading to
recognition and removal
Despite all the effort spent characterizing the mechanisms
of apoptosis, comparatively little has been focused on
surface changes on the apoptotic cell. A very clearly defined
change is the exposure of phosphatidylserine on the apoptotic cell surface, shown first on lymphocytes [22] and now
known to occur with essentially all apoptotic cells in
mammals. Phosphatidylserine also appears to be involved
in apoptotic cell removal in amphibians [23] and is seen
Review
during apoptosis in Drosophila [24]. Whether apoptotic
cells in C. elegans express phosphatidylserine on their surface
has still to be shown definitively.
While the mechanism of phosphatidylserine exposure is
still poorly understood at the molecular level, it appears to
require activation of a phospholipid ‘scramblase’ that
enhances bidirectional phospholipid flip-flop across the
membrane bilayer, and inactivation of an aminophospholipid translocase that would normally return the phosphatidylserine to the inner membrane leaflet. Effects on
both these systems can be seen during normal cell function
but are reversible; the key difference in apoptosis may be
the permanence of the effect. Candidate regulatory genes
for these activities have been identified [25–28] although
their exact role, identity or mode of action is by no means
clear. A search of the C. elegans gene database reveals a
molecule with considerable homology to the mammalian
scramblase. And a putative aminophospholipid translocase
— a member of the P-type ATPase family — is widely distributed, from plants to man [25,29–31]. Although the
validity of this candidate has been challenged, more recent
data in Arabidopsis support a role for these P-type ATPases
as aminophospholipid translocases [29].
Another class of surface changes that has been implicated
in apoptotic cell removal is alteration of membrane carbohydrates. The original observation of apoptotic cell recognition by macrophages suggested a lectin-like activity
[32,33]. Alterations to surface charge on apoptotic cells were
also attributed to carbohydrate moieties [32,34] although
specific sugars on the apoptotic cell surface were not identified in either of these studies. On the other hand there is
some evidence for increased expression of mannose on
apoptotic cells leading to their removal within the liver
[35,36]; this is an area that needs much more detailed
investigation.
A number of mechanisms for such changes to membrane
carbohydrates can be envisioned. New or altered glycosylated molecules may be supplied to the cell-surface from
internal membranous structures, possibly by direct fusion
of elements from the endoplasmic reticulum with the
plasma membrane. In addition, the normal turnover of
membrane glycoproteins is likely to be significantly altered
and decreased in apoptosis leading to gradual depletion of
key molecules or their normal glycosylation. Any enzymatic
alteration of surface oligosaccharides would require either
the presence of enzymes in or on the membrane surface —
so-called ectoenzymes — or in the extracellular medium.
This issue represents a potential problem in all studies of
apoptosis carried out in vitro, as some necrosis and cytolysis
is bound to occur and could contribute intracellular
enzymes to the system that might not be present during
the efficient removal usually seen in vivo.
R797
A completely different possibility is that appropriate glycoproteins become laterally redistributed or clustered in the
membrane of apoptotic cells. This could be a critical determinant of recognition. For example, members of the family
of defense collagens [37] that includes the C-type lectins
known as Collectins as well as the first component of the
classical complement pathway, C1q, have been shown to
bind to highly localized sites on apoptotic cells [38–40].
Mannose binding lectin is one of these proteins and not
surprisingly its attachment to apoptotic cells is blocked by
high concentrations of free mannose. It is noteworthy that
preliminary evidence shows that phosphatidylserine expression and membrane phospholipid redistribution can often
be patchy (our unpublished observations) and highly localized. The relationship between phospholipid redistribution
and other putative membrane changes to proteins or their
carbohydrate substituents is poorly described.
What is also not clear is the linkage between the two broad
signaling pathways leading to nuclear apoptotic changes —
those that involve mitochondria and those that do not —
and surface changes. How the aminophospholipid scramblase is inactivated is not known. As far as phospholipid
redistribution is concerned, cleavage of PKCδ by caspase 3
induces its unregulated activation, which, we suggest,
leads in turn to a more persistent induction of the phospholipid scramblase [41]. What is not clear is how the
actual phospholipid scrambling effect is mediated, which
is where questions about the involvement of the ABC transporters might come in. These are known to transport phospholipids across cell membranes and it is certainly possible
that ABC1 serves such a function in the apoptotic cell.
However this suggestion, which has been raised by a
number of groups (see [42,43]) is at present merely an
interesting speculation.
If ABC transporters are involved in phospholipid scrambling, the generality of phosphatidylserine expression during
apoptosis dictates the participation of more than one
member of this family as ABC1 is not generally distributed
among different cell types. The phospholipid scramblase
appears to require calcium and phosphorylation [41,44] for
its activity, self aggregates and is known to scramble phospholipids in artificial membrane systems [45], albeit with
low efficiency. It might therefore alter the membrane structure by itself to mediate transbilayer movement. However,
although potentially present, it was not identified in the
genetic screen for abnormal corpse removal in C. elegans.
Another element that must be put into the equation is the
suggestion that, in both mammals and the nematode,
phosphatidylserine is expressed on both the apoptotic cell
and the phagocyte during removal [7,43,46]. At this point,
therefore, the role of ABC1 is difficult to unravel. In the
mouse it seems to play a role in the phagocyte as
R798
Current Biology Vol 11 No 19
macrophages from knockout mice show defects in apoptotic cell uptake [43]. The problem here is that most cell
types have the capacity to recognize and ingest apoptotic
cells — presumably a very primitive function — once
again in the face of a restricted distribution of ABC1.
would require a receptor on the phagocyte and an important recent paper [57] shows that Mer, a member of the
Axl, Mer, Tyro3 family of receptor tyrosine kinases, may
play such a role. Critically, mer–/– mice show a marked
defect in removal of cells in the thymus.
Recognition of apoptotic cells
Whether the putative receptor, Mer, acts via Gas-6 and
exposed phosphatidylserine in vivo is not yet known — it
could also act along with some of the other recognition
molecules as a signaling agent. It is important to note in
this section that while many receptors have been suggested to participate in recognition of apoptotic cells
in vitro and even in reconstituted transfected cells, far fewer
candidates have been shown to play this role in vivo. In
part this is likely to reflect the significant redundancy, and
by implication, importance, of the removal processes.
Two possible issues of importance may be gleaned from
the above observations regarding surface changes on apoptotic cells. First, the classes of molecules that are altered,
carbohydrates and phospholipids, are recognized in vivo by
so-called pattern recognition molecules, which have recently
been associated with the innate immune system. Secondly, aggregation of glycoproteins and phospholipids in
the membrane of the apoptotic cell further fits this
concept in that many of these recognition processes are
thought to be low affinity, high avidity systems whose
effectiveness is markedly enhanced by local aggregation.
In mammals, phosphatidylserine expression on the apoptotic cell membrane appears to be a critical element for
recognition or uptake, despite the abundance of potential
recognition receptors [11–13,47,48]. In the absence of
phosphatidylserine expression, uptake is minimal and deliberate replenishment of outer leaflet phosphatidylserine
restores uptake [48]. The selectivity and stereospecificity
of this restoration system strongly implicates the newly
cloned phosphatidylserine receptor [12] in such uptake. A
simple model suggests a variety of tethering ligands with a
critical involvement of the phosphatidylserine receptor for
initiation of the engulfment process.
While a C. elegans homologue for the phosphatidylserine
receptor was identified from the gene database [12], it too
never appeared in the genetic screen for corpse removal.
To this point, its possible role in apoptotic cell removal in
nematodes is unclear (although it has been implicated in
amphibians [23]). Careful examination of the C. elegans
phosphatidylserine receptor candidate gene does reveal a
deletion in a portion of the putative intracellular domain
that might contain a tyrosine phosphorylation site. Whether
this suggests that in the nematode the receptor serves
more a tethering than signaling function remains to be
determined. In mammals, the receptor also appears to play
a key role in suppressing the inflammatory response during
apoptotic cell removal [12,48,49].
The phosphatidylserine receptor in the phagocyte membrane would be expected to interact directly with phosphatidylserine on the apoptotic cell. Other phosphatidylserine-binding membrane proteins include the
scavenger receptors (see below and [50–54]). Gas-6 is a
soluble protein that can recognize and bind phosphatidylserine on apoptotic cells [55,56] and could act as a
bridging molecule. Its ability to participate in uptake
As noted above, the first study of apoptotic cell recognition suggested the involvement of a lectin [33]. More
recently, the complement protein C1q was reported to
bind to blebs on apoptotic keratinocytes [38,39] and to
localized sites on other apoptotic cell types [40]. The same
was seen for mannose binding lectin and surfactant proteins A and D (Vandivier W and Ogden CA, unpublished
observations). Moreover, each of these molecules has been
observed to enhance uptake of apoptotic cells into
macrophages in vitro [40,58] and, for C1q [59] and surfactant protein D, in vivo as well (Vandivier W and Ogden
CA, unpublished observations). These molecules appear
to bind apoptotic cells through their globular head groups
— the pattern recognition regions — and signal for uptake
through their conserved collagen-like tails. Because of the
potential involvement of mannose on the apoptotic cell,
the mannose receptor may be an additional candidate, but
there is as yet no evidence to support its involvement in
mediating apoptotic cell recognition.
On the other hand, scavenger receptors in the A and B
group have been suggested to recognize apoptotic cells,
particularly SR-A [9,60] and CD36 [61–65] or SR-B1
[51–53,66]. In Drosophila, a member of the SR-B family,
Croquemort, has been shown to participate in corpse
removal [67,68]. The ligands involved on the apoptotic
cells are not known. Closely associated with CD36 activity
are the αv integrins: αvβ3 on macrophages [65] and αvβ5 on
dendritic cells [63] and retinal pigment epithelial cells
[69]. Bridging by thrombospondin has been suggested in
some of these CD36-mediated systems although, once
again, the apoptotic cell ligands are unknown. As CD36
can directly bind anionic phospholipids [50,54] the potential interactions in this system are likely to be complex.
ced-1, like the other six genes involved in C. elegans apoptotic cell removal, is also similar to a mammalian molecule,
in this case, the scavenger receptor from endothelial cells,
Review
SREC [70]. SREC binds and internalizes acetylated lowdensity lipoproteins. However, it is not widely distributed
among mammalian cell types and so cannot be solely
responsible for apoptotic cell recognition. Nor does it
exhibit the specificity for phosphatidylserine recognition,
in contrast to the phosphatidylserine receptor, that characterizes this phospholipid’s involvement in apoptotic cell
clearance. SREC has yet to be shown capable of mediating
apoptotic cell uptake, even in endothelial cells. Intriguingly, although their sequences appear unrelated, SREC
and the phosphatidylserine receptor are both found on the
tip of human chromosome 17; the biological significance of
this association is unknown.
CD14 represents an interesting candidate for apoptotic
cell recognition. This soluble or glycophosphoinositidelinked protein is involved in the appropriate interaction of
lipopolysaccharides with Toll-like receptor 4. It has been
implicated in recognition of apoptotic cells, particularly
apoptotic lymphocytes. Most of the recognition processes
do not seem to show much specificity for apoptotic cell
type so this selectivity may either be a special case or may
merely represent lack of detailed investigation in other
systems. The counter-ligand on the apoptotic cell has
been suggested to be ICAM3 [71] which is upregulated or
exposed on apoptotic lymphocytes through an unknown
mechanism. This again brings in something new, namely
a protein ligand. While it seems likely that CD14 represents a tethering rather than signaling ligand as it does not
exhibit a transmembrane domain, it may prove important
in immune system development and function. These
observations also raise the possibility that there will be
other cell specific protein–protein interactions involved in
apoptotic cell removal.
Finally, one cannot ignore the possibility that apoptotic
cells in mammals might be rendered more susceptible to
phagocytosis in vivo by more conventional antibody–antigen
interactions, the process of opsonization. For example, a
number of studies have suggested that apoptotic cells initiate attachment of complement C3bi followed by deposition of IgG, and subsequent uptake through complement
receptor 3, CR3 [72–74]. Once again, however, the structures on the apoptotic cells that might initiate this
opsonization are not fully defined.
R799
engulfment. CED-12 has recently been shown to participate in the CED-2, CED-5, CED-10 pathway in C. elegans.
Two mammalian homologues of CED-12, ELMO1 and 2,
seem to form a trimolecular complex with CrkII and
DOCK180 that results in activation of Rac-1 (Ravichandran K, personal communication). However, mutation of
any one of these four genes in C. elegans diminishes, but
does not eliminate, corpse removal.
CED-1, CED-6 and CED-7 seem to participate in an
alternative path for uptake: double mutants of any of these
three genes with one of the former group results in much
more profound abnormalities in apoptotic cell clearance
than the single mutation. CED-1 has already been discussed above and may represent a C. elegans recognition or
signaling receptor. CED-7 is an ABC transporter that may
be involved in both apoptotic cell and phagocyte. As discussed above, its activity, while very poorly understood, is
most likely to be involved in membrane alterations. CED6 resembles an adaptor protein with a phosphotyrosine
binding domain, and its mammalian homolog is implicated
in phagocytes during apoptotic cell engulfment [77,78].
How CED-6 interacts with putative surface receptors or
downstream effector molecules is not yet clear.
In contrast, evidence for two separate uptake pathways in
mammals is lacking. Possibly one or more of the different
recognition molecules could initiate uptake by using different signal pathways but, at least to us, this seems
unlikely. It has been suggested that uptake into professional phagocytes such as macrophages could be different
from ingestion by tissue cells [81]. However, the effect is
largely one of ingestion rate and efficiency and in our
hands does not show differences in receptor usage or
uptake mechanism. Even in C. elegans it is not clear how
the proposed pathways are related.
Uptake signaling pathways
As actin dependent processes are presumably involved,
Rho family GTPases are expected in both pathways. Rac-1
is the key candidate in the CED-2, 5, 10 cell uptake path
and studies in murine macrophages suggest the participation of Rac-1 and Cdc42 [80]. A key problem with many of
these investigations is the complexity of trying to sort out
signaling processes with multiple, or even unknown, recognition molecules and receptors, and initial signal transduction molecules. The ability to address the processes initiated
by an individual receptor would be of great value.
Gene complementation studies in C. elegans suggest two
related pathways for internalization of the apoptotic cells
by phagocytes [7,75,76]. CED-2, CED-5 and CED-10 represent homologs of mammalian CrkII, DOCK180 and Rac1 and constitute one such pathway. Studies in mammalian
systems also show these molecules are involved in apoptotic cell removal (see[43,77–80]). They are presumed to
act at the point of cytoskeletal reorganization during
It is clear that a critical issue in understanding apoptotic
cell removal is whether the uptake processes and signaling
pathways are unique. They are clearly different from Fc
receptor driven phagocytosis. Stimulated macropinocytosis
driven by growth factors such as epidermal growth factor
(EGF), platelet-derived growth factor (PDGF) and macrophage colony-stimulating factor (M-CSF) signaling through
R800
Current Biology Vol 11 No 19
Figure 2
Phagocytosis of cells by the zipper
mechanism or by stimulated
macropinocytosis. It is suggested that most
forms of apoptotic cell ingestion result from
the macropinocytotic process.
Zipper mechansim
Stimulated
macropinocytosis
Membrane
ruffles
Current Biology
their receptor tyrosine kinases may be a closer model.
However, very preliminary evidence obtained in collaboration with Dr Kodimangalam Ravichandran suggests to us
that upstream signaling elements resulting from growth
factor stimulation may be different from those seen in
apoptotic cell uptake. On the other hand, downstream molecules such as Rac-1 are common. The nematode studies
seem to support this implication.
From the point that Rho family GTPases become involved,
the signaling for cytoskeletal reorganisation, ruffling, membrane extension and eventual fusion, along with other elements of internalization, may not be specific for apoptotic
cells or their recognition receptors. However, key issues are
the relationship between Cdc42 and Rac; how and when
Rac-1 becomes localized to the site of uptake; whether the
interaction of an apoptotic cell initiates activation and
membrane alterations at only the local site of contact and
stimulation; and any potentially unique processes involved
in subsequent phagosome disposition, pH changes, lysosome and vesicle fusion. Because of the overall importance
of the apoptotic cell removal process and its consequences,
these are critical questions for future investigation.
Mechanisms of uptake
Ingestion of particles into cells occurs by a number of different mechanisms reflecting different receptors, signaling
pathways and physical uptake processes. Immunoglobulin
Fc receptor stimulation induces phagocytosis by a process
that involves the sequential ligation of the receptors by Ig
molecules on the particle as in a zipper [82–85]; this results
in a phagosome with membranes in close contact with
the ingested particle. Uptake through CR3–αmβ2 integrin
involves the particle appearing to sink into the phagocytic
cell, without the development of an obvious phagocytic
cup. We have recently suggested that uptake of apoptotic
cells through the types of receptors discussed above occurs
by a process more akin to macropinocytosis (Figure 2).
In stimulated macropinocytosis, induced, localized membrane ruffling is associated with enclosure of extracellular
fluid followed by internalization within a spacious phagosome. Particles attached to the surface are taken in at the
same time. In this system the receptors for macropinocytosis initiation could be on either the particle or the apoptotic cell, or could interact independently if the apoptotic
cell were already tethered to the phagocyte membrane.
We have called this a ‘tether and tickle’ mechanism ([86],
Hoffmann P and de Cathelineau A, unpublished observations) and thereby imply a two-step uptake process with a
requirement for apoptotic cell attachment and signaling
that, in many cases, may be mediated by different receptors (Figure 3).
The tether and tickle mechanism resembles that accompanying uptake of pathogenic Salmonella into epithelial cells.
Here the virulence factor SopE [87,88] is injected into the
cell by the bacterial Type III secretion system where it
activates members of the Rho family of GTPases and
causes membrane ruffling and macropinocytosis [89,90].
Salmonella lacking the virulence factor may still attach to
the cell surface but are not ingested unless ruffling and
Review
macropinocytosis are induced by alternative stimuli, such
as growth factors [91].
Ligation of some apoptotic cell recognition receptors such
as CD36 or αv integrins induces tethering of cells to macrophages but without uptake. Inclusion of phosphatidylserine on the target cell surface in this circumstance initiates
uptake. By contrast, phosphatidylserine on the target cell
membrane without the tethering ligand is ineffective, we
would suggest, because the affinity/avidity is too low to
initiate the process. Independent stimulation of the phosphatidylserine receptor in the presence of previously tethered cells also initiates their uptake — a bystander
mechanism akin to that outlined above for Salmonella.
This dual process may provide significant checks and balances in the removal process, minimizing for example,
uptake of cells that may transiently express phosphatidylserine during activation or of cells that may
become transiently attached to potential phagocytes in the
absence of the signaling receptor ligation, the tickle part of
the process.
Uptake of apoptotic cells by the tether and tickle mechanism would initially involve spacious phagosomes and the
concurrent ingestion of extracellular fluid. This was found
to be the case ([40], Hoffmann et al. unpublished observations), and was in marked contrast with uptake of cells,
even apoptotic cells, via the Fc receptor after opsonization;
here there is no evidence for fluid uptake into the phagosome. It should be noted that while macropinocytosis initially exhibits a spacious phagosome, this may soon be
followed by condensation of the phagosome to yield more
close apposition of the membranes to the ingested particles [89]. Although not studied to any degree, one might
expect differences in subsequent disposition of the phagosomes, in digestion of the apoptotic cells and final processing of the contents. The significant body of literature
showing macropinocytosis to be a key uptake mechanism
in antigen processing and presentation supports this, as
does the genetic evidence for specific genes involved in
corpse digestion in C. elegans [75,92].
Points of regulation
One aspect of regulation in the removal of apoptotic
corpses seen in mammalian systems is the stimulation of
macrophages to exhibit enhanced uptake. It is intriguing
to note that in most investigators’ hands, only a proportion
of macrophages, primary isolates or even cell lines, actually
engulf apoptotic cells in vitro. Whether this reflects subtypes of cells, varying abilities to undergo the putative
macropinocytotic process, cell–cell contact issues or heterogeneity in receptor expression, remains to be determined. Resident macrophages from lung or peritoneum
are markedly inefficient at apoptotic cell clearance as are
circulating monocytes. However, macrophages that have
R801
Figure 3
Apoptotic cell
Signaling ligands
Signaling receptors
Phagocyte
Tethering ligands
Tethering receptors
Signaling for membrane
ruffling and
macropinocytosis
Current Biology
A proposed two component uptake mechanism for apoptotic cells
involving a combination of tethering and signaling receptors — a ‘tether
and tickle’ process.
emigrated to, and matured within inflammatory sites are
very effective [93,94].
As monocytes mature into macrophages in vitro they too
develop an increased ability to recognize and ingest apoptotic cells. Part of this maturation may reflect stimulated
increase in expression of the phosphatidylserine receptor
[95,96]. It may also suggest levels of regulation that are
only beginning to be explored. For example, stimulation
of macrophage CD44 [97] and exposure to corticosteroids
[98] or to lipoxins [99] have each been shown to enhance
the ability of macrophages to recognize and engulf apoptotic cells. While the mechanisms for enhancement have
not been clarified, these processes do seem to be selective
for apoptotic cells.
Maturation of monocytes into immature dendritic cells
also increases their ability to ingest apoptotic cells. Immature dendritic cells show increased constitutive macropinocytosis so it may be that uptake in these cells is
regulated more by the nature of the tethering interaction
than specific stimulation of macropinocytotic signaling.
Regulation of uptake efficiency is expected in resident
tissue cells as well but has not yet been investigated to any
significant extent. It would also seem likely that other
combinations of stimuli and environmental factors yield
circumstances that block or delay apoptotic cell recognition and uptake. For example, defective clearance of apoptotic inflammatory cells in cystic fibrosis has recently been
attributed in part to cleavage of the recognition receptors
by elastase (Vandivier et al., unpublished observations).
R802
Current Biology Vol 11 No 19
Another area of increased interest is the likelihood that
cells, including apoptotic cells, may under certain circumstances express ‘don’t eat me’ signals. The demonstration
that CD47 expression by erythrocytes blocks their phagocytosis, through an interaction with the signal regulatory
protein SIRPα [100,101], lends credence to such a concept.
Whether this system, or others, operates in the context of
apoptotic cell uptake remains to be firmly established.
Consequences of recognition and removal
Apoptotic cell clearance is efficient and non-inflammatory
and may often be mediated in vivo by neighboring cells.
Great efforts are currently being expended on understanding the sequelae of apoptotic cell deletion, largely because
of its active anti-inflammatory and suggested anti-immunogenic effects. Such effects have been discussed in some
detail (see [9–11,79,102–104]). Following initial observations that uptake does not induce the release of inflammatory mediators, it soon became apparent that inflammation
is actively suppressed, both in vitro and in vivo, through
the production of inhibitory factors such as transforming
growth factor (TGF) β, prostaglandin E2 (PGE2) and interleukin-10 (IL-10).
We, and others, have implicated the phosphatidylserine
receptor and phosphatidylserine expression on the apoptotic cells as a major player in this suppression and have
recently suggested that its effect may be extended to
explain the inhibition of dendritic cell maturation and
antigen presentation in the adaptive immune response
[79,105]. This is in keeping with the suggested distinction
between the interaction with dendritic cells of apoptotic
cells — which inhibits maturation and antigen presentation — and necrotic cells [14,15,102,106–108]. It is worth
noting in this regard that induced apoptosis and cell deletion in normal tissues often leads to replenishment and
repair, in contrast to the removal of excess cells seen
during development. It seems likely to us that growth and
differentiation factors other than TGFβ may be produced
in response to apoptotic cell recognition that could contribute to this return to normal structure and function.
The obverse of this overall beneficial and essentially
normal process are the circumstances where suppression of
inflammation or immunity is essentially harmful. A clear
example of this was shown in tissue responses to Trypanosoma cruzi, the cause of Chagas disease. Here, apoptotic cells enhance the survival of the parasites in macrophages through their induction of TGFβ and PGE2 [109].
Similarly there is increasing evidence that apoptosis in
tumor cells might also have an overall negative effect on
the host as a consequence of local suppression of inflammation and immunity. TGFβ itself has long been associated with the development of fibrotic reactions in tissues
— a process that may lead to an important restoration of
structural integrity in a skin wound, but long-term impairment in function in the lung or liver. These types of
responses emphasize the need for balance in the response
to apoptotic cell removal in normal cell turnover.
In situations where cells are produced to excess, for
example myofibroblasts in fibrotic wound healing, even
here the processes of apoptosis and cell clearance appear
critical to their resolution. A number of studies have
addressed the ability of macrophages not only to engulf
apoptotic cells but also to induce apoptosis in appropriate
targets, especially in the context of tissue reorganization
during development or after inflammatory or fibrotic
changes [110–112]. Although not explicitly investigated in
most of these experimental systems, a coordinated induction of apoptosis and cell removal seems likely to occur.
Potential mechanisms include the production of oxidants,
nitric oxide (NO), tumor necrosis factor (TNF) α or Fas
ligand (FasL), and attachment of the macrophage to the
target cell could readily enhance both apoptosis and subsequent removal.
The genetic approach already discussed in C. elegans has
recently made an intriguing contribution to our understanding of the combined apoptosis and engulfment process
[113,114]. Apoptosis in worms is blocked in the absence of
CED-3 or CED-4. However, in partial ced-3 mutants,
showing only reduced numbers of apoptotic cells, the combined mutation of engulfment genes blocks apoptosis. The
implication is that engulfment can contribute to the induction of cell death initiated by less than optimal activation of
caspases. Once again the mechanisms are not yet defined
although Green and Beere [115] have suggested a possible
early discharge of lysosomal hydrolases akin to processes
described many years ago in mammalian phagocytes
[116–118] and colloquially called ‘frustrated phagocytosis’
or ‘regurgitation while feeding’. The two C. elegans studies
also suggest that the terminal, caspase-dependent ‘execution’ step in apoptosis may not be as irreversible as is often
implied — a concept that is perhaps more readily understood in the apparently more complex regulation processes
seen during apoptosis in mammalian cells [119].
The take-home message for our understanding of apoptotic cell clearance is that the complexities of apoptotic
cell engulfment are beginning to succumb to investigation,
are likely to be unique and primitive, and appear to
exhibit remarkable similarities across disparate members
of the animal kingdom.
References
1. Kerr JF, Wyllie AH, Currie AR: Apoptosis: a basic biological
phenomenon with wide-ranging implications in tissue kinetics.
Br J Cancer 1972, 26:239-257.
2. Wyllie AH, Kerr JF, Currie AR: Cell death: the significance of
apoptosis. Int Rev Cytol 1980, 68:251-306.
Review
3. Cohen J: Knife-edge of design. Nature 2001, 411:529.
4. Gumienny TL, Lambie E, Hartwieg E, Horvitz HR, Hengartner MO:
Genetic control of programmed cell death in the Caenorhabditis
elegans hermaphrodite germline. Development 1999,
126:1011-1022.
5. Chung S, Gumienny TL, Hengartner MO, Driscoll M: A common set
of engulfment genes mediates removal of both apoptotic and
necrotic cell corpses in C. elegans. Nat Cell Biol 2000, 2:931-937.
6. Liu QA, Hengartner MO: The molecular mechanism of programmed
cell death in C. elegans. Ann New York Acad Sci 1999, 887:92-104.
7. Zhou Z, Hartwieg E, Horvitz HR: CED-1 is a transmembrane
receptor that mediates cell corpse engulfment in C. elegans. Cell
2001, 104:43-56.
8. Gumienny TL, Hengartner MO: How the worm removes corpses:
the nematode C. elegans as a model system to study engulfment.
Cell Death Differ 2001, 8:564-568.
9. Platt N, da Silva RP, Gordon S: Recognizing death: the
phagocytosis of apoptotic cells. Trends Cell Biol 1998, 8:365-372.
10. Fadok VA, Henson PM: Apoptosis: getting rid of the bodies. Curr
Biol 1998, 8:R693-695.
11. Savill J, Fadok V: Corpse clearance defines the meaning of cell
death. Nature 2000, 407:784-788.
12. Fadok VA, Bratton DL, Rose DM, Pearson A, Ezekewitz RA, Henson
PM: A receptor for phosphatidylserine-specific clearance of
apoptotic cells. Nature 2000, 405:85-90.
13. Fadok VA, Bratton DL, Henson PM: Phagocyte receptors for
apoptotic cells: recognition, uptake and consequences. J Clin
Invest 2001, in press.
14. Fadok V, Bratton DL, Guthrie L, Henson PM: Differential effects of
apoptotic vs. lysed cells on macrophage production of cytokines:
role of proteases. J Immunol 2001, 166:6847-6854.
15. Sauter B, Albert ML, Francisco L, Larsson M, Somersan S, Bhardwaj
N: Consequences of cell death: exposure to necrotic tumor cells,
but not primary tissue cells or apoptotic cells, induces the
maturation of immunostimulatory dendritic cells. J Exp Med 2000,
191:423-434.
16. Moynault A, Luciani MF, Chimini G: ABC1, the mammalian
homologue of the engulfment gene ced-7, is required during
phagocytosis of both necrotic and apoptotic cells. Biochem Soc
Trans 1998, 26:629-635.
17. Chimini G: Engulfing by lipids: a matter of taste. Cell Death Differ
2001, 8:545-548.
18. Bodzioch M, Orso E, Klucken J, Langmann T, Bottcher A, Diederich
W, Drobnik W, Barlage S, Buchler C, Porsch-Ozcurumez M, et al.:
The gene encoding ATP-binding cassette transporter 1 is mutated
in Tangier disease. Nat Genet 1999, 22:347-351.
19. Rust S, Rosier M, Funke H, Real J, Amoura Z, Piette JC, Deleuze JF,
Brewer HB, Duverger N, Denefle P, Assmann G: Tangier disease is
caused by mutations in the gene encoding ATP-binding cassette
transporter 1. Nat Genet 1999, 22:352-355.
20. Lawn RM, Wade DP, Garvin MR, Wang X, Schwartz K, Porter JG,
Seilhamer JJ, Vaughan AM, Oram JF: The Tangier disease gene
product ABC1 controls the cellular apolipoprotein-mediated lipid
removal pathway. J Clin Invest 1999, 104:R25-R31.
21. Wood W, Turmaine M, Weber R, Camp V, Maki RA, McKercher SR,
Martin P: Mesenchymal cells engulf and clear apoptotic footplate
cells in macrophageless PU.1 null mouse embryos. Development
2000, 127:5245-5252.
22. Fadok VA, Voelker DR, Campbell PA, Cohen JJ, Bratton DL,
Henson PM: Exposure of phosphatidylserine on the surface of
apoptotic lymphocytes triggers specific recognition and removal
by macrophages. J Immunol 1992, 148:2207-2216.
23. Nera MS, Vanderbeek G, Johnson RO, Ruben LN, Clothier RH:
Phosphatidylserine expression on apoptotic lymphocytes of
Xenopus laevis, the South African clawed toad, as a signal for
macrophage recognition. Dev Comp Immunol 2000, 24:641-652.
24. van den Eijnde SM: Phosphatidylserine exposure by apoptotic cells
is phylogenetically conserved. Apoptosis 1998, 3:9-16.
25. Ding J, Wu Z, Crider BP, Ma Y, Li X, Slaughter C, Gong L, Xie XS:
Identification and functional expression of four isoforms of
ATPase II, the putative aminophospholipid translocase. Effect of
isoform variation on the ATPase activity and phospholipid
specificity. J Biol Chem 2000, 275:23378-23386.
26. Wiedmer T, Zhou Q, Kwoh DY, Sims PJ: Identification of three new
members of the phospholipid scramblase gene family. Biochim
Biophys Acta 2000, 1467:244-253.
R803
27. Tang X, Halleck MS, Schlegel RA, Williamson P: A subfamily of Ptype ATPases with aminophospholipid transporting activity.
Science 1996, 272:1495-1497.
28. Zhou Q, Zhao J, Stout JG, Luhm RA, Wiedmer T, Sims PJ: Molecular
cloning of human plasma membrane phospholipid scramblase.
A protein mediating transbilayer movement of plasma membrane
phospholipids. J Biol Chem 1997, 272:18240-18244.
29. Gomes E, Jakobsen MK, Axelsen KB, Geisler M, Palmgren MG:
Chilling tolerance in Arabidopsis involves ALA1, a member of a
new family of putative aminophospholipid translocases. Plant Cell
2000, 12:2441-2454.
30. Chen CY, Ingram MF, Rosal PH, Graham TR: Role for Drs2p, a
P-type ATPase and potential aminophospholipid translocase, in
yeast late Golgi function. J Cell Biol 1999, 147:1223-1236.
31. Daleke DL, Lyles JV: Identification and purification of aminophospholipid flippases. Biochim Biophys Acta 2000, 1486:108-127.
32. Morris RG, Hargreaves AD, Duvall E, Wyllie AH: Hormone-induced
cell death. 2. Surface changes in thymocytes undergoing
apoptosis. Am J Pathol 1984, 115:426-436.
33. Duvall E, Wyllie AH, Morris RG: Macrophage recognition of cells
undergoing programmed cell death (apoptosis). Immunology
1985, 56:351-358.
34. Savill JS, Henson PM, Haslett C: Phagocytosis of aged human
neutrophils by macrophages is mediated by a novel ‘chargesensitive’ recognition mechanism. J Clin Invest 1989, 84:1518-1527.
35. Dini L: Recognizing death: liver phagocytosis of apoptotic cells.
Eur J Histochem 2000, 44:217-227.
36. Falasca L, Bergamini A, Serafino A, Balabaud C, Dini L: Human
Kupffer cell recognition and phagocytosis of apoptotic peripheral
blood lymphocytes. Exp Cell Res 1996, 224:152-162.
37. Tenner AJ: Membrane receptors for soluble defense collagens.
Curr Opin Immunol 1999, 11:34-41.
38. Korb LC, Ahearn JM: C1q binds directly and specifically to surface
blebs of apoptotic human keratinocytes: complement deficiency
and systemic lupus erythematosus revisited. J Immunol 1997,
158:4525-4528.
39. Navratil JS, Watkins SC, Wisnieski JJ, Ahearn JM: The globular
heads of C1q specifically recognize surface blebs of apoptotic
vascular endothelial cells. J Immunol 2001, 166:3231-3239.
40. Ogden CA, de Cathelineau A, Hoffmann PR, Bratton DL,
Ghebrehiwet B, Fadok VA, Henson PM: C1q and Mannose Binding
Lectin (MBL) engagement of cell surface calreticulin and CD91
initiates macropinocytosis and uptake of apoptotic cells.
J Exp Med 2001, in press.
41. Frasch SC, Henson PM, Kailey JM, Richter DA, Janes MS, Fadok VA,
Bratton DL: Regulation of phospholipid scramblase activity during
apoptosis and cell activation by protein kinase Cdelta. J Biol Chem
2000, 275:23065-23073.
42. Toti F, Schindler V, Riou JF, Lombard-Platet G, Fressinaud E, Meyer D,
Uzan A, Le Pecq JB, Mandel JL, Freyssinet JM: Another link between
phospholipid transmembrane migration and ABC transporter
gene family, inferred from a rare inherited disorder of
phosphatidylserine externalization. Biochem Biophys Res Commun
1997, 241:548-552.
43. Hamon Y, Broccardo C, Chambenoit O, Luciani MF, Toti F, Chaslin S,
Freyssinet JM, Devaux PF, McNeish J, Marguet D, Chimini G: ABC1
promotes engulfment of apoptotic cells and transbilayer
redistribution of phosphatidylserine. Nat Cell Biol 2000, 2:399-406.
44. Stout JG, Zhou Q, Wiedmer T, Sims PJ: Change in conformation of
plasma membrane phospholipid scramblase induced by
occupancy of its Ca2+ binding site. Biochemistry 1998,
37:14860-14866.
45. Basse F, Stout JG, Sims PJ, Wiedmer T: Isolation of an erythrocyte
membrane protein that mediates Ca2+-dependent transbilayer
movement of phospholipid. J Biol Chem 1996, 271:17205-17210.
46. Callahan MK, Williamson P, Schlegel RA: Surface expression of
phosphatidylserine on macrophages is required for phagocytosis
of apoptotic thymocytes. Cell Death Differ 2000, 7:645-653.
47. Schlegel RA, Callahan MK, Williamson P: The central role of
phosphatidylserine in the phagocytosis of apoptotic thymocytes.
Ann New York Acad Sci 2000, 926:217-225.
48. Fadok VA, de Cathelineau A, Daleke DL, Henson PM, Bratton DL:
Loss of phospholipid asymmetry and surface exposure of
phosphatidylserine is required for phagocytosis of apoptotic cells
by macrophages and fibroblasts. J Biol Chem 2001,
276:1071-1077.
R804
Current Biology Vol 11 No 19
49. Fadok VA, McDonald PP, Bratton DL, Henson PM: Regulation of
macrophage cytokine production by phagocytosis of apoptotic
and post-apoptotic cells. Biochem Soc Trans 1998, 26:653-656.
50. Rigotti A, Acton SL, Krieger M: The class B scavenger receptors
SR-BI and CD36 are receptors for anionic phospholipids. J Biol
Chem 1995, 270:16221-16224.
51. Shiratsuchi A, Kawasaki Y, Ikemoto M, Arai H, Nakanishi Y: Role of
class B scavenger receptor type I in phagocytosis of apoptotic rat
spermatogenic cells by Sertoli cells. J Biol Chem 1999,
274:5901-5908.
52. Svensson PA, Johnson MS, Ling C, Carlsson LM, Billig H, Carlsson B:
Scavenger receptor class B type I in the rat ovary: possible role in
high density lipoprotein cholesterol uptake and in the recognition
of apoptotic granulosa cells. Endocrinology 1999, 140:2494-2500.
53. Fukasawa M, Adachi H, Hirota K, Tsujimoto M, Arai H, Inoue K: SRB1,
a class B scavenger receptor, recognizes both negatively charged
liposomes and apoptotic cells. Exp Cell Res 1996, 222:246-250.
54. Tait JF, Smith C: Phosphatidylserine receptors: role of CD36 in
binding of anionic phospholipid vesicles to monocytic cells. J Biol
Chem 1999, 274:3048-3054.
55. Nakano T, Ishimoto Y, Kishino J, Umeda M, Inoue K, Nagata K,
Ohashi K, Mizuno K, Arita H: Cell adhesion to phosphatidylserine
mediated by a product of growth arrest-specific gene 6. J Biol
Chem 1997, 272:29411-29414.
56. Ishimoto Y, Ohashi K, Mizuno K, Nakano T: Promotion of the uptake
of PS liposomes and apoptotic cells by a product of growth
arrest-specific gene, gas6. J Biochem 2000, 127:411-417.
57. Scott RS, McMahon EJ, Pop SM, Reap EA, Caricchio R, Cohen PL,
Earp HS, Matsushima GK: Phagocytosis and clearance of apoptotic
cells is mediated by MER. Nature 2001, 411:207-211.
58. Schagat TL, Wofford JA, Wright JR: Surfactant protein A enhances
alveolar macrophage phagocytosis of apoptotic neutrophils.
J Immunol 2001, 166:2727-2733.
59. Taylor PR, Carugati A, Fadok VA, Cook HT, Andrews M, Carroll MC,
Savill JS, Henson PM, Botto M, Walport MJ: A hierarchical role for
classical pathway complement proteins in the clearance of
apoptotic cells in vivo. J Exp Med 2000, 192:359-366.
60. Platt N, Suzuki H, Kurihara Y, Kodama T, Gordon S: Role for the
class A macrophage scavenger receptor in the phagocytosis of
apoptotic thymocytes in vitro. Proc Natl Acad Sci USA 1996,
93:12456-12460.
61. Ren Y, Silverstein RL, Allen J, Savill J: CD36 gene transfer confers
capacity for phagocytosis of cells undergoing apoptosis. J Exp
Med 1995, 181:1857-1862.
62. Fadok VA, Warner ML, Bratton DL, Henson PM: CD36 is required for
phagocytosis of apoptotic cells by human macrophages that use
either a phosphatidylserine receptor or the vitronectin receptor
(alpha v beta 3). J Immunol 1998, 161:6250-6257.
63. Albert ML, Pearce SF, Francisco LM, Sauter B, Roy P, Silverstein RL,
Bhardwaj N: Immature dendritic cells phagocytose apoptotic cells
via alphavbeta5 and CD36, and cross-present antigens to
cytotoxic T lymphocytes. J Exp Med 1998, 188:1359-1368.
64. Navazo MD, Daviet L, Savill J, Ren Y, Leung LL, McGregor JL:
Identification of a domain (155-183) on CD36 implicated in the
phagocytosis of apoptotic neutrophils. J Biol Chem 1996,
271:15381-15385.
65. Savill J, Hogg N, Ren Y, Haslett C: Thrombospondin cooperates with
CD36 and the vitronectin receptor in macrophage recognition of
neutrophils undergoing apoptosis. J Clin Invest 1992, 90:1513-1522.
66. Imachi H, Murao K, Hiramine C, Sayo Y, Sato M, Hosokawa H, Ishida T,
Kodama T, Quehenberger O, Steinberg D, Takahara J: Human
scavenger receptor B1 is involved in recognition of apoptotic
thymocytes by thymic nurse cells. Lab Invest 2000, 80:263-270.
67. Franc NC, Heitzler P, Ezekowitz RA, White K: Requirement for
croquemort in phagocytosis of apoptotic cells in Drosophila.
Science 1999, 284:1991-1994.
68. Franc NC, Dimarcq JL, Lagueux M, Hoffmann J, Ezekowitz RA:
Croquemort, a novel Drosophila hemocyte/macrophage receptor
that recognizes apoptotic cells. Immunity 1996, 4:431-443.
69. Finnemann SC, Bonilha VL, Marmorstein AD, Rodriguez-Boulan E:
Phagocytosis of rod outer segments by retinal pigment epithelial
cells requires alpha(v)beta5 integrin for binding but not for
internalization. Proc Natl Acad Sci USA 1997, 94:12932-12937.
70. Adachi H, Tsujimoto M, Arai H, Inoue K: Expression cloning of a
novel scavenger receptor from human endothelial cells. J Biol
Chem 1997, 272:31217-31220.
71. Moffatt OD, Devitt A, Bell ED, Simmons DL, Gregory CD:
Macrophage recognition of ICAM-3 on apoptotic leukocytes.
J Immunol 1999, 162:6800-6810.
72. Mevorach D: Opsonization of apoptotic cells. Implications for
uptake and autoimmunity. Ann New York Acad Sci 2000,
926:226-235.
73. Mevorach D, Mascarenhas JO, Gershov D, Elkon KB: Complementdependent clearance of apoptotic cells by human macrophages.
J Exp Med 1998, 188:2313-2320.
74. Takizawa F, Tsuji S, Nagasawa S: Enhancement of macrophage
phagocytosis upon iC3b deposition on apoptotic cells. FEBS Lett
1996, 397:269-272.
75. Ellis RE, Jacobson DM, Horvitz HR: Genes required for the
engulfment of cell corpses during programmed cell death in
Caenorhabditis elegans. Genetics 1991, 129:79-94.
76. Hengartner MO: Apoptosis: corralling the corpses. Cell 2001,
104:325-328.
77. Liu QA, Hengartner MO: Human CED-6 encodes a functional
homologue of the Caenorhabditis elegans engulfment protein
CED-6. Curr Biol 1999, 9:1347-1350.
78. Su HP, Brugnera E, Van Criekinge W, Smits E, Hengartner M,
Bogaert T, Ravichandran KS: Identification and characterization of a
dimerization domain in CED-6, an adapter protein involved in
engulfment of apoptotic cells. J Biol Chem 2000, 275:9542-9549.
79. Tosello-Trampont A-C, Brugnera E, Ravichandran KS: Evidence for a
conserved role for CrkII and Rac in engulfment of apoptotic cells.
J Biol Chem 2001, 276:13797-13802.
80. Leverrier Y, Ridley AJ: Requirement for Rho GTPases and PI 3kinases during apoptotic cell phagocytosis by macrophages. Curr
Biol 2001, 11:195-199.
81. Parnaik R, Raff MC, Scholes J: Differences between the clearance
of apoptotic cells by professional and non-professional
phagocytes. Curr Biol 2000, 10:857-860.
82. Griffin FM, Jr, Griffin JA, Leider JE, Silverstein SC: Studies on the
mechanism of phagocytosis. I. Requirements for circumferential
attachment of particle-bound ligands to specific receptors on the
macrophage plasma membrane. J Exp Med 1975, 142:1263-1282.
83. Allen LA, Aderem A: Molecular definition of distinct cytoskeletal
structures involved in complement- and Fc receptor-mediated
phagocytosis in macrophages. J Exp Med 1996, 184:627-637.
84. Allen LA, Aderem A: Mechanisms of phagocytosis. Curr Opin
Immunol 1996, 8:36-40.
85. Swanson JA, Baer SC: Phagocytosis by zippers and triggers.
Trends Cell Biol 1995, 5:89-93.
86. Henson PM, Bratton DL, Fadok VA: The phosphatidylserine
receptor: a crucial molecular switch. Nat Rev Mol Cell Biol 2001,
2:7-15.
87. Hardt WD, Chen LM, Schuebel KE, Bustelo XR, Galan JE: S.
typhimurium encodes an activator of Rho GTPases that induces
membrane ruffling and nuclear responses in host cells. Cell 1998,
93:815-826.
88. Rudolph MG, Weise C, Mirold S, Hillenbrand B, Bader B,
Wittinghofer A, Hardt WD: Biochemical analysis of SopE from
Salmonella typhimurium, a highly efficient guanosine nucleotide
exchange factor for RhoGTPases. J Biol Chem 1999,
274:30501-30509.
89. Alpuche-Aranda CM, Racoosin EL, Swanson JA, Miller SI: Salmonella
stimulate macrophage macropinocytosis and persist within
spacious phagosomes. J Exp Med 1994, 179:601-608.
90. Francis CL, Ryan TA, Jones BD, Smith SJ, Falkow S: Ruffles induced
by Salmonella and other stimuli direct macropinocytosis of
bacteria. Nature 1993, 364:639-642.
91. Galan JE, Pace J, Hayman MJ: Involvement of the epidermal growth
factor receptor in the invasion of cultured mammalian cells by
Salmonella typhimurium. Nature 1992, 357:588-589.
92. Lyon CJ, Evans CJ, Bill BR, Otsuka AJ, Aguilera RJ: The C. elegans
apoptotic nuclease NUC-1 is related in sequence and activity to
mammalian DNase II. Gene 2000, 252:147-154.
93. Newman SL, Henson JE, Henson PM: Phagocytosis of senescent
neutrophils by human monocyte-derived macrophages and rabbit
inflammatory macrophages. J Exp Med 1982, 156:430-442.
94. Fadok VA, Savill JS, Haslett C, Bratton DL, Doherty DE, Campbell PA,
Henson PM: Different populations of macrophages use either the
vitronectin receptor or the phosphatidylserine receptor to
recognize and remove apoptotic cells. J Immunol 1992,
149:4029-4035.
Review
95. Fadok VA, Voelker DR, Campbell PA, Bratton DL, Cohen JJ, Noble PW,
Riches DW, Henson PM: The ability to recognize phosphatidylserine
on apoptotic cells is an inducible function in murine bone marrowderived macrophages. Chest 1993, 103:102S.
96. Fadok VA, Warner ML, Bratton DL, Henson PM: CD36 is required for
phagocytosis of apoptotic cells by human macrophages that use
either a phosphatidylserine receptor or the vitronectin receptor
(alpha v beta 3). J Immunol 1998, 161:6250-6257.
97. Hart SP, Dougherty GJ, Haslett C, Dransfield I: CD44 regulates
phagocytosis of apoptotic neutrophil granulocytes, but not
apoptotic lymphocytes, by human macrophages. J Immunol 1997,
159:919-925.
98. Liu Y, Cousin JM, Hughes J, Van Damme J, Seckl JR, Haslett C,
Dransfield I, Savill J, Rossi AG: Glucocorticoids promote
nonphlogistic phagocytosis of apoptotic leukocytes. J Immunol
1999, 162:3639-3646.
99. Godson C, Mitchell S, Harvey K, Petasis NA, Hogg N, Brady HR:
Cutting edge: lipoxins rapidly stimulate nonphlogistic
phagocytosis of apoptotic neutrophils by monocyte-derived
macrophages. J Immunol 2000, 164:1663-1667.
100. Oldenborg PA, Gresham HD, Lindberg FP: CD47-signal regulatory
protein alpha (SIRPalpha) regulates Fcgamma and complement
receptor-mediated phagocytosis. J Exp Med 2001, 193:855-862.
101. Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD,
Lindberg FP: Role of CD47 as a marker of self on red blood cells.
Science 2000, 288:2051-2054.
102. Stern M, Savill J, Haslett C: Human monocyte-derived macrophage
phagocytosis of senescent eosinophils undergoing apoptosis.
Mediation by alpha v beta 3/CD36/thrombospondin recognition
mechanism and lack of phlogistic response. Am J Pathol 1996,
149:911-921.
103. Voll RE, Herrmann M, Roth EA, Stach C, Kalden JR, Girkontaite I:
Immunosuppressive effects of apoptotic cells. Nature 1997,
390:350-351.
104. Stach CM, Turnay X, Voll RE, Kern PM, Kolowos W, Beyer TD, Kalden
JR, Herrmann M: Treatment with annexin V increases
immunogenicity of apoptotic human T-cells in Balb/c mice. Cell
Death Differ 2000, 7:911-915.
105. Binder RJ, Han DK, Srivastava PK: CD91: a receptor for heat shock
protein gp96. Nat Immunol 2000, 1:151-155.
106. Basu S, Binder RJ, Suto R, Anderson KM, Srivastava PK: Necrotic but
not apoptotic cell death releases heat shock proteins, which
deliver a partial maturation signal to dendritic cells and activate
the NF-kappa B pathway. Int Immunol 2000, 12:1539-1546.
107. Gallucci S, Matzinger P: Danger signals: SOS to the immune
system: Curr Opin Immunol 2001, 13:114-119.
108. Cocco RE, Ucker DS: Distinct modes of macrophage recognition
for apoptotic and necrotic cells are not specified exclusively by
phosphatidylserine exposure. Mol Biol Cell 2001, 12:919-930.
109. Freire-de-Lima CG, Nascimento DO, Soares MB, Bozza PT, CastroFaria-Neto HC, de Mello FG, DosReis GA, Lopes MF: Uptake of
apoptotic cells drives the growth of a pathogenic trypanosome in
macrophages. Nature 2000, 403:199-203.
110. Lang RA, Bishop JM: Macrophages are required for cell death and
tissue remodeling in the developing mouse eye. Cell 1993,
74:453-462.
111. Diez-Roux G, Argilla M, Makarenkova H, Ko K, Lang RA: Macrophages
kill capillary cells in G1 phase of the cell cycle during programmed
vascular regression. Development 1999, 126:2141-2147.
112. Duffield JS, Erwig LP, Wei X, Liew FY, Rees AJ, Savill JS: Activated
macrophages direct apoptosis and suppress mitosis of mesangial
cells. J Immunol 2000, 164:2110-2119.
113. Hoeppner DJ, Hengartner MO, Schnabel R: Engulfment genes
cooperate with ced-3 to promote cell death in Caenorhabditis
elegans. Nature 2001, 412:202-206.
114. Reddien PW, Cameron S, Horvitz HR: Phagocytosis promotes
programmed cell death in C. elegans. Nature 2001, 412:198-202.
115. Green DR, Beere HM: Apoptosis. Mostly dead. Nature 2001,
412:133-135.
116 Henson PM: The immunologic release of constituents from
neutrophil leukocytes. II. Mechanisms of release during
phagocytosis, and adherence to nonphagocytosable surfaces.
J Immunol 1971, 107:1547-1557.
117. Henson PM: Mechanisms of exocytosis in phagocytic
inflammatory cells. Parke-Davis Award Lecture. Am J Pathol 1980,
101:494-511.
R805
118. Weissmann G, Serhan C, Korchak HM, Smolen JE: Mechanisms of
mediator release from neutrophils. Adv Exp Med Biol 1984,
172:527-552.
119. Leist M, Jaattela M: Four deaths and a funeral. From caspases to
alternative mechanisms. Nat Rev Mol Cell Biol 2001, 2:589-598.