Although millions of cells in the human body will undergo programmed cell death each day, dying cells are rarely detected under homeostatic settings in vivo. The swift removal of dying cells is due to the rapid recruitment of phagocytes to the site of cell death which then recognise and engulf the dying cell. Apoptotic cell clearance — the engulfment of apoptotic cells by phagocytes — is a well-defined process governed by a series of molecular factors including ‘find-me’, ‘eat-me’, ‘don't eat-me’ and ‘good-bye’ signals. However, in recent years with the rapid expansion of the cell death field, the removal of other necrotic-like cell types has drawn much attention. Depending on the type of death, dying cells employ different mechanisms to facilitate engulfment and elicit varying functional impacts on the phagocyte, from wound healing responses to inflammatory cytokine secretion. Nevertheless, despite the mechanism of death, the clearance of dying cells is a fundamental process required to prevent the uncontrolled release of pro-inflammatory mediators and inflammatory disease. This mini-review summarises the current understandings of: (i) apoptotic, necrotic, necroptotic and pyroptotic cell clearance; (ii) the functional consequences of dying cell engulfment and; (iii) the outstanding questions in the field.

For over 50 years apoptosis has dominated basic and translational research in the cell death field, representing the most well-characterised type of cell death. However, this traditionally immuno-silent form of programmed cell death represents just one of many pathways in which a cell can program itself to die. In 2018, twelve different regulated forms of cell death were described, highlighting the significant expansion of the cell death field [1]. In particular, the discovery of necroptosis and pyroptosis provides a significant contrast with the anti-inflammatory properties of apoptosis, and the stochastic nature of primary necrosis (Figure 1). Regardless of the mechanism of death, the swift removal of dying cells by professional (i.e. macrophages) and non-professional (i.e. epithelial cells) phagocytes remains paramount to maintain physiological homeostasis. For example, the induction of cell death and removal of dying cells has a fundamental role in embryonic development [2], tissue repair [3,4] and resolution of inflammation [5,6]. However, the persistence of dead cells and rupture of the plasma membrane allows the release of intracellular contents including damage-associated molecular patterns (DAMPs) which can trigger a robust inflammatory response [7–10]. Defective clearance of dying cells is closely associated with the onset and pathogenesis of inflammatory disease such as atherosclerosis [11,12], autoimmune disorders such as systemic lupus erythematosus and rheumatoid arthritis [9,13–16], and asthma [17]. Thus, dying cells employ a variety of mechanisms to recruit, be recognised and be engulfed by phagocytes. Here, this mini-review highlights the mechanisms underpinning dying cell clearance, and discusses the functional impact of phagocytosis on the surrounding environment.

Schematic summary of the molecular mechanisms driving cell death.

Figure 1.
Schematic summary of the molecular mechanisms driving cell death.

Intrinsic and extrinsic apoptotic signals are received and converge at the activation of caspase 3/7. Caspase 3-cleaved GSMDE and NINJ1 may facilitate the progression to secondary necrosis. Primary necrosis is traditionally stochastic in nature resulting in uncontrolled membrane rupture. In the presence of caspase inhibition, TNF-α can induce necroptosis through binding the TNF-R which results in the activation of RIPK1/3 and formation MLKL pores at the membrane. Pyroptosis is mediated via either the canonical (caspase 1) or non-canonical (caspase 4/5/11) pathway which converge with the formation of GSDMD membrane pores.

Figure 1.
Schematic summary of the molecular mechanisms driving cell death.

Intrinsic and extrinsic apoptotic signals are received and converge at the activation of caspase 3/7. Caspase 3-cleaved GSMDE and NINJ1 may facilitate the progression to secondary necrosis. Primary necrosis is traditionally stochastic in nature resulting in uncontrolled membrane rupture. In the presence of caspase inhibition, TNF-α can induce necroptosis through binding the TNF-R which results in the activation of RIPK1/3 and formation MLKL pores at the membrane. Pyroptosis is mediated via either the canonical (caspase 1) or non-canonical (caspase 4/5/11) pathway which converge with the formation of GSDMD membrane pores.

Close modal

Apoptosis

Apoptosis is largely driven by one of two pathways: the intrinsic/mitochondrial pathway and the extrinsic/receptor-mediated pathway. Both rely on the activation of the executioner caspases 3 and 7 to regulate the dismantling of the dying cell (Figure 1) [18]. Notably, cytotoxic lymphocyte killing also induces apoptosis via a Granzyme B-mediated mechanism. Similar to the induction of apoptosis, the clearance of apoptotic cells is a tightly controlled and well-studied process, and can be separated into three steps: recruitment, engagement and engulfment. Phagocytes are initially recruited to the site of apoptotic death by sensing ‘find-me’ signals actively released by apoptotic cells. These include soluble nucleotides such as ATP and UTP [19,20], sphinosine-1-phosphate (S1P) [21], lysophosphatidylcholine (LCP) [22] and MCP-1 [23]. Recruited phagocytes then engage with the apoptotic cell by binding either directly or via bridging molecules to an array of ‘eat-me’ signals exposed on the apoptotic cells outer-membrane. This includes calreticulin (CRT, both endogenous [24] and exogenous CRT secreted by phagocytes [25,26]), thrombospondin [27], ICAM3 [28], pentraxin 3 [29] and most notably, the phospholipid phosphatidylserine (PtdSer) [30]. Although normally located on the inner plasma membrane leaflet, PtdSer translocates to the outer leaflet during apoptosis through caspase 3/7 activation of the key scramblase Xrk8, and inactivation of flippases ATP11A and ATP11C which prevent PtdSer exposure on healthy cells [31–33].

Phagocytes are equipped with a diverse repertoire of engulfment receptors which engage directly with ‘eat-me’ signals, including TIM1/3/4 [34–36], BAI1 [37], RAGE [38], TLT2 [39], CD300b [40] and Stablin-2 [41]. Moreover, MFG-E8 and Gas6/Protein S can act as bridging molecules between PtdSer on the apoptotic cell and phagocytic intregrins and TAM receptors (Tyro, Axl, MerTK) to facilitate phagocytic-apoptotic cell engagement, respectively [42–45]. Similarly, the complement protein C1q and mannose-binding lectin can bind to exposed PtdSer and bridge with phagocytic receptors such as Megf10 and CRT/CD19 [46–49]. Apoptosis is also associated with dramatic DNA fragmentation, and a recent study identified that clusterin binding to histones exposed on the apoptotic cell surface exhibit opsoninic behaviour to aid cell clearance [50]. The efficiency of apoptotic cell clearance is also dependent on the apoptotic particle size. Recent studies have demonstrated that the fragmentation of apoptotic cells into extracellular vesicles known as apoptotic bodies (∼1–5 μm in diameter) is a tightly controlled process regulated by ROCK1 [51], PANX1 [52] and Plexin B2 [53]. As apoptotic bodies also expose the ‘eat-me’ signal PtdSer, the disassembly of apoptotic cells into apoptotic bodies generates numerous ‘bite-sized’ pieces that aid efficient engulfment by surrounding phagocytes [51,53,54]. Whether apoptotic bodies release ‘find-me’ signals to aid phagocytic recruitment to the initial site of cell death is unclear. Together, interactions between the phagocyte and the apoptotic fragments through these various mechanisms trigger an array of downstream signalling steps such as cytoskeletal reorganisation required to mediate phagocytosis [55].

It is important to note that ‘eat-me’ signal recognition can be attenuated if outcompeted by enhanced or clustered ‘don't eat-me’ signals such as CD47 [56,57], CD31 [58], and more recently CD24 [59] (Figure 2). The recognition of ‘don't eat-me’ signals negatively regulate engulfment, preventing the unnecessary clearance of healthy cells. However, cancer cells often exploit these mechanisms and up-regulate ‘don't eat-me’ signals to evade phagocytosis [60].

Molecular signals of dying cell clearance.

Figure 2.
Molecular signals of dying cell clearance.

Schematic summary of the molecular mechanisms which facilitate apoptotic, necrotic, necroptotic and pyroptotic cell clearance. This includes the exposure of ‘eat-me’ (green) and ‘don't eat-me’ (red) signals, secretion of ‘find-me’ signals (blue) and release of ‘good-bye’ signals (purple).

Figure 2.
Molecular signals of dying cell clearance.

Schematic summary of the molecular mechanisms which facilitate apoptotic, necrotic, necroptotic and pyroptotic cell clearance. This includes the exposure of ‘eat-me’ (green) and ‘don't eat-me’ (red) signals, secretion of ‘find-me’ signals (blue) and release of ‘good-bye’ signals (purple).

Close modal

Primary and secondary necrosis

Necrosis, either primary or secondary (occurring after the completion of apoptosis), is traditionally an unregulated form of cell death largely characterised by stochastic membrane lysis (Figure 1) [61]. It was recently suggested that caspase cleavage of Gasdermin E (GSDME) may mediate the progression of apoptosis to secondary necrosis through inducing membrane lysis [62] however, results are conflicting [10,63]. Alternatively, NINJ1 may regulate necrotic-cell membrane permeabilization [10]. In contrast with apoptotic cells which tightly regulate the activation of PANX1 channels and release of ‘find-me’ signals such as ATP, necrotic cells may stochastically release ATP as a by-product of uncontrolled membrane permeabilization [61]. Consequently, primary necrotic cells can release significantly higher levels of ATP (compared with apoptotic cells) and may be more efficient at inducing phagocyte recruitment [64]. Thus, ATP is a necrotic ‘find-me’ signal [64,65] which may function in concert or independently of other necrotic ‘find-me’ signals including formyl-peptides [66] and chemokines [67].

Once recruited to the site of cell death, phagocytes may internalised apoptotic and necrotic cells via different mechanisms (Figure 2) [68,69]. In comparison with the series of flippases and scramblases which regulate PtdSer exposure during apoptosis [31–33], the stochastic loss of phospholipid asymmetry by necrotic cells may result in limited or varying PtdSer exposure upon membrane lysis [64]. Although necrotic cells characteristically possess substantial Annexin V (AV) staining in flow cytometry-based assays [70], this should not be used to measure ‘eat-me’ signal exposure as AV will also bind PtdSer on the inner plasma membrane leaflet of necrotic cells. In fact, key phagocytic receptors such as TIM4 which mediate apoptotic cell engulfment through binding PtdSer poorly recognise necrotic cells [64]. Although necrotic cells can be cleared through PtdSer-mediated pathways [71–74], other mechanisms exist and contribute to their removal. This includes the exposure and binding of adhesion molecules such as E-cadherin [71], keratin [75], plasmin [76], as well as complement molecules including IgM, C1q and properdin [77–81] and HRG [82]. In a model of acute kidney injury, the protein AIM was also shown to label necrotic debris, interact with phagocytic KIM-1 and aid necrotic cell clearance, whereas the addition of AV to attenuate PtdSer recognition did not influence engulfment [83]. The phagocytic receptors CD14, CD36 and integrins αvβ3 may also contribute to the recognition of necrotic cell ‘eat-me’ signals and their timely removal [79].

Necroptosis

In comparison with the clearance of apoptotic and necrotic cells, the removal of cells dying via other cell death mechanisms such as necroptosis is only beginning to be defined. Necroptosis can be activated by the TNF pathway and is driven by the executioners RIPK1/3 and MLKL in the presence of caspase inhibition, such as during viral infections (Figure 1) [84]. Thus, many of the key regulators activated by caspase 3/7 (e.g PANX1 and Xrk8) required to facilitate clearance mechanisms (e.g. ATP release and PtdSer exposure) are not typically active during necroptosis. Although ATP release by necroptotic cells has been reported, whether this occurs prior to or as a consequence of membrane permeabilization has not been confirmed [85]. Recent findings have identified that necroptotic cells can expose PtdSer prior to membrane permeabilization, and this is dependent on the key necroptotic regulators RIPK3 and MLKL [86–88]. Therefore, the PtdSer-binding molecule MFG-E8 can recognise necroptotic cells and overexpression of the phagocytic receptor TIM4 can boost necroptotic cell clearance [88,89]. Similarly, supplementation of AV can attenuate necroptotic cell uptake [85]. In addition to PtdSer, the lipid mediator Resolvin D1 may also mediate necroptotic cell clearance by inducing phagocytic CRT secretion which can label and aid the recognition of necroptotic bone marrow-derived macrophages (BMDM) [90]. In contrasting findings, knock down or supplementation of soluble CRT was unable to alter the clearance of necroptotic fibroblasts [89] and may highlight potential cell-type specific clearance mechanisms. In line with this, as necroptotic BMDMs possess substantial expression of the ‘don't eat-me’ signal CD47, such cells may require additional factors, such as CRT, to mediate their efficient clearance [90]. Necroptotic cells also release small PtdSer positive extracellular vesicles akin to apoptotic bodies, termed necroptotic bodies [86,88]. Whether these necroptotic bodies also contribute to the efficiency of necroptotic cell clearance remains an unanswered question of interest.

Pyroptosis

Pyroptosis is an inflammatory form of programmed cell death triggered by the recognition of pathogen-associated molecular patterns such as bacterial LPS and DAMPs such as ATP [91]. It is initiated by either the canonical (caspase 1) or non-canonical pathway (caspase 4/5/11) which converge with activation of Gasdermin D (GSDMD) [91] (Figure 1). Similar to necroptotic cell clearance, the molecular mechanisms underpinning the removal of pyroptotic cells are still being defined (Figure 2). The clearance of pyroptotic cells is of significant interest as pyroptosis is widely implicated in inflammatory pathologies including Alzheimer's disease [92–94], liver fibrosis [95,96] and Salmonella infection [97,98]. Similar to the cleavage of PANX1 by caspase 3/7 during apoptosis, PANX1 is also activated by caspase 1/11 during pyroptosis and aids the release of ATP ‘find-me’ signals to mediate phagocytic recruitment [64,99–101]. Pyroptotic cells also secrete IL-1β and IL-18 in a cell-lysis independent manner through GSDMD pores to recruit phagocytes [102,103].

Once recruited to the site of cell death, phagocytes can engage with exposed PtdSer on the pyroptotic cell surface via bridging molecules (MFG-E8) or directly by scavenger receptors (TIM4) [64,89]. The mechanism of PtdSer exposure during pyroptosis is not dependent on caspase 1 [64] and whether it is an active or passive event remains elusive. Given that the phospholipid scramblase TMEM16F can be activated via Ca2+ signalling [104,105], whether such scramblases contribute to PtdSer exposure during cell death modalities without caspase 3/7 activity, such as pyroptosis, would be of interest to determine. Nevertheless, supplementation of AV has also been shown ineffective in blocking pyroptotic cell uptake, suggesting that other factors contribute to pyroptotic cell clearance [89,99]. In line with this, complement proteins can contribute to the rapid removal of pyroptotic cells, as mice deficient in the complement protein C3 are unable to recruit phagocytes to the site of death nor clear pyroptotic cells efficiently [99]. Clearance could further be impaired by broad inhibition of scavenger receptors, suggesting that C3 may act as a bridging molecule between pyroptotic cells and phagocytic scavenger receptors to mediate clearance [99].

The engulfment hierarchy

As the persistence of dying cells can trigger a breadth of inflammatory disease, swaying the mechanism of cell death to ensure swift, immunoprotective clearance is an exciting therapeutic potential. Moreover, understanding the engulfment hierarchy, i.e. which type of dying cells are cleared more efficiently, is of significant interest. Overall, the literature suggests that apoptotic cell clearance trumps the removal of necrotic-like cells [64,68,73,86,89,106]. As necrotic cells possess varying levels of the notable ‘eat-me’ signal PtdSer, phagocytic receptors may poorly recognise necrotic cells [64]. Consequently, phagocytes may require more time to engulf necrotic cells compared with their apoptotic counterparts [73]. Additionally, in comparison with apoptotic cells which rapidly bleb and fragment into apoptotic bodies, necrotic cells typically generate a single large bleb and remain as one cellular entity [107]. Given the role of dying cell fragmentation in aiding cell clearance [51,53], this may also provide a possible explanation for the inefficiency of necrotic cell clearance and the different mechanisms that contribute to their removal, compared with apoptotic cell uptake [68,73]. The clearance of apoptotic cells was also shown to be more efficient than necroptotic cell engulfment in both in vitro and in vivo settings, and also than pyroptotic cells in vitro [64,86,106]. However, contrasting findings have also been reported [89].

It is difficult to directly compare kinetics and phagocytic efficiencies between studies as the time post-induction of target cell death, phagocyte-to-target cell ratio and engulfment time often vary greatly. Moreover, kinetic comparison within studies must ensure equal levels of cell death to accurately compare phagocytic efficiencies. In vitro engulfment assays are also not representative of physiological conditions where various types of phagocytic cells (i.e. macrophages and epithelial cells) are present, and neighbouring cells may undergo different forms of cell death simultaneously (i.e. apoptosis or necrosis). Notably, competition phagocytosis assays have investigated whether apoptotic and necrotic cells could out-compete one another but results are conflicting [69,74]. Nevertheless, at a simplistic level, cells that expose ‘eat-me’ signals during the early stages of death (i.e. apoptosis), are expected to be cleared more rapidly [73]. Additionally, the secretion of multiple ‘find-me’ signals, vast number of ‘eat-me’ signals and significant redundancies in the phagocytic receptors which regulate apoptotic cell engulfment all strengthen the case for apoptotic cell clearance as the most efficient. However, increased interest in cell death and clearance pathways, and new findings such as the identification of PtdSer exposure prior to membrane permeabilization during necroptosis [86–88] may change our understanding of the engulfment hierarchy.

The consequence of death and dinner

Like the induction of cell death, the clearance of cells dying via different mechanisms can elicit distinct inflammatory signatures and impact the downstream immune response such as wound healing. Apoptosis is a traditionally immune-silent process which results in the direct or indirect release of anti-inflammatory mediators. For example, apoptotic cells secrete an array of anti-inflammatory factors such as IL-10, [108], TGF-β [109], and MFG-E8 [110]. Moreover, sensing of apoptotic ‘find-me’ signals such as S1P can both enhance cell clearance and induce phagocytic secretion of TGF-β, whilst decreasing pro-inflammatory factors like TNF-α and IL-6 [111]. Akin to ‘eat-me’ and ‘find-me’ signals, a new engulfment signal was recently described coined ‘good-bye’ signals. Apoptotic cells can release ‘good-bye’ signals in form of metabolites such as spermidine which induce anti-inflammatory gene expression in surrounding phagocytes, as well as wound healing, cytoskeletal organisation and anti-apoptotic responses [5]. The engulfment of apoptotic cells further contributes to inflammation control whereby upon uptake, phagocytes secrete anti-inflammatory factors including TGF-β [112,113] and IL-10 [114] and angiogenic factors to mediate wound healing such as VEGF [115], whilst limiting pro-inflammatory cytokine secretion [113]. Thus, not only do apoptotic cells prepare themselves for efficient clearance, but they also modulate the surrounding environment to prime phagocytes for engulfment and maintain anti-inflammatory conditions. As such, many studies have harnessed the anti-inflammatory properties of apoptotic cells and their clearance to combat robust inflammatory disease such as rheumatoid arthritis [5,16,116,117].

In contrast with the anti-inflammatory properties of apoptotic cells and their engulfment, necrosis is largely associated with robust inflammation. Necrotic cells undergo rapid membrane lysis and release a wide variety of intracellular contents including the pro-inflammatory cytokines IL-1α [118] and MIF [119], and DAMPs including HMGB1 [120,121], HSP70/90 [122,123] and DNA [121]. Although some studies have proposed necrotic cells to be more effective in recruiting phagocytes to the site of death through these signals [118], the pro-inflammatory consequence of necrosis likely outcompetes the benefit of rapid phagocytic recruitment. Moreover, once recruited to the site of necrosis, phagocytosis and sensing of necrotic debris can further exacerbate inflammation whereby phagocytes release pro-inflammatory cytokines such as IL-8 and TNF-α [82,124].

Necroptosis and pyroptosis are also associated with robust inflammation. Necroptotic cells release an array of pro-inflammatory mediators such as IL-8, IL-1, CXCL2 and cyclophilin A [125–127], and engulfment of necroptotic cells further exacerbates inflammation by triggering phagocytic TNF-α and MCP-1 secretion [86]. Similarly, pyroptotic cells secrete IL-1β, IL-18, TNF-α and IL-6 which can drive inflammatory disease such as liver fibrosis and arthritis [95,128]. Although the inflammatory consequence of pyroptotic cell clearance on the phagocytosing cell is unclear, engulfment of ‘NLRP3 inflammasome particles’ or the inflammasome-associated adaptor protein complexes ‘ASC specks’ can elicit phagocytic inflammatory cytokine secretion [96,129]. Altogether, as a single stimuli can elicit multiple forms of cell death, such as the induction of necrosis, apoptosis, necroptosis and potentially pyroptosis by influenza A virus [130], the functional impact of dead cell clearance in physiological settings and disease is complex and requires comprehensive investigations.

The swift removal of dying cells is paramount to prevent disease onset and understanding the molecular mechanisms underpinning their clearance is vital. Moreover, harnessing this knowledge to develop novel therapeutics and boost dead cell clearance in inflammatory disease settings where clearance is aberrant possesses exciting clinical potential. Although many of the major molecular components contributing to efficient cell clearance have been described, there still remains a significant knowledge gap.

Exploring the mechanistic differences between targets and phagocytes

Although recent literature has shed light on how necroptotic and pyroptotic cells are recognised and phagocytosed, this literature merely represents the tip of the iceberg. Given the vast number of machineries that mediate the removal of apoptotic and necrotic cells, other factors in addition to ones currently described likely contribute to necroptotic and pyroptotic cell clearance. Moreover, the clearance of cells undergoing alternative cell death pathways such as ferroptosis, parthanatos and NETosis is poorly understood but also of significant interest. For example, the defective clearance of NETs has been observed in inflammatory disease such as respiratory distress syndrome whereby soluble components in the bronchioalveolar lavage fluid of patients could impair phagocytic NET uptake [131]. Therefore, whether known or novel engulfment receptors contribute to NET removal would be of great interest. Understanding the different mechanisms of dead cell clearance is especially important for disease settings which elicit multiple cell death pathways simultaneously and potentially require a multifaceted therapeutic strategy. Moreover, whether professional and non-professional phagocytes can recognise their targets via different modalities or receptors, or whether uptake elicits a different response is yet to be determined. The interplay between professional and non-professional phagocytes is especially interesting as upon apoptotic cell engulfment, macrophages can secrete IGF-1 and enhance the phagocytic efficiency of surrounding non-professional phagocytes [6]. Whether professional phagocytes have a superior role in the clearance of inflammatory cells (i.e. pyroptotic cells), or differences within immune cell subsets exist (i.e. M1 vs M2 macrophages), are also outstanding questions. Notably, it was reported that ‘large’-DCs were more efficient in phagocytosing necrotic cells than ‘small’-DCs [132]. However, whether this was due to mechanistic differences rather than restricted size and phagocytic capacity is unclear.

Clinical potential of harnessing dead cell clearance

It is well established that aberrant clearance of dying cells can trigger inflammatory disease. For example, impairment of cell clearance by clusterin or MFG-E8 deficiency results in autoimmune disease-like symptoms [14,50]. Moreover, inflammation and atherosclerotic plaque formation may in part be due to the accumulation of apoptotic cells from impaired phagocytosis during atherosclerosis [11,12]. Therefore, boosting cell clearance represents an exciting therapeutic potential and may be achieved by exploiting various clearance mechanisms. Blocking the ‘don't eat-me’ signal CD47 with monoclonal antibodies during atherosclerosis has shown promise in boosting apoptotic cell clearance and reducing disease burden in mice [133]. Notably, targeting the expression of ‘eat-me’ signals is yet to be explored but may also represent a suitable approach. Up-regulating engulfment receptors or ‘priming’ macrophages to enhance engulfment likely represents the most efficient therapeutic strategy. Genetically overexpressing the phagocytic receptor BAI1 has already been shown to boost apoptotic cell engulfment in vivo and attenuate disease-associated inflammation in mice [134]. However, the translation of these therapeutic strategies to the clinic still remains a challenge.

Recent advances have revealed the tight molecular control underpinning the disassembly of apoptotic cells into apoptotic bodies and demonstrated the importance of this process in aiding rapid cell clearance [51,53]. Therefore, simultaneously inducing apoptosis and boosting the disassembly of apoptotic cells may provide an effective approach to enhance cell clearance in disease settings such as solid tumours where cell death and their swift removal is crucial. The antibiotic Trovafloxacin was identified as the first pharmaceutical enhancer of apoptotic body formation [52,135] and thus, Trovafloxacin or similar PANX1 inhibitors may be suitable candidates to investigate such therapeutic strategies. Furthermore, as mentioned above, whether necroptotic or pyroptotic cells also fragment into smaller vesicles (i.e. necroptotic bodies and pyroptotic bodies) which aid phagocytosis remains an unanswered question of clinical relevance.

The efficient removal of dying cells is regulated by a complex and redundant series of machineries which can elicit both pro- and anti-inflammatory effects on the phagocyte and surrounding environment. Although how phagocytes are recruited to, recognise and engulf other dying cells beyond apoptosis is still being defined, the ability to translate these findings clinically and treat inflammatory disease is an exciting prospect for the cell clearance field.

  • Cell death and the removal of dying cells is tightly linked to a variety of inflammatory disease. Thus, understanding the molecular mechanisms responsible for phagocytic clearance and the functional impact of engulfment on the phagocyte is essential for the development of novel disease therapeutics.

  • Efficient dead cell clearance can be split into three individual steps including recruitment, recognition and engulfment which are mediated by the release and exposure of ‘find-me’, ‘eat-me’, ‘don't eat-me’ and ‘good-bye’ signals.

  • With the rapid expansion of the cell death field, further research is needed to understand how different cell types, and cells dying via different mechanisms are cleared, the impact this has on the phagocytosing cell and how this can be targeted therapeutically.

The author declares that there are no competing interests associated with this manuscript.

I would like to acknowledge the CASS Foundation Medicine/Science Grants for their financial support.

Open access for this article was enabled by the participation of Walter and Eliza Hall Institute in an all-inclusive Read & Publish pilot with Portland Press and the Biochemical Society under a transformative agreement with CAUL.

G.K.A.-S. wrote and edited the manuscript.

I would like to thank A/Prof I Poon and J Liu for their scientific conversations, input and insights which helped construct this manuscript.

AIM

Apoptosis inhibitor of macrophage

ATP

Adenosine triphosphate

BAI1

Brain-specific angiogenesis inhibitor 1

DCs

Dendritic cells

FADD

Fas-associated protein with death domain

GSDMD-N

Gasdermin D N-terminal

GSDME-N

Gasdermin E N-terminal

ICAM3

Intracellular adhesion molecule 3

IGF-1

Insulin-like growth factor 1

MCP-1

Monocyte chemoattractant protein 1

MFG-E8

Milk fat globule epidermal growth factor 8

MLKL

Mixed lineage kinase domain-like

NINJ1

Nerve injury-induced protein 1

NLRP3

NLR family pyrin domain containing 3

P

Phosphorylation

PANX1

Pannexin 1

RAGE

Receptor for advanced glycation endproducts

RIPK1/3

Receptor-interacting serine/threonine-protein kinase 1/3

ROCK1

Rho-associated coiled-coil containing protein kinase 1

TNF

Tumour necrosis factor

TNFR

Tumour necrosis factor receptor

Ub

Ubiquitination

UTP

Uridine triphosphate

1
Galluzzi
,
L.
,
Vitale
,
I.
,
Aaronson
,
S.A.
,
Abrams
,
J.M.
,
Adam
,
D.
,
Agostinis
,
P.
et al. (
2018
)
Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018
.
Cell Death Differ.
25
,
486
541
2
Qu
,
X.
,
Zou
,
Z.
,
Sun
,
Q.
,
Luby-Phelps
,
K.
,
Cheng
,
P.
,
Hogan
,
R.N.
et al. (
2007
)
Autophagy gene-dependent clearance of apoptotic cells during embryonic development
.
Cell
128
,
931
946
3
Yang
,
Z.
,
Li
,
Q.
,
Wang
,
X.
,
Jiang
,
X.
,
Zhao
,
D.
,
Lin
,
X.
et al. (
2018
)
C-type lectin receptor LSECtin-mediated apoptotic cell clearance by macrophages directs intestinal repair in experimental colitis
.
Proc. Natl Acad. Sci. U.S.A.
115
,
11054
11059
4
Bosurgi
,
L.
,
Cao
,
Y.G.
,
Cabeza-Cabrerizo
,
M.
,
Tucci
,
A.
,
Hughes
,
L.D.
,
Kong
,
Y.
et al. (
2017
)
Macrophage function in tissue repair and remodeling requires IL-4 or IL-13 with apoptotic cells
.
Science
356
,
1072
1076
5
Medina
,
C.B.
,
Mehrotra
,
P.
,
Arandjelovic
,
S.
,
Perry
,
J.S.A.
,
Guo
,
Y.
,
Morioka
,
S.
et al. (
2020
)
Metabolites released from apoptotic cells act as tissue messengers
.
Nature
580
,
130
135
6
Han
,
C.Z.
,
Juncadella
,
I.J.
,
Kinchen
,
J.M.
,
Buckley
,
M.W.
,
Klibanov
,
A.L.
,
Dryden
,
K.
et al. (
2016
)
Macrophages redirect phagocytosis by non-professional phagocytes and influence inflammation
.
Nature
539
,
570
574
7
Poon
,
I.K.H.
,
Lucas
,
C.D.
,
Rossi
,
A.G.
and
Ravichandran
,
K.S.
(
2014
)
Apoptotic cell clearance: basic biology and therapeutic potential
.
Nat. Rev. Immunol.
14
,
166
180
8
Vanden Berghe
,
T.
,
Vanlangenakker
,
N.
,
Parthoens
,
E.
,
Deckers
,
W.
,
Devos
,
M.
,
Festjens
,
N.
et al. (
2010
)
Necroptosis, necrosis and secondary necrosis converge on similar cellular disintegration features
.
Cell Death Differ.
17
,
922
930
9
Cohen
,
P.L.
,
Caricchio
,
R.
,
Abraham
,
V.
,
Camenisch
,
T.D.
,
Charles Jennette
,
J.
,
Roubey
,
R.A.S.
et al. (
2002
)
Delayed apoptotic cell clearance and lupus-like autoimmunity in mice lacking the c-mer membrane tyrosine kinase
.
J. Exp. Med.
196
,
135
140
10
Kayagaki
,
N.
,
Kornfeld
,
O.S.
,
Lee
,
B.L.
,
Stowe
,
I.B.
,
O'Rourke
,
K.
,
Li
,
Q.
et al. (
2021
)
NINJ1 mediates plasma membrane rupture during lytic cell death
.
Nature
591
,
131
136
11
Schrijvers
,
D.M.
,
De Meyer
,
G.R.Y.
,
Kockx
,
M.M.
,
Herman
,
A.G.
and
Martinet
,
W.
(
2005
)
Phagocytosis of apoptotic cells by macrophages is impaired in atherosclerosis
.
Arterioscler. Thromb. Vasc. Biol.
25
,
1256
1261
12
Thorp
,
E.
,
Cui
,
D.
,
Schrijvers
,
D.M.
,
Kuriakose
,
G.
and
Tabas
,
I.
(
2008
)
Mertk receptor mutation reduces efferocytosis efficiency and promotes apoptotic cell accumulation and plaque necrosis in atherosclerotic lesions of apoe-/- mice
.
Arterioscler. Thromb. Vasc. Biol.
28
,
1421
1428
13
Herrmann
,
M.
,
Voll
,
R.E.
,
Zoller
,
O.M.
,
Hagenhofer
,
M.
,
Ponner
,
B.B.
and
Kalden
,
J.R.
(
1998
)
Impaired phagocytosis of apoptotic cell material by monocyte-derived macrophages from patients with systemic lupus erythematosus
.
Arthritis Rheum.
41
,
1241
1250
14
Hanayama
,
R.
,
Tanaka
,
M.
,
Miyasaka
,
K.
,
Aozasa
,
K.
,
Koike
,
M.
,
Uchiyama
,
Y.
et al. (
2004
)
Autoimmune disease and impaired uptake of apoptotic cells in MFG-E8-deficient mice
.
Science
304
,
1147
1150
15
Tas
,
S.W.
,
Quartier
,
P.
,
Botto
,
M.
and
Fossati-Jimack
,
L.
(
2006
)
Macrophages from patients with SLE and rheumatoid arthritis have defective adhesion in vitro, while only SLE macrophages have impaired uptake of apoptotic cells
.
Ann. Rheum. Dis.
65
,
216
221
16
Saas
,
P.
,
Bonnefoy
,
F.
,
Toussirot
,
E.
and
Perruche
,
S.
(
2017
)
Harnessing apoptotic cell clearance to treat autoimmune arthritis
.
Front. Immunol.
8
,
1191
17
Grabiec
,
A.M.
,
Denny
,
N.
,
Doherty
,
J.A.
,
Happonen
,
K.E.
,
Hankinson
,
J.
,
Connolly
,
E.
et al. (
2017
)
Diminished airway macrophage expression of the Axl receptor tyrosine kinase is associated with defective efferocytosis in asthma
.
J. Allergy Clin. Immunol.
140
,
1144
1146.e4
18
Elmore
,
S.
(
2007
)
Apoptosis: a review of programmed cell death
.
Toxicol. Pathol.
35
,
495
516
19
Elliott
,
M.R.
,
Chekeni
,
F.B.
,
Trampont
,
P.C.
,
Lazarowski
,
E.R.
,
Kadl
,
A.
,
Walk
,
S.F.
et al. (
2009
)
Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance
.
Nature
461
,
282
286
20
Chekeni
,
F.B.
,
Elliott
,
M.R.
,
Sandilos
,
J.K.
,
Walk
,
S.F.
,
Kinchen
,
J.M.
,
Lazarowski
,
E.R.
et al. (
2010
)
Pannexin 1 channels mediate “find-me” signal release and membrane permeability during apoptosis
.
Nature
467
,
863
867
21
Gude
,
D.R.
,
Alvarez
,
S.E.
,
Paugh
,
S.W.
,
Mitra
,
P.
,
Yu
,
J.
,
Griffiths
,
R.
et al. (
2008
)
Apoptosis induces expression of sphingosine kinase 1 to release sphingosine-1-phosphate as a “come-and-get-me” signal
.
FASEB J.
22
,
2629
2638
22
Lauber
,
K.
,
Bohn
,
E.
,
Kröber
,
S.M.
,
Xiao
,
Y.J.
,
Blumenthal
,
S.G.
,
Lindemann
,
R.K.
et al. (
2003
)
Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal
.
Cell
113
,
717
730
23
Cullen
,
S.P.
,
Henry
,
C.M.
,
Kearney
,
C.J.
,
Logue
,
S.E.
,
Feoktistova
,
M.
,
Tynan
,
G.A.
et al. (
2013
)
Fas/CD95-induced chemokines can serve as “ find-me” signals for apoptotic cells
.
Mol. Cell
49
,
1034
1048
24
Gardai
,
S.J.
,
McPhillips
,
K.A.
,
Frasch
,
S.C.
,
Janssen
,
W.J.
,
Starefeldt
,
A.
,
Murphy-Ullrich
,
J.E.
et al. (
2005
)
Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte
.
Cell
123
,
321
334
25
Feng
,
M.
,
Marjon
,
K.D.
,
Zhu
,
F.
,
Weissman-Tsukamoto
,
R.
,
Levett
,
A.
,
Sullivan
,
K.
et al. (
2018
)
Programmed cell removal by calreticulin in tissue homeostasis and cancer
.
Nat. Commun.
9
,
3194
26
Feng
,
M.
,
Chen
,
J.Y.
,
Weissman-Tsukamoto
,
R.
,
Volkmer
,
J.P.
,
Ho
,
P.Y.
,
McKenna
,
K.M.
et al. (
2015
)
Macrophages eat cancer cells using their own calreticulin as a guide: roles of TLR and Btk
.
Proc. Natl Acad. Sci. U.S.A.
112
,
2145
2150
27
Krispin
,
A.
,
Bledi
,
Y.
,
Atallah
,
M.
,
Trahtemberg
,
U.
,
Verbovetski
,
I.
,
Nahari
,
E.
et al. (
2006
)
Apoptotic cell thrombospondin-1 and heparin-binding domain lead to dendritic-cell phagocytic and tolerizing states
.
Blood
108
,
3580
3589
28
Moffatt
,
O.D.
,
Devitt
,
A.
,
Bell
,
E.D.
,
Simmons
,
D.L.
and
Gregory
,
C.D.
(
1999
)
Macrophage recognition of ICAM-3 on apoptotic leukocytes
.
J. Immunol.
162
,
6800
6810
PMID:
[PubMed]
29
Jaillon
,
S.
,
Jeannin
,
P.
,
Hamon
,
Y.
,
Frémaux
,
I.
,
Doni
,
A.
,
Bottazzi
,
B.
et al. (
2009
)
Endogenous PTX3 translocates at the membrane of late apoptotic human neutrophils and is involved in their engulfment by macrophages
.
Cell Death Differ.
16
,
465
474
30
Fadok
,
V.A.
,
De Cathelineau
,
A.
,
Daleke
,
D.L.
,
Henson
,
P.M.
and
Bratton
,
D.L.
(
2001
)
Loss of phospholipid asymmetry and surface exposure of phosphatidylserine is required for phagocytosis of apoptotic cells by macrophages and fibroblasts
.
J. Biol. Chem.
276
,
1071
1077
31
Nagata
,
S.
,
Suzuki
,
J.
,
Segawa
,
K.
and
Fujii
,
T.
(
2016
)
Exposure of phosphatidylserine on the cell surface
.
Cell Death Differ.
23
,
952
961
32
Segawa
,
K.
,
Kurata
,
S.
,
Yanagihashi
,
Y.
,
Brummelkamp
,
T.R.
,
Matsuda
,
F.
and
Nagata
,
S.
(
2014
)
Caspase-mediated cleavage of phospholipid flippase for apoptotic phosphatidylserine exposure
.
Science
344
,
1164
1168
33
Suzuki
,
J.
,
Denning
,
D.P.
,
Imanishi
,
E.
,
Horvitz
,
H.R.
and
Nagata
,
S.
(
2013
)
Xk-related protein 8 and CED-8 promote phosphatidylserine exposure in apoptotic cells
.
Science
341
,
403
406
34
Kobayashi
,
N.
,
Karisola
,
P.
,
Peña-Cruz
,
V.
,
Dorfman
,
D.M.
,
Jinushi
,
M.
,
Umetsu
,
S.E.
et al. (
2007
)
TIM-1 and TIM-4 glycoproteins bind phosphatidylserine and mediate uptake of apoptotic cells
.
Immunity
27
,
927
940
35
Nakayama
,
M.
,
Akiba
,
H.
,
Takeda
,
K.
,
Kojima
,
Y.
,
Hashiguchi
,
M.
,
Azuma
,
M.
et al. (
2009
)
Tim-3 mediates phagocytosis of apoptotic cells and cross-presentation
.
Blood
113
,
3821
3830
36
Miyanishi
,
M.
,
Tada
,
K.
,
Koike
,
M.
,
Uchiyama
,
Y.
,
Kitamura
,
T.
and
Nagata
,
S.
(
2007
)
Identification of Tim4 as a phosphatidylserine receptor
.
Nature
450
,
435
439
37
Park
,
D.
,
Tosello-Trampont
,
A.C.
,
Elliott
,
M.R.
,
Lu
,
M.
,
Haney
,
L.B.
,
Ma
,
Z.
et al. (
2007
)
BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module
.
Nature
450
,
430
434
38
He
,
M.
,
Kubo
,
H.
,
Morimoto
,
K.
,
Fujino
,
N.
,
Suzuki
,
T.
,
Takahasi
,
T.
et al. (
2011
)
Receptor for advanced glycation end products binds to phosphatidylserine and assists in the clearance of apoptotic cells
.
EMBO Rep.
12
,
358
364
39
de Freitas
,
A.
,
Banerjee
,
S.
,
Xie
,
N.
,
Cui
,
H.
,
Davis
,
K.I.
,
Friggeri
,
A.
et al. (
2012
)
Identification of TLT2 as an engulfment receptor for apoptotic cells
.
J. Immunol.
188
,
6381
6388
40
Murakami
,
Y.
,
Tian
,
L.
,
Voss
,
O.H.
,
Margulies
,
D.H.
,
Krzewski
,
K.
and
Coligan
,
J.E.
(
2014
)
CD300b regulates the phagocytosis of apoptotic cells via phosphatidylserine recognition
.
Cell Death Differ.
21
,
1746
1757
41
Park
,
S.Y.
,
Jung
,
M.Y.
,
Kim
,
H.J.
,
Lee
,
S.J.
,
Kim
,
S.Y.
,
Lee
,
B.H.
et al. (
2008
)
Rapid cell corpse clearance by stabilin-2, a membrane phosphatidylserine receptor
.
Cell Death Differ.
15
,
192
201
42
Anderson
,
H.A.
,
Maylock
,
C.A.
,
Williams
,
J.A.
,
Paweletz
,
C.P.
,
Shu
,
H.
and
Shacter
,
E.
(
2003
)
Serum-derived protein S binds to phosphatidylserine and stimulates the phagocytosis of apoptotic cells
.
Nat. Immunol.
4
,
87
91
43
Akakura
,
S.
,
Singh
,
S.
,
Spataro
,
M.
,
Akakura
,
R.
,
Kim
,
J.
,
Albert
,
M.L.
et al. (
2004
)
The opsonin MFG-E8 is a ligand for the αvβ5 integrin and triggers DOCK180-dependent Rac1 activation for the phagocytosis of apoptotic cells
.
Exp. Cell Res.
292
,
403
416
44
Hanayama
,
R.
,
Tanaka
,
M.
,
Miwa
,
K.
,
Shinohara
,
A.
,
Iwamatsu
,
A.
and
Nagata
,
S.
(
2002
)
Identification of a factor that links apoptotic cells to phagocytes
.
Nature
417
,
182
187
45
Dransfield
,
I.
,
Zagórska
,
A.
,
Lew
,
E.D.
,
Michail
,
K.
and
Lemke
,
G.
(
2015
)
Mer receptor tyrosine kinase mediates both tethering and phagocytosis of apoptotic cells
.
Cell Death Dis.
6
,
e1646
46
Iram
,
T.
,
Ramirez-Ortiz
,
Z.
,
Byrne
,
M.H.
,
Coleman
,
U.A.
,
Kingery
,
N.D.
,
Means
,
T.K.
et al. (
2016
)
Megf10 Is a receptor for C1Q that mediates clearance of apoptotic cells by astrocytes
.
J. Neurosci.
36
,
5185
5192
47
Taylor
,
P.R.
,
Carugati
,
A.
,
Fadok
,
V.A.
,
Cook
,
H.T.
,
Andrews
,
M.
,
Carroll
,
M.C.
et al. (
2000
)
A hierarchical role for classical pathway complement proteins in the clearance of apoptotic cells in vivo
.
J. Exp. Med.
192
,
359
366
48
Païdassi
,
H.
,
Tacnet-Delorme
,
P.
,
Garlatti
,
V.
,
Darnault
,
C.
,
Ghebrehiwet
,
B.
,
Gaboriaud
,
C.
et al. (
2008
)
C1q binds phosphatidylserine and likely acts as a multiligand-bridging molecule in apoptotic cell recognition
.
J. Immunol.
180
,
2329
2338
49
Ogden
,
C.A.
,
DeCathelineau
,
A.
,
Hoffmann
,
P.R.
,
Bratton
,
D.
,
Fadok
,
B.
,
Ghebrehiwet
,
V.A.
et al. (
2001
)
C1q and mannose binding lectin engagement of cell surface calreticulin and CD91 initiates macropinocytosis and uptake of apoptotic cells
.
J. Exp. Med.
194
,
781
796
50
Cunin
,
P.
,
Beauvillain
,
C.
,
Miot
,
C.
,
Augusto
,
J.F.
,
Preisser
,
L.
,
Blanchard
,
S.
et al. (
2016
)
Clusterin facilitates apoptotic cell clearance and prevents apoptotic cell-induced autoimmune responses
.
Cell Death Dis.
7
,
e2215
51
Tixeira
,
R.
,
Phan
,
T.K.
,
Caruso
,
S.
,
Shi
,
B.
,
Atkin-Smith
,
G.K.
,
Nedeva
,
C.
et al. (
2020
)
ROCK1 but not LIMK1 or PAK2 is a key regulator of apoptotic membrane blebbing and cell disassembly
.
Cell Death Differ.
27
,
102
116
52
Poon
,
I.K.H.
,
Chiu
,
Y.H.
,
Armstrong
,
A.J.
,
Kinchen
,
J.M.
,
Juncadella
,
I.J.
,
Bayliss
,
D.A.
et al. (
2014
)
Unexpected link between an antibiotic, pannexin channels and apoptosis
.
Nature
507
,
329
334
53
Atkin-Smith
,
G.K.
,
Miles
,
M.A.
,
Tixeira
,
R.
,
Lay
,
F.T.
,
Duan
,
M.
,
Hawkins
,
C.J.
et al. (
2019
)
Plexin B2 is a regulator of monocyte apoptotic cell disassembly
.
Cell Rep.
29
,
1821
1831.e3
54
Witasp
,
E.
,
Uthaisang
,
W.
,
Elenström-Magnusson
,
C.
,
Hanayama
,
R.
,
Tanaka
,
M.
,
Nagata
,
S.
et al. (
2007
)
Bridge over troubled water: milk fat globule epidermal growth factor 8 promotes human monocyte-derived macrophage clearance of non-blebbing phosphatidylserine-positive target cells [2]
.
Cell Death Differ.
14
,
1063
1065
55
Park
,
S.Y.
and
Kim
,
I.S.
(
2017
)
Engulfment signals and the phagocytic machinery for apoptotic cell clearance
.
Exp. Mol. Med.
49
,
e331
56
Lv
,
Z.
,
Bian
,
Z.
,
Shi
,
L.
,
Niu
,
S.
,
Ha
,
B.
,
Tremblay
,
A.
et al. (
2015
)
Loss of cell surface CD47 clustering formation and binding avidity to SIRPα facilitate apoptotic cell clearance by macrophages
.
J. Immunol.
195
,
661
67
57
Oldenborg
,
P.A.
,
Gresham
,
H.D.
and
Lindberg
,
F.P.
(
2001
)
CD47-signal regulatory protein α (SIRPα) regulates Fcγ and complement receptor-mediated phagocytosis
.
J. Exp. Med.
193
,
855
862
58
Brown
,
S.
,
Heinisch
,
I.
,
Ross
,
E.
,
Shaw
,
K.
,
Buckley
,
C.O.
and
Savill
,
J.
(
2002
)
Apoptosis disables CD31-mediated cell detachment from phagocytes promoting binding and engulfment
.
Nature
418
,
200
203
59
Barkal
,
A.A.
,
Brewer
,
R.E.
,
Markovic
,
M.
,
Kowarsky
,
M.
,
Barkal
,
S.A.
,
Zaro
,
B.W.
et al. (
2019
)
CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy
.
Nature
572
,
392
396
60
Feng
,
M.
,
Jiang
,
W.
,
Kim
,
B.Y.S.
,
Zhang
,
C.C.
,
Fu
,
Y.X.
and
Weissman
,
I.L.
(
2019
)
Phagocytosis checkpoints as new targets for cancer immunotherapy
.
Nat. Rev. Cancer
19
,
568
586
61
Sachet
,
M.
,
Liang
,
Y.Y.
and
Oehler
,
R.
(
2017
)
The immune response to secondary necrotic cells
.
Apoptosis
22
,
1189
1204
62
Rogers
,
C.
,
Fernandes-Alnemri
,
T.
,
Mayes
,
L.
,
Alnemri
,
D.
,
Cingolani
,
G.
and
Alnemri
,
E.S.
(
2017
)
Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death
.
Nat. Commun.
8
,
14128
63
Tixeira
,
R.
,
Shi
,
B.
,
Parkes
,
M.A.F.
,
Hodge
,
A.L.
,
Caruso
,
S.
,
Hulett
,
M.D.
et al. (
2018
)
Gasdermin E does Not limit apoptotic cell disassembly by promoting early onset of secondary necrosis in jurkat T cells and THP-1 monocytes
.
Front. Immunol.
9
,
2842
64
Wang
,
Q.
,
Imamura
,
R.
,
Motani
,
K.
,
Kushiyama
,
H.
,
Nagata
,
S.
and
Suda
,
T.
(
2013
)
Pyroptotic cells externalize eat-me and release find-me signals and are efficiently engulfed by macrophages
.
Int. Immunol.
5
,
363
372
65
Wang
,
J.
and
Kubes
,
P.
(
2016
)
A reservoir of mature cavity macrophages that can rapidly invade visceral organs to affect tissue repair
.
Cell
165
,
668
678
66
McDonald
,
B.
,
Pittman
,
K.
,
Menezes
,
G.B.
,
Hirota
,
S.A.
,
Slaba
,
I.
,
Waterhouse
,
C.C.M.
et al. (
2010
)
Intravascular danger signals guide neutrophils to sites of sterile inflammation
.
Science
330
,
362
366
67
Marques
,
P.E.
,
Amaral
,
S.S.
,
Pires
,
D.A.
,
Nogueira
,
L.L.
,
Soriani
,
F.M.
,
Lima
,
B.H.F.
et al. (
2012
)
Chemokines and mitochondrial products activate neutrophils to amplify organ injury during mouse acute liver failure
.
Hepatology
56
,
1971
1982
68
Krysko
,
D.V.
,
Denecker
,
G.
,
Festjens
,
N.
,
Gabriels
,
S.
,
Parthoens
,
E.
,
D'Herde
,
K.
et al. (
2006
)
Macrophages use different internalization mechanisms to clear apoptotic and necrotic cells
.
Cell Death Differ.
13
,
2011
2022
69
Cocco
,
R.E.
and
Ucker
,
D.S.
(
2001
)
Distinct modes of macrophage recognition for apoptotic and necrotic cells are not specified exclusively by phosphatidylserine exposure
.
Mol. Biol. Cell
12
,
777
1188
70
Jiang
,
L.
,
Tixeira
,
R.
,
Caruso
,
S.
,
Atkin-Smith
,
G.K.
,
Baxter
,
A.A.
,
Paone
,
S.
et al. (
2016
)
Monitoring the progression of cell death and the disassembly of dying cells by flow cytometry
.
Nat. Protoc.
11
,
655
663
71
Schwegler
,
M.
,
Wirsing
,
A.M.
,
Dollinger
,
A.J.
,
Abendroth
,
B.
,
Putz
,
F.
,
Fietkau
,
R.
et al. (
2015
)
Clearance of primary necrotic cells by non-professional phagocytes
.
Biol. Cell
107
,
372
387
72
Li
,
Z.
,
Venegas
,
V.
,
Nagaoka
,
Y.
,
Morino
,
E.
,
Raghavan
,
P.
,
Audhya
,
A.
et al. (
2015
)
Necrotic cells actively attract phagocytes through the collaborative action of two distinct PS-exposure mechanisms
.
PLoS Genet.
11
,
e1005285
73
Brouckaert
,
G.
,
Kalai
,
M.
,
Krysko
,
D.V.
,
Saelens
,
X.
,
Vercammen
,
D.
,
Ndlovu
,
M.
et al. (
2004
)
Phagocytosis of necrotic cells by macrophages is phosphatidylserine dependent and does not induce inflammatory cytokine production
.
Mol. Biol. Cell
15
,
1089
1100
74
Budai
,
Z.
,
Ujlaky-Nagy
,
L.
,
Kis
,
G.N.
,
Antal
,
M.
,
Bankó
,
C.
,
Bacsó
,
Z.
et al. (
2019
)
Macrophages engulf apoptotic and primary necrotic thymocytes through similar phosphatidylserine-dependent mechanisms
.
FEBS Open Bio
9
,
446
456
75
Cao
,
L.
,
Chang
,
H.
,
Shi
,
X.
,
Peng
,
C.
and
He
,
Y.
(
2016
)
Keratin mediates the recognition of apoptotic and necrotic cells through dendritic cell receptor DEC205/CD205
.
Proc. Natl Acad. Sci. U.S.A.
113
,
13438
13443
76
Borg
,
R.J.
,
Samson
,
A.L.
,
Au
,
A.E.L.
,
Scholzen
,
A.
,
Fuchsberger
,
M.
,
Kong
,
Y.Y.
et al. (
2015
)
Dendritic cell-mediated phagocytosis but not immune activation is enhanced by plasmin
.
PLoS ONE
10
,
e0131216
77
Poon
,
I.K.H.
,
Hulett
,
M.D.
and
Parish
,
C.R.
(
2010
)
Molecular mechanisms of late apoptotic/necrotic cell clearance
.
Cell Death Differ.
17
,
381
397
78
Gaipl
,
U.S.
,
Kuenkele
,
S.
,
Voll
,
R.E.
,
Beyer
,
T.D.
,
Kolowos
,
W.
,
Heyder
,
P.
et al. (
2001
)
Complement binding is an early feature of necrotic and a rather late event during apoptotic cell death
.
Cell Death Differ.
8
,
327
334
79
Böttcher
,
A.
,
Gaipl
,
U.S.
,
Fürnrohr
,
B.G.
,
Herrmann
,
M.
,
Girkontaite
,
I.
,
Kalden
,
J.R.
et al. (
2006
)
Involvement of phosphatidylserine, αvβ3, CD14, CD36, and complement C1q in the phagocytosis of primary necrotic lymphocytes by macrophages
.
Arthritis Rheum.
54
,
927
938
80
Xu
,
W.
,
Berger
,
S.P.
,
Trouw
,
L.A.
,
de Boer
,
H.C.
,
Schlagwein
,
N.
,
Mutsaers
,
C.
et al. (
2008
)
Properdin binds to late apoptotic and necrotic cells independently of C3b and regulates alternative pathway complement activation
.
J. Immunol.
180
,
7613
7621
81
Liang
,
Y.Y.
,
Arnold
,
T.
,
Michlmayr
,
A.
,
Rainprecht
,
D.
,
Perticevic
,
B.
,
Spittler
,
A.
et al. (
2014
)
Serum-dependent processing of late apoptotic cells for enhanced efferocytosis
.
Cell Death Dis.
5
,
e1264
82
Poon
,
I.K.H.
,
Hulett
,
M.D.
and
Parish
,
C.R.
(
2010
)
Histidine-rich glycoprotein is a novel plasma pattern recognition molecule that recruits IgG to facilitate necrotic cell clearance via FcγRI on phagocytes
.
Blood
115
,
2473
2482
83
Arai
,
S.
,
Kitada
,
K.
,
Yamazaki
,
T.
,
Takai
,
R.
,
Zhang
,
X.
,
Tsugawa
,
Y.
et al. (
2016
)
Apoptosis inhibitor of macrophage protein enhances intraluminal debris clearance and ameliorates acute kidney injury in mice
.
Nat. Med.
22
,
183
193
84
Weinlich
,
R.
,
Oberst
,
A.
,
Beere
,
H.M.
and
Green
,
D.R.
(
2017
)
Necroptosis in development, inflammation and disease
.
Nat. Rev. Mol. Cell Biol.
18
,
127
136
85
Wang
,
Q.
,
Ju
,
X.
,
Zhou
,
Y.
and
Chen
,
K.
(
2015
)
Necroptotic cells release find-me signal and are engulfed without proinflammatory cytokine production
.
Vitr. Cell. Dev. Biol. Anim.
51
,
1033
1039
86
Zargarian
,
S.
,
Shlomovitz
,
I.
,
Erlich
,
Z.
,
Hourizadeh
,
A.
,
Ofir-Birin
,
Y.
,
Croker
,
B.A.
et al. (
2017
)
Phosphatidylserine externalization, “necroptotic bodies” release, and phagocytosis during necroptosis
.
PLoS Biol.
15
,
e2002711
87
Chen
,
J.
,
Kuroki
,
S.
,
Someda
,
M.
and
Yonehara
,
S.
(
2019
)
Interferon-induces the cell surface exposure of phosphatidylserine by activating the protein MLKL in the absence of caspase-8 activity
.
J. Biol. Chem.
294
,
11994
12006
88
Gong
,
Y.N.
,
Guy
,
C.
,
Olauson
,
H.
,
Becker
,
J.U.
,
Yang
,
M.
,
Fitzgerald
,
P.
et al. (
2017
)
ESCRT-III acts downstream of MLKL to regulate necroptotic cell death and its consequences
.
Cell
169
,
286
300.e16
89
Lu
,
J.
,
Shi
,
W.
,
Liang
,
B.
,
Chen
,
C.
,
Wu
,
R.
,
Lin
,
H.
et al. (
2019
)
Efficient engulfment of necroptotic and pyroptotic cells by nonprofessional and professional phagocytes
.
Cell Discov.
5
,
39
90
Gerlach
,
B.D.
,
Marinello
,
M.
,
Heinz
,
J.
,
Rymut
,
N.
,
Sansbury
,
B.E.
,
Riley
,
C.O.
et al. (
2020
)
Resolvin D1 promotes the targeting and clearance of necroptotic cells
.
Cell Death Differ.
27
,
525
539
91
Man
,
S.M.
,
Karki
,
R.
and
Kanneganti
,
T.D.
(
2017
)
Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases
.
Immunol. Rev.
277
,
61
75
92
Tan
,
M.S.
,
Tan
,
L.
,
Jiang
,
T.
,
Zhu
,
X.C.
,
Wang
,
H.F.
,
Jia
,
C.D.
et al. (
2014
)
Amyloid-β induces NLRP1-dependent neuronal pyroptosis in models of Alzheimer's disease
.
Cell Death Dis.
5
,
e1382
93
Han
,
C.
,
Yang
,
Y.
,
Guan
,
Q.
,
Zhang
,
X.
,
Shen
,
H.
,
Sheng
,
Y.
et al. (
2020
)
New mechanism of nerve injury in Alzheimer's disease: β-amyloid-induced neuronal pyroptosis
.
J. Cell. Mol. Med.
24
,
8078
8090
94
Milner
,
M.T.
,
Maddugoda
,
M.
,
Götz
,
J.
,
Burgener
,
S.S.
and
Schroder
,
K.
(
2021
)
The NLRP3 inflammasome triggers sterile neuroinflammation and Alzheimer's disease
.
Curr. Opin. Immunol.
68
,
116
124
95
Wree
,
A.
,
Eguchi
,
A.
,
Mcgeough
,
M.D.
,
Pena
,
C.A.
,
Johnson
,
C.D.
,
Canbay
,
A.
et al. (
2014
)
NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice
.
Hepatology
59
,
898
910
96
Gaul
,
S.
,
Leszczynska
,
A.
,
Alegre
,
F.
,
Kaufmann
,
B.
,
Johnson
,
C.D.
,
Adams
,
L.A.
et al. (
2021
)
Hepatocyte pyroptosis and release of inflammasome particles induce stellate cell activation and liver fibrosis
.
J. Hepatol.
74
,
156
167
97
Bierschenk
,
D.
,
Monteleone
,
M.
,
Moghaddas
,
F.
,
Baker
,
P.J.
,
Masters
,
S.L.
,
Boucher
,
D.
et al. (
2019
)
The Salmonella pathogenicity island-2 subverts human NLRP3 and NLRC4 inflammasome responses
.
J. Leukoc. Biol.
105
,
401
410
98
Qu
,
Y.
,
Misaghi
,
S.
,
Newton
,
K.
,
Maltzman
,
A.
,
Izrael-Tomasevic
,
A.
,
Arnott
,
D.
et al. (
2016
)
NLRP3 recruitment by NLRC4 during Salmonella infection
.
J. Exp. Med.
213
,
877
885
99
Jorgensen
,
I.
,
Zhang
,
Y.
,
Krantz
,
B.A.
and
Miao
,
E.A.
(
2016
)
Pyroptosis triggers pore-induced intracellular traps (PITs) that capture bacteria and lead to their clearance by efferocytosis
.
J. Exp. Med.
213
,
2113
2128
100
Yang
,
D.
,
He
,
Y.
,
Muñoz-Planillo
,
R.
,
Liu
,
Q.
and
Núñez
,
G.
(
2015
)
Caspase-11 requires the pannexin-1 channel and the purinergic P2X7 pore to mediate pyroptosis and endotoxic shock
.
Immunity
43
,
923
932
101
Pelegrin
,
P.
and
Surprenant
,
A.
(
2006
)
Pannexin-1 mediates large pore formation and interleukin-1β release by the ATP-gated P2X7 receptor
.
EMBO J.
25
,
5071
5082
102
Jorgensen
,
I.
,
Lopez
,
J.P.
,
Laufer
,
S.A.
and
Miao
,
E.A.
(
2016
)
IL-1β, IL-18, and eicosanoids promote neutrophil recruitment to pore-induced intracellular traps following pyroptosis
.
Eur. J. Immunol.
46
,
2761
2766
103
Heilig
,
R.
,
Dick
,
M.S.
,
Sborgi
,
L.
,
Meunier
,
E.
,
Hiller
,
S.
and
Broz
,
P.
(
2018
)
The Gasdermin-D pore acts as a conduit for IL-1β secretion in mice
.
Eur. J. Immunol.
48
,
584
592
104
Fujii
,
T.
,
Sakata
,
A.
,
Nishimura
,
S.
,
Eto
,
K.
and
Nagata
,
S.
(
2015
)
TMEM16F is required for phosphatidylserine exposure and microparticle release in activated mouse platelets
.
Proc. Natl Acad. Sci. U.S.A.
112
,
12800
12805
105
Suzuki
,
J.
,
Fujii
,
T.
,
Imao
,
T.
,
Ishihara
,
K.
,
Kuba
,
H.
and
Nagata
,
S.
(
2013
)
Calcium-dependent phospholipid scramblase activity of TMEM 16 protein family members
.
J. Biol. Chem.
288
,
13305
13316
106
Klöditz
,
K.
and
Fadeel
,
B.
(
2019
)
Three cell deaths and a funeral: macrophage clearance of cells undergoing distinct modes of cell death
.
Cell Death Discov.
5
,
65
107
Baxter
,
A.A.
,
Poon
,
I.K.H.
and
Hulett
,
M.D.
(
2017
)
The plant defensin NaD1 induces tumor cell death via a non-apoptotic, membranolytic process
.
Cell Death Discov.
3
,
16102
108
Voll
,
R.E.
,
Herrmann
,
M.
,
Roth
,
E.A.
,
Stach
,
C.
,
Kalden
,
J.R.
and
Girkontaite
,
I.
(
1997
)
Immunosuppressive effects of apoptotic cells [9]
.
Nature
390
,
350
351
109
Chen
,
W.J.
,
Frank
,
M.E.
,
Jin
,
W.
and
Wahl
,
S.M.
(
2001
)
TGF-β released by apoptotic T cells contributes to an immunosuppressive milieu
.
Immunity
14
,
715
725
110
Brissette
,
M.J.
,
Lepage
,
S.
,
Lamonde
,
A.S.
,
Sirois
,
I.
,
Groleau
,
J.
,
Laurin
,
L.P.
et al. (
2012
)
MFG-E8 released by apoptotic endothelial cells triggers anti-inflammatory macrophage reprogramming
.
PLoS ONE
7
,
e36368
111
Luo
,
B.
,
Gan
,
W.
,
Liu
,
Z.
,
Shen
,
Z.
,
Wang
,
J.
,
Shi
,
R.
et al. (
2016
)
Erythropoeitin signaling in macrophages promotes dying cell clearance and immune tolerance
.
Immunity
44
,
287
302
112
Huynh
,
M.L.N.
,
Fadok
,
V.A.
and
Henson
,
P.M.
(
2002
)
Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-β1 secretion and the resolution of inflammation
.
J. Clin. Invest.
109
,
41
50
113
Fadok
,
V.A.
,
Bratton
,
D.L.
,
Konowal
,
A.
,
Freed
,
P.W.
,
Westcott
,
J.Y.
and
Henson
,
P.M.
(
1998
)
Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-β, PGE2, and PAF
.
J. Clin. Invest.
101
,
890
898
114
Juncadella
,
I.J.
,
Kadl
,
A.
,
Sharma
,
A.K.
,
Shim
,
Y.M.
,
Hochreiter-Hufford
,
A.
,
Borish
,
L.
et al. (
2013
)
Apoptotic cell clearance by bronchial epithelial cells critically influences airway inflammation
.
Nature
493
,
547
551
115
Golpon
,
H.A.
,
Fadok
,
V.A.
,
Taraseviciene-Stewart
,
L.
,
Scerbavicius
,
R.
,
Sauer
,
C.
,
Welte
,
T.
et al. (
2004
)
Life after corpse engulfment: phagocytosis of apoptotic cells leads to VEGF secretion and cell growth
.
FASEB J.
18
,
1716
1718
116
Notley
,
C.A.
,
Brown
,
M.A.
,
Wright
,
G.P.
and
Ehrenstein
,
M.R.
(
2011
)
Natural IgM is required for suppression of inflammatory arthritis by apoptotic cells
.
J. Immunol.
186
,
4967
4972
117
Van Lent
,
P.L.E.M.
,
Licht
,
R.
,
Dijkman
,
H.
,
Holthuysen
,
A.E.M.
,
Berden
,
J.H.M.
and
Van Den Berg
,
W.B.
(
2001
)
Uptake of apoptotic leukocytes by synovial lining macrophages inhibits immune complex-mediated arthritis
.
J. Leukoc. Biol.
70
,
708
14
118
Cohen
,
I.
,
Rider
,
P.
,
Carmi
,
Y.
,
Braiman
,
A.
,
Dotan
,
S.
,
White
,
M.R.
et al. (
2010
)
Differential release of chromatin-bound IL-1α discriminates between necrotic and apoptotic cell death by the ability to induce sterile inflammation
.
Proc. Natl Acad. Sci. U.S.A.
107
,
2574
2579
119
Dankers
,
W.
,
Hasnat
,
M.A.
,
Swann
,
V.
,
Alharbi
,
A.
,
Lee
,
J.P.W.
,
Cristofaro
,
M.A.
et al. (
2020
)
Necrotic cell death increases the release of macrophage migration inhibitory factor by monocytes/macrophages
.
Immunol. Cell Biol.
98
,
782
790
120
Scaffidi
,
P.
,
Misteli
,
T.
and
Bianchi
,
M.E.
(
2002
)
Release of chromatin protein HMGB1 by necrotic cells triggers inflammation
.
Nature
418
,
191
195
121
Beyer
,
C.
,
Stearns
,
N.A.
,
Giessl
,
A.
,
Distler
,
J.H.W.
,
Schett
,
G.
and
Pisetsky
,
D.S.
(
2012
)
The extracellular release of DNA and HMGB1 from Jurkat T cells during in vitro necrotic cell death
.
Innate Immun.
18
,
727
737
122
Basu
,
S.
,
Binder
,
R.J.
,
Suto
,
R.
,
Anderson
,
K.M.
and
Srivastava
,
P.K.
(
2000
)
Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-κB pathway
.
Int. Immunol.
12
,
1539
1546
123
El Mezayen
,
R.
,
El Gazzar
,
M.
,
Seeds
,
M.C.
,
McCall
,
C.E.
,
Dreskin
,
S.C.
and
and Nicolls
,
M.R.
(
2007
)
Endogenous signals released from necrotic cells augment inflammatory responses to bacterial endotoxin
.
Immunol. Lett.
111
,
36
44
124
Su
,
L.
,
Li
,
N.
,
Tang
,
H.
,
Lou
,
Z.
,
Chong
,
X.
,
Zhang
,
C.
et al. (
2018
)
Kupffer cell-derived TNF-α promotes hepatocytes to produce CXCL1 and mobilize neutrophils in response to necrotic cells
.
Cell Death Dis.
9
,
323
125
Christofferson
,
D.E.
and
Yuan
,
J.
(
2010
)
Cyclophilin A release as a biomarker of necrotic cell death
.
Cell Death Differ.
17
,
1942
1943
126
Zhu
,
K.
,
Liang
,
W.
,
Ma
,
Z.
,
Xu
,
D.
,
Cao
,
S.
,
Lu
,
X.
et al. (
2018
)
Necroptosis promotes cell-autonomous activation of proinflammatory cytokine gene expression article
.
Cell Death Dis.
9
,
500
127
Duprez
,
L.
,
Takahashi
,
N.
,
Van Hauwermeiren
,
F.
,
Vandendriessche
,
B.
,
Goossens
,
V.
,
Vanden Berghe
,
T.
et al. (
2011
)
RIP kinase-dependent necrosis drives lethal systemic inflammatory response syndrome
.
Immunity
35
,
908
918
128
Wu
,
X.Y.
,
Li
,
K.T.
,
Yang
,
H.X.
,
Yang
,
B.
,
Lu
,
X.
,
Zhao
,
L.D.
et al. (
2020
)
Complement C1q synergizes with PTX3 in promoting NLRP3 inflammasome over-activation and pyroptosis in rheumatoid arthritis
.
J. Autoimmun.
106
,
102336
129
Franklin
,
B.S.
,
Bossaller
,
L.
,
De Nardo
,
D.
,
Ratter
,
J.M.
,
Stutz
,
A.
,
Engels
,
G.
et al. (
2014
)
The adaptor ASC has extracellular and “prionoid” activities that propagate inflammation
.
Nat. Immunol.
15
,
727
737
130
Atkin-Smith
,
G.K.
,
Duan
,
M.
,
Chen
,
W.
and
Poon
,
I.K.H.
(
2018
)
The induction and consequences of Influenza A virus-induced cell death
.
Cell Death Dis.
9
,
1002
131
Grégoire
,
M.
,
Uhel
,
F.
,
Lesouhaitier
,
M.
,
Gacouin
,
A.
,
Guirriec
,
M.
,
Mourcin
,
F.
et al. (
2018
)
Impaired efferocytosis and neutrophil extracellular trap clearance by macrophages in ARDS
.
Eur. Respir. J.
52
,
1702590
132
Trahtemberg
,
U.
,
Grau
,
A.
,
Tabib
,
A.
,
Atallah
,
M.
,
Krispin
,
A.
and
Mevorach
,
D.
(
2016
)
Identification and characterization of two human monocyte-derived dendritic cell subpopulations with different functions in dying cell clearance and different patterns of cell death
.
PLoS ONE
11
,
e0162984
133
Kojima
,
Y.
,
Volkmer
,
J.P.
,
McKenna
,
K.
,
Civelek
,
M.
,
Lusis
,
A.J.
,
Miller
,
C.L.
et al. (
2016
)
CD47-blocking antibodies restore phagocytosis and prevent atherosclerosis
.
Nature
536
,
86
90
134
Lee
,
C.S.
,
Penberthy
,
K.K.
,
Wheeler
,
K.M.
,
Juncadella
,
I.J.
,
Vandenabeele
,
P.
,
Lysiak
,
J.J.
et al. (
2016
)
Boosting apoptotic cell clearance by colonic epithelial cells attenuates inflammation in vivo
.
Immunity
44
,
807
820
135
Atkin-Smith
,
G.K.
,
Tixeira
,
R.
,
Paone
,
S.
,
Mathivanan
,
S.
,
Collins
,
C.
,
Liem
,
M.
et al. (
2015
)
A novel mechanism of generating extracellular vesicles during apoptosis via a beads-on-a-string membrane structure
.
Nat. Commun.
6
,
7439
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY-NC-ND). Open access for this article was enabled by the participation of Walter and Eliza Hall Institute in an all-inclusive Read & Publish pilot with Portland Press and the Biochemical Society under a transformative agreement with CAUL.