Вы находитесь на странице: 1из 12

pubs.acs.

org/accounts Article

Dissecting Programmed Cell Death with Small Molecules


Yingjie Bai, Hiu C. Lam, and Xiaoguang Lei*

Cite This: https://dx.doi.org/10.1021/acs.accounts.9b00600 Read Online

ACCESS Metrics & More Article Recommendations

CONSPECTUS: Programmed cell death (PCD) is fundamentally an


See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

indispensable process in all cellular activities, including cell develop-


ment, wound healing, and immune surveillance of tumors (Galluzzi, L.
et al. Cell Death Differ. 2018, 25, 486−541). Malfunctioning of PCD has
Downloaded via UNIV OF BIRMINGHAM on April 16, 2020 at 22:41:02 (UTC).

been shown to be closely related to human diseases such as acute


pancreatitis, neurodegenerative diseases, and diverse types of cancers.
To date, multiple PCD processes have been discovered and the
corresponding regulatory pathways have been elucidated. For example,
apoptosis and autophagy are two PCD mechanisms that have been well
studied by sophisticated models and probe toolkits. However, limited
genetic and chemical tools for other types of PCD hamper the
elucidation of their molecular mechanisms. Our group has been
studying PCD using both function-oriented synthesis and chemical
biology strategies, including the development of diverse chemical
probes based on novel PCD modulators. For instance, in the
development of downstream programmed necrosis (or necroptosis)
inhibitor necrosulfonamide, we used a chemical probe to unveil a functional protein that was not previously implicated in
necroptosis, mixed lineage kinase domain-like protein (MLKL). In addition, high throughput screening and medicinal chemistry
enabled the discovery of bioymifi, a small molecule agonist which selectively causes oligomerization of the death receptor 5 (DR5),
to induce extrinsic apoptosis. Furthermore, we developed a biomimetic synthetic strategy based on diverse Diels−Alder reactions in
the total syntheses of ainsliadimers A and B, ainsliatrimers A and B, and gonchnatiolides A−C, which are natural product inhibitors
or activators for PCD. Using synthetic ainsliadimer A probe, we elucidated that ainsliadimer A inhibits the NF-κB pathway by
covalently binding to Cys46 of IKKβ and triggers apoptosis of cancer cells. We have also revealed that IKKβ is allosterically inhibited
by ainsliadimer A. In addition to total synthesis, we have developed a bioorthogonal click hetero-Diels−Alder cycloaddition of vinyl
thioether and o-quinolinone quinone methide (TQ-ligation) to facilitate small molecule target identification. The combination of
total synthesis and TQ-ligation enables subcellular imaging and identification of the cellular target of ainsliatrimer A to be PPARγ. In
addition, TQ-ligation has been applied in the discovery of heat shock protein 90 (HSP90) as one of the functional target proteins for
kongensin A. We also confirmed that kongensin A covalently attaches to Cys420 within HSP90 and demonstrated that kongensin A
blocks the interaction between HSP90 and CDC37 and subsequently inhibits necroptosis. Our development of these diverse PCD
modulators provides not only effective chemical tools for fundamental biomedical research, but also the foundation for drug
discovery targeting important human diseases such as cancers and inflammation caused by malfunction of PCD.

■ INTRODUCTION
Programmed cell death (PCD) can be classified into apoptosis
parthanatos, entotic cell death, and mitotic catastrophe as RCD
pathways.9 Since the characterization of novel PCD-related
and nonapoptotic PCDs based on the nature of ubiquitous signaling pathways has not yet been completed, space remains
causes and outcome of cell death. Apoptosis, the prototype
for component characterizations and development of small
PCD, was first described by Kerr et al. in 1972,1 characterized
by its morphological changes, unique biochemistry, and lack of molecule modulators for PCDs.
inflammation.1−4 Besides apoptosis, typical nonapoptotic
PCDs including autophagy,5 necroptosis,6 ferroptosis,7 and Received: November 29, 2019
pyroptosis,8 have been discovered recently.9,10 In addition to
PCDs, regulated cell death (RCD) has emerged to extend the
definition of PCDs to include the promiscuous cell death
pathways.9 In 2018, the Nomenclature Committee on Cell
Death accepted MPT-driven necrosis, NETotic cell deaths,11

© XXXX American Chemical Society https://dx.doi.org/10.1021/acs.accounts.9b00600


A Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

Figure 1. Small molecules modulating programmed cell death pathways including necroptosis, apoptosis, and the NF-κB pathway.

Indeed, recent discoveries of PCDs (e.g., ferroptosis9 and therapeutics for PCD-related human diseases such as cancers
oncosis12) were initiated from their corresponding inhibitors and autoimmune diseases.
or PCD pathway components via chemical screenings using
cellular phenotypes as readouts. However, the target
identification of these small molecule inhibitors posed a
■ DISSECTING NECROPTOSIS WITH
NECROSULFONAMIDE
challenge. Bioorthogonal reactions have been broadly applied Necroptosis was first introduced by Yuan and co-workers in
in bioimaging and target identifications.13,14 This methodology 2006, along with an antinecroptosis compound named as
might be applied in the study of PCDs with the corresponding necrostatin-1 (Nec-1) which targets receptor-interacting
modulators by attaching chemical handles for various purposes protein 1 (RIPK1).6 Subsequently, multiple antinecroptosis
(e.g., attachment of biotin for affinity purification or compounds targeting RIPK1 or RIPK3 have been identified.21
fluorescence for visualization). The rapid reaction time, high It has been shown that RIPK3 can phosphorylate itself and its
spatiotemporal resolution, unique reactivity, and high specific- partner RIPK1, within the necrosome, an alternative name for
ity features of these chemical biological tools are comple- the RIPK1/RIPK3 complex.21 However, the biological
mentary to conventional studies in biochemistry and genetics. consequences of the phosphorylation related to necroptosis
Therefore, despite the fact that the reagents might be and the necroptotic downstream substrate(s) of RIPK1/
somewhat toxic,15 bioorthogonal reactions have been currently RIPK3 complex remained elusive. Therefore, we initiated a
applied in many research areas including microbiology, program in further deciphering the molecular pathway of
oncology, developmental biology, and neurobiology.14,16−19 necroptosis. We used a cell-based assay of necroptosis for high-
Herein, we summarize our efforts in revealing the throughput screening to discover antinecroptosis small
mechanisms of PCDs using chemical biology approaches molecules. Two previously established cell lines, Jurkat and
with various small molecule probes and bioorthogonal HT-29 cells, were each treated with a combination of TNF-α
to initiate programmed cell death, a cell-permeable small
reactions which were developed in our laboratory (Figure 1).
molecule that mimics Smac function (Smac mimetic) to
First, we will discuss our chemical biology studies which led to
enhance the cell death signal, and the pan-caspase inhibitor z-
the discovery of PCD modulators necrosulfonamide and VAD-fmk to block apoptosis and initiate necroposis.22,23 After
bioymifi. Next, we present our endeavors using natural product screening ∼200 000 compounds using this cell-based necrop-
total synthesis and subsequent target identification of PCD- tosis assay, we identified a sulfonylaminopyrimidine hit as a
inducers ainsliadimer A and ainsliatrimer A. Moreover, we necroptosis inhibitor with an IC50 of less than 1 μM. Shortly
introduce the combination of biomimetic synthesis and TQ- after structure−activity relationship (SAR) study, we discov-
ligation that facilitates the target identifications of these PCD ered a more potent derivative, with an IC50 of 124 nM, which
modulators. Ultimately, the research summarized in this we named as necrosulfonamide. Subsequent cell-based bio-
Account alongside other studies of PCDs from different logical evaluations of how necrosulfonamide inhibits necrop-
research groups5,7,20 has collectively contributed to stimulate tosis were performed, and the results showed that the
fundamental biological discoveries and develop potential formation of RIPK3 punctae in the necrosulfonamide treated
B https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

Figure 2. Necrosulfonamide targets MLKL for antinecroptosis activity. (A) Live cell imaging upon the treatment of necrosulfonamide (NSA) and
necrostatin-1 (Nec-1). Expressing the mCherry-RIPK3 fusion protein in HeLa cells rendered them necrosis responsive. T, TNF-α; S, Smac
mimetic; Z, z-VAD-fmk. (B) Pull-down of RIPK3 in necrosulfonamide treated cells revealed the interacting protein MLKL. (C) Structure of the
chemical probe for target identification. (D) Graphical abstract of the mode of action on how necrosulfonamide blocks necroptosis.

cells was significantly different from the one of Nec-1 treated After the functional study of MLKL, we then proposed a
cells. These results suggested that the cellular target(s) and hypothesis that MLKL might be the direct cellular target of
mode of action for necrosulfonamide might be different from necrosulfonamide. Therefore, we designed and synthesized a
Nec-1 (Figure 2A).24 necrosulfonamide probe for the pull-down assay. From our
We then conducted co-immunoprecipitation experiments SAR studies, we have gained crucial information about the
with necrosulfonamide to examine the possible RIPK3- heterocyclic moiety of necrosulfonamide could tolerate
interacting proteins. Indeed, pull down experiments and modifications without compromising its biological activity.
subsequent mass spectrometry analysis revealed that the Then a small molecule probe was prepared which contained a
mixed lineage kinase-domain like protein (MLKL) is an biotin tag, and a rigid polyproline linker between biotin and
necrosulfonamide to enhance the binding activity of the probe
interacting partner of RIPK3 (Figure 2B). MLKL was later
to the potential target proteins (Figure 2C). Upon the
confirmed to be a crucial new component of necrosome, the
treatment of RIP3-overexpressing HeLa cell lysates with this
RIPK1/RIPK3-containing complex that promotes necroptosis. probe, a subsequent pull-down assay by streptavidin yielded
The inhibition of necroptosis mediated by siRNA knockdown endogenous MLKL, which could be competed off by the
of MLKL further demonstrated that MLKL is indispensable for addition of excess necrosulfonamide as a binding competitor.
the necroptosis pathway. Therefore, our search for RIPK3 Mutagenesis experiments also revealed that the C86S mutant
downstream inhibitors and the subsequent discovery of of MLKL still mediated necroptosis despite the addition of
necrosulfonamide have revealed that MLKL as one of the necrosulfonamide, suggesting that the Cys86 residue of MLKL
central and essential components in the necroptosis process is essential for the binding of necrosulfonamide. Moreover,
(Figure 2D).24 previous SAR studies of necrosulfonamide suggested that the
C https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

Figure 3. Bioymifi is a selective death receptor 5 (DR5) agonist to induce extrinsic apoptosis. (A) Structures of lead compounds A1, A2, C2, and
bioymifi. (B) RNA inferencing indicated that A2C2 (a 1:9 mixture of compounds A2 and C2) and bioymifi selectively target DR5. Error bars
represent the SD of experimental duplicates. (C) Bioymifi induced FLAG-labeled DR5 aggregation (red color). (D) Graphical abstract of the
mechanism on how bioymifi induces extrinsic apoptosis of cancer cells by selectively activating DR5. B, bioymifi; DISC, death-inducing signaling
complex; FADD, Fas-associated with death domain protein; TRADD, TNF receptor-associated death domain.

α,β-unsaturated amide group might act as a Michael acceptor was not induced when cells were treated with either A2 or C2
that covalently attaches to MLKL. Collectively, these data alone. Our attention then turned to de novo syntheses of many
revealed that MLKL plays a significant role in initiating other analogues to further explore the SAR. Ultimately, we
necroptosis and necrosulfonamide is a potent and selective developed a bromine-substituted thiazolidin-4-one derivative,
small molecule inhibitor of necroptosis by covalently targeting namely, bioymifi, which showed an improved activity with an
MLKL.24 EC50 at 2 μM for inducing apoptosis (Figure 3A).27

■ DISCOVERY OF THE FIRST DR5 AGONIST


BIOYMIFI AS A POTENTIAL ANTICANCER AGENT
We then determined which apoptotic pathway was targeted
by bioymifi.24 Our initial study showed that the pan-caspase
inhibitor z-VAD-FMK rescued bioymifi-treated cells; therefore,
Because selective activation of specific apoptotic pathway has we proposed that a caspase-dependent pathway would be
been proven to be an effective anticancer strategy, we were involved in apoptosis. We excluded the intrinsic apoptotic
interested in searching for a compound to activate extrinsic pathway, as RNAi targeting caspase-9 did not improve cell
apoptosis, which is an apoptotic pathway initiated by death survival after treatment with bioymifi. Moreover, biochemical
receptors. 25,26 By screening over 200 000 compounds studies showed that caspase-3, caspase-8, and PARP are
combined with Smac mimetic in T98G cells, we discovered cleaved consecutively after the treatment of bioymifi,
an apoptosis-stimulating hit called A1 (Figure 3A). However, suggesting bioymifi kills cancer cells via the extrinsic apoptotic
our synthetic A1 was unfortunately not biologically active. pathway. We then hypothesized the target of bioymifi could be
After an extensive investigation, we revealed that the structure a death receptor. After screening all known members of death
of “A1” compound from the commercial chemical library was receptors by RNAi, we found that death receptor 5 (DR5) is a
wrongly assigned. The real composition of “A1” was a 1:9 possible cellular target of bioymifi (Figure 3B). Further
mixture of A2 and C2 (Figure 3A). To our surprise, apoptosis isothermal titration calorimetry (ITC) experiments illustrated
D https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

Figure 4. Collective and biomimetic syntheses of sesquiterpene lactone oligomers.

that bioymifi reversibly binds to the ECD domain of DR5 with type sesquiterpenoid dehydrozaluzanin C. Starting from the
a Kd of 1.2 μM similar to its LC50 to cancer cells. From our commercially available santonin, dehydrozaluzanin C was
studies, DR5 was found to be a functional cellular target of synthesized in 12 steps. After screening conditions for the
bioymifi to inducing extrinsic apoptosis.27 hetero-Diels−Alder dimerization of dehydrozaluzanin C,
Since death receptors have been shown to oligomerize to BINOL was found to be the optimal catalyst in promoting
activate the downstream apoptosis executors by the activation the biomimetic dimerization. Hydrolysis of the alkenyl ether of
of TNFα,28 we wondered if bioymifi triggers a similar the dimeric product, followed by cyclization via an intra-
apoptosis-inducing mechanism. Therefore, full-length Flag- molecular aldol reaction furnished the total synthesis of
tagged DR5 from cells was treated with bioymifi, assayed with ainsliadimer A.33
semi-denaturing detergent agarose gel electrophoresis (SDD- We then extended the synthesis and demonstrated that the
AGE), and followed by immunoblot analysis. The high similar biomimetic strategy could be applied for the syntheses
molecule weight of DR5-Flag polymers indicated the of other sesquiterpenoid oligomers, including ainsliatrimers A
aggregation of DR5 was induced by bioymifi or A2C2. In and B, gochnatiolides A−C, and ainsliadimer B (Figure 4).34,35
addition, aggregation of the purified DR5-ECD was observed The collective total syntheses of gochnatiolides A−C and
after the treatment with bioymifi or A2C2, which concluded ainsliadimer B were achieved in two steps and three steps,
that ECD is required for aggregation. Subsequent immuno- respectively, from dehydrozaluzanin C used as the common
fluorescence studies on DR5-overexpressed HCC115 tumor precursor. The biomimetic Diels−Alder reaction was applied
cells treated with bioymifi supported that bioymifi induces as a key step under our optimized conditions to construct the
oligomerization of DR5 on the cellular level (Figure 3C). In polycyclic ring systems. The sesquiterpenoid trimers, ainslia-
conclusion, we have developed bioymifi as the first small trimers A and B, were constructed in four steps via two
molecule agonist for DR5 to trigger extrinsic apoptosis of sequential biomimetic Diels−Alder reactions from dehydroza-
cancer cells (Figure 3D).27 luzanin C. The first enantioselective syntheses also allowed the


unambiguous confirmation of the chemical structure and
COLLECTIVE AND BIOMIMETIC SYNTHESES OF absolute and relative configurations of these sesquiterpenoid
SESQUITERPENOID NATURAL PRODUCTS oligomers such as gochnatiolide B and ainsliatrimers A and B.
More importantly, this biomimetic strategy has accelerated the
A family of sesquiterpenoid oligomer natural products were development of effective small molecule probes for further
isolated from Gochnatia and Ainslia plants. They have shown target identification of ainsliadimer A and ainsliatrimer A.33−36


potent biological activity to induce apoptosis of various tumor
cells, but the modes of action were unknown (Figure 4).29−32
We aimed to develop an efficient approach to access these MODE OF ACTION STUDY FOR AINSLIADIMER A
complex natural products to facilitate the subsequent mode of The biomimetic synthesis of ainsliadimer A has laid the
action studies. Retrosynthetic analysis suggested that ainslia- foundation for our mechanistic study of the apoptosis-inducing
dimer A might be derived from two molecules of guaianolide- activity of this natural product.33 First, we investigated which
E https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

Figure 5. Ainsliadimer A covalently targets IKKβ to activate the NF-κB pathway and trigger apoptosis. (A) Structures of ainsliadimer A derived
positive probe and negative probe. (B) Pull-down assays by ainsliadimer A probes indicated the binding of IKKβ and IKKα. (C) Site-directed
mutagenesis revealed that Cys46 is the binding site. (D) Graphical abstract of the mode of action for ainsliadimer A.

apoptotic pathways were triggered by ainsliadimer A. We A inhibits the phosphorylation of IκB to block the NF-κB
found that the pan-caspase inhibitor z-VAD-FMK can rescue pathway, which shares a comparable mechanism to the known
BCG-823 cells treated with ainsliadimer A, indicating the IκB kinase (IKK) inhibitor BMS-345541 in both LPS and
apoptosis pathway is involved. In addition, through real-time poly(I:C) induced models, suggesting ainsliadimer A might
PCR analysis, we found that multiple apoptosis inhibitory directly block the NF-κB signaling pathway.37
proteins including Bcl-xl, c-IAP, and FLIP were globally We further designed and synthesized an ainsliadimer A-
downregulated, suggesting the regulatory proteins correspond- derived chemical probe based on our SAR studies. This probe
ing to their transcription factor pathway could be targeted.37 contains an ainsliadimer A warhead, a PEG linker, and a biotin
Subsequent investigation of the components in the nuclear tag. Given the fact that saturation of the α,β unsaturated
factor-κB (NF-κB) signaling pathway showed that ainsliadimer lactones led to the loss of function for ainsliadimer A, we also
F https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

constructed a negative control (NC) probe through selective A and kongensin A (Figure 6C).41,42 We will discuss these
reduction of the two conjugated alkene moieties for pull-down endeavors in details in the following sections.


assay (Figure 5A). We then used these two probes in the pull-
down experiments with cell lysates which were previously AINSLIATRIMER A INDUCES APOPTOSIS BY
activated by TNFα treatments. Two specific protein bands, ACTIVATING PPARγ
IKKα and IKKβ, were identified by mass spectrometry analysis
(Figure 5B). To reveal the binding site of ainsliadimer A (1), In the mode of action study of the apoptosis inducer
we individually mutated the nine conserved cysteine residues ainsliatrimer A,41 we first synthesized a bioactive natural
of IKKβ into alanine residues (Figure 5C). Among these product probe in which a vinyl thioether was attached onto the
mutants, C46A was the sole mutant that lost the interaction 3″-O position of ainsliatrimer A (TV-ainsliatrimer A) (Figure
with ainsliadimer A (1). Further LC-MS/MS analysis also 7A). The HeLa cells were treated with TV-ainsliatrimer A,
confirmed that Cys46 residue of IKKβ is covalently attached followed by incubation with fluorescein-modified oQQM
by ainsliadimer A.37 precursor which would undergo “click” hetero-Diels−Alder
Taken together, we have elucidated the detailed mode of reaction with the vinyl thioether of TV-ainsliatrimer A,
action for ainsliadimer A. This remarkable natural product resulting a distribution of the fluorescent small molecule
selectively and covalently targets IKKβ to activate the NF-κB probes in cells. Pretarget imaging showed that fluorescence was
pathway and trigger apoptosis of cancer cells.37 visualized in the nucleus exclusively, suggesting the cellular


targets of ainsliatrimer A should be located in the nucleus
TQ-LIGATION FACILITATES NATURAL PRODUCT (Figure 7A, B). Next, we investigated which nuclear protein(s)
TARGET IDENTIFICATION might bind to ainsliatrimer A. We therefore synthesized the
biotinylated ainsliatrimer A probe using the TQ-ligation
In order to facilitate natural product target identification, we strategy as well as the negative control probe from the
have developed an efficient and robust bioorthogonal ligation saturated TV-ainsliatrimer A derivative. Treatment of HeLa
(TQ-ligation) by “click” hetero-Diels−Alder cycloaddition of cell lysates with these two probes, followed by pull-down assay
o-quinolinone quinone methide and vinyl thioether under using the extracted cell nuclear proteins and LC-MS/MS
physiological conditions (Figure 6A).38 The compatibility of analysis, revealed that histone deacetylase 2 (HDAC2) and
peroxisome proliferator activated receptor γ (PPARγ) as two
candidate cellular targets (Figure 7D). siRNA knockdown at
the protein level demonstrated that PPARγ knockdown could
rescue cells treated with ainsliatrimer A while HDAC2
knockdown could not. Further validation experiments
elucidated that ainsliatrimer A is a selective PPARγ agonist
to trigger apoptosis.41 Along with our discovery, other
independent studies also demonstrated that the activation of
PPARγ could induce apoptosis.43,44 Therefore, the PPARγ
agonists such as ainsliatrimer A could serve as a promising lead
for anticancer therapy.

■ KONGENSIN A IS AN EFFECTIVE INHIBITOR OF


NECROPTOSIS BY TARGETING HSP90
To continue to search for novel small molecules modulating
programmed cell death such as necroptosis, we screened our
in-house ∼300 000 small molecule library and discovered
kongensin A (KA), a diterpenoid natural product isolated from
Croton kongensis as a hit to block necroptosis.45 Initial
validation assays showed that KA blocks necroptosis efficiently
at 2 μM. To identify the cellular target of kongensin A, we first
Figure 6. New bioorthogonal ligation (TQ-ligation). (A) First prepared a positive probe by attaching vinyl thioether onto KA,
generation TQ-ligation. (B) Second generation TQ-ligation. (C) and a negative control probe from KA derivative (Figure 8A).
Target identification of natural products using TQ-ligation. HT-29 cells were incubated with the KA probe and NC probe,
followed by the treatment of biotin-modified oQQM precursor
TQ-ligation has been demonstrated in the imaging of taxol for TQ-ligation. Pull-down assay followed by LC-MS/MS
derivative in live cells.38 The second generation TQ-ligation analysis revealed that one of the cellular targets for KA is heat
was subsequently developed to improve the kinetic rate up to shock protein 90 (HSP90). In addition, Cys420 residue was
18-fold compared to the prototype (Figure 6B).39 Moreover, confirmed to be the binding site by LC-MS/MS analysis and
the second generation TQ ligation is mutually orthogonal to site-directed mutagenesis (Figure 8B−D).42
the traditional strain-promoted azide−alkyne cyclization Moreover, coimmunoprecipitation of HSP90 in kongensin
(SPAAC) both in vitro and in vivo.39,40 We also found that A-treated HeLa lysates revealed that KA inhibits the protein−
the second generation TQ ligation has a comparable rate protein interaction between HSP90 and its co-chaperone cell
constant to that of the widely used SPAAC (k2 = 7.6 × 10−2 division cycle protein 37 (CDC37), a protein which can
M−1 s−1 in CH3CN).39 We further demonstrated that the TQ- stabilize multiple kinases such as CDK4.46 In addition, we
ligation is an effective means to facilitate our target further demonstrated that the HSP90−CDC37 complex is
identifications of the apoptosis inducers such as ainsliatrimer crucial for RIPK3-dependent necroptosis.47 In summary, we
G https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

Figure 7. TQ-ligation facilitated the elucidation of mode of action for ainsliatrimer A. (A) Designed ainsliatrimer A TV-probe and fluorescein-
oQQM for imaging cellular distribution of ainsliatrimer A. (B) Fluorescent microscopy indicates nuclear localization of ainsliatrimer A. (C)
Structures of ainsliatrimer A probe and negative control probe. (D) Pull-down assays by ainsliatrimer A probe revealed potential binding proteins.

have elucidated the mode of action of KA: the covalent immune disorders, which are caused by dysfunction of the
interaction of KA to Cys420 of HSP90 disrupts the formation regulated cell death machinery.
of the HSP90−CDC37 complex and subsequently inhibits Forward chemical genetics has been proven to be a very
RIPK3-dependent necroptosis.42 This study provided a new powerful approach to the discovery of new targets. However,
means to develop a HSP90 inhibitor for the treatment of the most significant challenge associated with this approach is
necroptosis-related diseases such as inflammation and identification of the functional targets of small-molecule
atherosclerosis.48 probes. The great potential of chemical genetics cannot be


realized without the establishment of efficient means for target
CONCLUSION identification. In this regard, synthetic chemistry plays an
important role in the development of effective chemical
Our laboratory conducts research at the interface between probes, especially for the complex natural product derived
chemistry and biology. We systematically use bioactive small probes. Many issues such as thorough SAR studies, the choice
molecules including complex natural products to dissect of labeling, and target ID methods should be carefully
fundamental biological process and to develop novel considered for the chemical probe design and development.
therapeutic agents for the currently intractable human diseases. Several leading chemical biologists have recently published a
Over the past decade, we have extensively utilized various small commentary describing the key properties of successful
molecule probes to dissect the extrinsic programmed cell death chemical probes.49 These include high potency, good
pathways, including both apoptosis and programmed necrosis selectivity, and the availability of appropriate control
(necroptosis), and illuminated a number of new molecular compounds. In our studies, we have inevitably applied many
mechanisms underlying these fundamental cellular processes. different approaches, including direct labeling with biotin tags,
These discoveries not only significantly enriched our current utilization of bioorthogonal ligation methods, or application of
understanding of programmed cell death mechanisms, but also pretarget imaging. In our opinion, no unified method could be
led to the discovery of a number of potential drug leads for the successfully applied in all cases. Extensive evaluations of
treatment of cancers, ischemia-reperfusion injury, and auto- different approaches should be performed in order to obtain
H https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

Figure 8. Kongensin A blocks necroptosis by inhibiting the protein−protein interaction of HSP90 and its co-chaperone CDC37. (A) Structures of
kongensin A and kongensin A thiovinyl ether probe. (B) Target identification revealed covalent binding of kongensin A on HSP90. (C) List of
possible interacting proteins for kongensin A. (D) Site-directed mutagenesis indicated the binding site of Hsp90 is C420. (E) Graphical abstract of
the mode of action of kongensin A.

the most conclusive results. Overall, our work to date provides In conclusion, we can certainly envision that the combined
a model workflow for the transformation of bioactive small efforts of function-oriented synthesis and forward chemical
molecules including complex natural products into effective genetics will continue to uncover the unknown molecular
chemical probes and the identification of their functional mechanisms of fundamental cellular process such as pro-
protein targets. grammed cell death to advance our understanding of human
I https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research


pubs.acs.org/accounts Article

biology and provide new meaningful approaches to combat ACKNOWLEDGMENTS


human diseases. We are grateful to our students, postdocs, and collaborators

■ AUTHOR INFORMATION
Corresponding Author
who have been involved in these studies over the past decade.
Financial support from the National Natural Science
Foundation of China Grant (21625201, 21961142010,
21661140001, 91853202, and 21521003), the National Key
Xiaoguang Lei − Beijing National Laboratory for Molecular Research and Development Program of China
Sciences, Key Laboratory of Bioorganic Chemistry and (2017YFA0505200), and the Beijing Outstanding Young
Molecular Engineering of Ministry of Education, Synthetic and Scientist Program (BJJWZYJH01201910001001) is gratefully
Functional Biomolecules Center, College of Chemistry and acknowledged.


Molecular Engineering and Peking-Tsinghua Center for Life
Science, Peking University, Beijing 100871, China;
orcid.org/0000-0002-0380-8035; Email: xglei@ REFERENCES
pku.edu.cn (1) Kerr, J. F. R.; Wyllie, A. H.; Currie, A. R. Apoptosis - Basic
Biological Phenomenon with Wide-Ranging Implications in Tissue
Authors Kinetics. Br. J. Cancer 1972, 26, 239−257.
(2) Elmore, S. Apoptosis: A review of programmed cell death.
Yingjie Bai − Beijing National Laboratory for Molecular
Toxicol. Pathol. 2007, 35, 495−516.
Sciences, Key Laboratory of Bioorganic Chemistry and (3) Paweletz, N. Walther Flemming: pioneer of mitosis research.
Molecular Engineering of Ministry of Education, Synthetic and Nat. Rev. Mol. Cell Biol. 2001, 2, 72−75.
Functional Biomolecules Center, College of Chemistry and (4) Kerr, J. F. R. History of the events leading to the formulation of
Molecular Engineering, Peking University, Beijing 100871, the apoptosis concept. Toxicology 2002, 181, 471−474.
China (5) Debnath, J.; Baehrecke, E. H.; Kroemer, G. Does autophagy
Hiu C. Lam − Beijing National Laboratory for Molecular contribute to cell death? Autophagy 2005, 1, 66−74.
Sciences, Key Laboratory of Bioorganic Chemistry and (6) Degterev, A.; Huang, Z. H.; Boyce, M.; Li, Y. Q.; Jagtap, P.;
Molecular Engineering of Ministry of Education, Synthetic and Mizushima, N.; Cuny, G. D.; Mitchison, T. J.; Moskowitz, M. A.;
Functional Biomolecules Center, College of Chemistry and Yuan, J. Y. Chemical inhibitor of nonapoptotic cell death with
Molecular Engineering, Peking University, Beijing 100871, therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 2005,
China 1, 112−119.
(7) Dixon, S. J.; Lemberg, K. M.; Lamprecht, M. R.; Skouta, R.;
Complete contact information is available at: Zaitsev, E. M.; Gleason, C. E.; Patel, D. N.; Bauer, A. J.; Cantley, A.
https://pubs.acs.org/10.1021/acs.accounts.9b00600 M.; Yang, W. S.; Morrison, B.; Stockwell, B. R. Ferroptosis: An Iron-
Dependent Form of Nonapoptotic Cell Death. Cell 2012, 149, 1060−
Notes 1072.
(8) Bergsbaken, T.; Fink, S. L.; Cookson, B. T. Pyroptosis: host cell
The authors declare no competing financial interest. death and inflammation. Nat. Rev. Microbiol. 2009, 7, 99−109.
(9) Galluzzi, L.; Vitale, I.; Aaronson, S. A.; Abrams, J. M.; Adam, D.;
Biographies
Agostinis, P.; Alnemri, E. S.; Altucci, L.; Amelio, I.; Andrews, D. W.;
Yingjie Bai received his B.Sc. degree at Peking University in 2017. He Annicchiarico-Petruzzelli, M.; Antonov, A. V.; Arama, E.; Baehrecke,
is currently conducting his Ph.D. research under the supervision of E. H.; Barlev, N. A.; Bazan, N. G.; Bernassola, F.; Bertrand, M. J. M.;
Bianchi, K.; Blagosklonny, M. V.; Blomgren, K.; Borner, C.; Boya, P.;
Prof. Xiaoguang Lei at Peking University to elucidate biological
Brenner, C.; Campanella, M.; Candi, E.; Carmona-Gutierrez, D.;
functions of human endogenous lipids. Cecconi, F.; Chan, F. K. M.; Chandel, N. S.; Cheng, E. H.; Chipuk, J.
Hiu Chun Lam was born and raised in Hong Kong. In 2013, he E.; Cidlowski, J. A.; Ciechanover, A.; Cohen, G. M.; Conrad, M.;
received his Bachelor degree in Chemistry with 1st class Honours at Cubillos-Ruiz, J. R.; Czabotar, P. E.; D’Angiolella, V.; Dawson, T. M.;
the University of Adelaide, Australia. In 2018, Hiu completed his Dawson, V. L.; De Laurenzi, V.; De Maria, R.; Debatin, K. M.;
DeBerardinis, R. J.; Deshmukh, M.; Di Daniele, N.; Di Virgilio, F.;
Ph.D. in Chemistry under the supervision of Associate Professor
Dixit, V. M.; Dixon, S. J.; Duckett, C. S.; Dynlacht, B. D.; El-Deiry, W.
Jonathan George at the University of Adelaide. Hiu is currently a S.; Elrod, J. W.; Fimia, G. M.; Fulda, S.; Garcia-Saez, A. J.; Garg, A.
postdoctoral fellow in Prof. Xiaoguang Lei’s group at Peking D.; Garrido, C.; Gavathiotis, E.; Golstein, P.; Gottlieb, E.; Green, D.
University. His research interest focuses on the total synthesis and R.; Greene, L. A.; Gronemeyer, H.; Gross, A.; Hajnoczky, G.;
mode of action study of complex natural products. Hardwick, J. M.; Harris, I. S.; Hengartner, M. O.; Hetz, C.; Ichijo, H.;
Jaattela, M.; Joseph, B.; Jost, P. J.; Juin, P. P.; Kaiser, W. J.; Karin, M.;
Xiaoguang Lei was born and raised in Beijing, China. He received
Kaufmann, T.; Kepp, O.; Kimchi, A.; Kitsis, R. N.; Klionsky, D. J.;
BSc degree in chemistry at Peking University in 2001 and then Knight, R. A.; Kumar, S.; Lee, S. W.; Lemasters, J. J.; Levine, B.;
received his Ph.D. in 2006 under the supervision of Prof. John. A. Linkermann, A.; Lipton, S. A.; Lockshin, R. A.; Lopez-Otin, C.; Lowe,
Porco at Boston University. After 2 years as a postdoctoral fellow in S. W.; Luedde, T.; Lugli, E.; MacFarlane, M.; Madeo, F.; Malewicz,
Prof. Samuel J. Danishefsky’s laboratory at Columbia University, Prof. M.; Malorni, W.; Manic, G.; et al. Molecular mechanisms of cell
Lei joined the faculty at the National Institute of Biological Sciences death: recommendations of the Nomenclature Committee on Cell
(NIBS) in Beijing as a Principal Investigator and Director of Death 2018. Cell Death Differ. 2018, 25, 486−541.
Chemistry Center. In 2014, Prof. Lei moved to Peking University and (10) Formigli, L.; Papucci, L.; Tani, A.; Schiavone, N.; Tempestini,
A.; Orlandini, G. E.; Capaccioli, S.; Orlandini, S. Z. Aponecrosis:
joined the faculty in the College of Chemistry and Molecular
Morphological and biochemical exploration of a syncretic process of
Engineering. He is currently a Full Professor of Chemistry and cell death sharing apoptosis and necrosis. J. Cell. Physiol. 2000, 182,
Chemical Biology at Peking University. His research program focuses 41−49.
on natural product synthesis, chemical biology, synthetic biology, and (11) Remijsen, Q.; Kuijpers, T. W.; Wirawan, E.; Lippens, S.;
drug discovery. Vandenabeele, P.; Vanden Berghe, T. Dying for a cause: NETosis,

J https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

mechanisms behind an antimicrobial cell death modality. Cell Death (32) Wu, Z. J.; Xu, X. K.; Shen, Y. H.; Su, J.; Tian, J. M.; Liang, S.;
Differ. 2011, 18, 581−588. Li, H. L.; Liu, R. H.; Zhang, W. D. Ainsliadimer A, A new
(12) Weerasinghe, P.; Buja, L. M. Oncosis: an important non- sesquiterpene lactone dimer with an unusual carbon skeleton from
apoptotic mode of cell death. Exp. Mol. Pathol. 2012, 93, 302−308. Ainsliaea macrocephala. Org. Lett. 2008, 10, 2397−2400.
(13) Kolb, H. C.; Finn, M. G.; Sharpless, K. B. Click Chemistry: (33) Li, C.; Yu, X. L.; Lei, X. G. A Biomimetic Total Synthesis of
Diverse Chemical Function from a Few Good Reactions. Angew. (+)-Ainsliadimer A. Org. Lett. 2010, 12, 4284−4287.
Chem., Int. Ed. 2001, 40, 2004−2021. (34) Li, C.; Dian, L. Y.; Zhang, W. D.; Lei, X. G. Biomimetic
(14) Saxon, E.; Bertozzi, C. R. Cell surface engineering by a modified Syntheses of (−)-Gochnatiolides A-C and (−)-Ainsliadimer B. J. Am.
Staudinger reaction. Science 2000, 287, 2007−2010. Chem. Soc. 2012, 134, 12414−12417.
(15) McKay, C. S.; Finn, M. G. Click Chemistry in Complex (35) Li, C.; Dong, T.; Dian, L. Y.; Zhang, W. D.; Lei, X. G.
Mixtures: Bioorthogonal Bioconjugation. Chem. Biol. 2014, 21, 1075− Biomimetic syntheses and structural elucidation of the apoptosis-
1101. inducing sesquiterpenoid trimers: (−)-ainsliatrimers A and B. Chem.
(16) Zhang, Z. J.; Wang, Y. C.; Yang, X.; Hang, H. C. Chemical Sci. 2013, 4, 1163−1167.
Reporters for Exploring Microbiology and Microbiota Mechanisms. (36) Li, C.; Jones, A. X.; Lei, X. G. Synthesis and mode of action of
ChemBioChem 2020, 21, 19−32. oligomeric sesquiterpene lactones. Nat. Prod. Rep. 2016, 33, 602−611.
(17) Agarwal, P.; Bertozzi, C. R. Site-specific antibody-drug (37) Dong, T.; Li, C.; Wang, X.; Dian, L. Y.; Zhang, X. G.; Li, L.;
conjugates: the nexus of bioorthogonal chemistry, protein engineer- Chen, S.; Cao, R.; Li, L.; Huang, N.; He, S. D.; Lei, X. G. Ainsliadimer
ing, and drug development. Bioconjugate Chem. 2015, 26, 176−192. A selectively inhibits IKK α/β by covalently binding a conserved
(18) Agarwal, P.; Beahm, B. J.; Shieh, P.; Bertozzi, C. R. Systemic cysteine. Nat. Commun. 2015, 6, 6522.
Fluorescence Imaging of Zebrafish Glycans with Bioorthogonal (38) Li, Q.; Dong, T.; Liu, X. H.; Lei, X. G. A Bioorthogonal
Chemistry. Angew. Chem., Int. Ed. 2015, 54, 11504−11510. Ligation Enabled by Click Cycloaddition of o-Quinolinone Quinone
(19) Schanzenbacher, C. T.; Sambandan, S.; Langer, J. D.; Schuman, Methide and Vinyl Thioether. J. Am. Chem. Soc. 2013, 135, 4996−
E. M. Nascent Proteome Remodeling following Homeostatic Scaling 4999.
at Hippocampal Synapses. Neuron 2016, 92, 358−371. (39) Zhang, X. Y.; Dong, T.; Li, Q.; Liu, X. H.; Li, L.; Chen, S.; Lei,
(20) Stockwell, B. R.; Friedmann Angeli, J. P.; Bayir, H.; Bush, A. I.; X. G. Second Generation TQ-Ligation for Cell Organelle Imaging.
Conrad, M.; Dixon, S. J.; Fulda, S.; Gascon, S.; Hatzios, S. K.; Kagan, ACS Chem. Biol. 2015, 10, 1676−1683.
V. E.; Noel, K.; Jiang, X.; Linkermann, A.; Murphy, M. E.; (40) Agard, N. J.; Prescher, J. A.; Bertozzi, C. R. A strain-promoted
Overholtzer, M.; Oyagi, A.; Pagnussat, G. C.; Park, J.; Ran, Q.; [3 + 2] azide-alkyne cycloaddition for covalent modification of
Rosenfeld, C. S.; Salnikow, K.; Tang, D.; Torti, F. M.; Torti, S. V.; blomolecules in living systems. J. Am. Chem. Soc. 2004, 126, 15046−
Toyokuni, S.; Woerpel, K. A.; Zhang, D. D. Ferroptosis: A Regulated 15047.
Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. (41) Li, C.; Dong, T.; Li, Q.; Lei, X. G. Probing the Anticancer
Cell 2017, 171, 273−285. Mechanism of (−)-Ainsliatrimer A through Diverted Total Synthesis
(21) Declercq, W.; Vanden Berghe, T.; Vandenabeele, P. RIP kinases
and Bioorthogonal Ligation. Angew. Chem., Int. Ed. 2014, 53, 12111−
at the crossroads of cell death and survival. Cell 2009, 138, 229−232.
12115.
(22) He, S.; Wang, L.; Miao, L.; Wang, T.; Du, F.; Zhao, L.; Wang,
(42) Li, D. R.; Li, C.; Li, L.; Chen, S.; Wang, L.; Li, Q.; Wang, X. D.;
X. Receptor interacting protein kinase-3 determines cellular necrotic
Lei, X. G.; Shen, Z. R. Natural Product Kongensin A is a Non-
response to TNF-α. Cell 2009, 137, 1100−1111.
Canonical HSP90 Inhibitor that Blocks RIP3-dependent Necroptosis.
(23) Li, L.; Thomas, R. M.; Suzuki, H.; De Brabander, J. K.; Wang,
Cell Chem. Biol. 2016, 23, 257−266.
X.; Harran, P. G. A small molecule Smac mimic potentiates TRAIL-
(43) Ahmadian, M.; Suh, J. M.; Hah, N.; Liddle, C.; Atkins, A. R.;
and TNFα-mediated cell death. Science 2004, 305, 1471−1474.
(24) Sun, L. M.; Wang, H. Y.; Wang, Z. G.; He, S. D.; Chen, S.; Liao, Downes, M.; Evans, R. M. PPARγ signaling and metabolism: the
D. H.; Wang, L.; Yan, J. C.; Liu, W. L.; Lei, X. G.; Wang, X. D. Mixed good, the bad and the future. Nat. Med. 2013, 19, 557−566.
Lineage Kinase Domain-like Protein Mediates Necrosis Signaling (44) Grommes, C.; Landreth, G. E.; Heneka, M. T. Antineoplastic
Downstream of RIP3 Kinase. Cell 2012, 148, 213−227. effects of peroxisome proliferator-activated receptor gamma agonists.
(25) Green, D. R. Apoptotic pathways: ten minutes to dead. Cell Lancet Oncol. 2004, 5, 419−429.
2005, 121, 671−674. (45) Chen, W.; Yang, X. D.; Zhao, J. F.; Yang, J. H.; Zhang, H. B.; Li,
(26) Wiezorek, J.; Holland, P.; Graves, J. Death receptor agonists as Z. Y.; Li, L. Three new, 1-oxygenated ent-8,9-secokaurane diterpenes
a targeted therapy for cancer. Clin. Cancer Res. 2010, 16, 1701−1708. from Croton kongensis. Helv. Chim. Acta 2006, 89, 537−541.
(27) Wang, G.; Wang, X.; Yu, H.; Wei, S.; Williams, N.; Holmes, D. (46) Stepanova, L.; Leng, X. H.; Parker, S. B.; Harper, J. W.
L.; Halfmann, R.; Naidoo, J.; Wang, L.; Li, L.; Chen, S.; Harran, P.; Mammalian p50(Cdc37) is a protein kinase-targeting subunit of
Lei, X.; Wang, X. Small-molecule activation of the TRAIL receptor Hsp90 that binds and stabilizes Cdk4. Genes Dev. 1996, 10, 1491−
DR5 in human cancer cells. Nat. Chem. Biol. 2013, 9, 84−89. 1502.
(28) Siegel, R. M.; Muppidi, J. R.; Sarker, M.; Lobito, A.; Jen, M.; (47) Li, D. R.; Xu, T.; Cao, Y.; Wang, H. Y.; Li, L.; Chen, S.; Wang,
Martin, D.; Straus, S. E.; Lenardo, M. J. SPOTS: signaling protein X. D.; Shen, Z. R. A cytosolic heat shock protein 90 and cochaperone
oligomeric transduction structures are early mediators of death CDC37 complex is required for RIP3 activation during necroptosis.
receptor-induced apoptosis at the plasma membrane. J. Cell Biol. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 5017−5022.
2004, 167, 735−744. (48) Linkermann, A.; Green, D. R. Necroptosis. N. Engl. J. Med.
(29) Bohlmann, F.; Ahmed, M.; Jakupovic, J.; King, R. M.; 2014, 370, 455−465.
Robinson, H. Dimeric Sesquiterpene Lactones and Kolavane (49) Arrowsmith, C. H.; Audia, J. E.; Austin, C.; Baell, J.; Bennett, J.;
Derivatives from Gochnatia-Paniculata. Phytochemistry 1983, 22, Blagg, J.; Bountra, C.; Brennan, P. E.; Brown, P. J.; Bunnage, M. E.;
191−195. Buser-Doepner, C.; Campbell, R. M.; Carter, A. J.; Cohen, P.;
(30) Bohlmann, F.; Zdero, C.; Schmedahirschmann, G.; Jakupovic, Copeland, R. A.; Cravatt, B.; Dahlin, J. L.; Dhanak, D.; Edwards, A.
J.; Dominguez, X. A.; King, R. M.; Robinson, H. Dimeric M.; Frederiksen, M.; Frye, S. V.; Gray, N.; Grimshaw, C. E.;
Guaianolides and Other Constituents from Gochnatia Species. Hepworth, D.; Howe, T.; Huber, K. V.; Jin, J.; Knapp, S.; Kotz, J. D.;
Phytochemistry 1986, 25, 1175−1178. Kruger, R. G.; Lowe, D.; Mader, M. M.; Marsden, B.; Mueller-
(31) Wang, Y.; Shen, Y. H.; Jin, H. Z.; Fu, J. J.; Hu, X. J.; Qin, J. J.; Fahrnow, A.; Muller, S.; O’Hagan, R. C.; Overington, J. P.; Owen, D.
Liu, J. H.; Chen, M.; Yan, S. K.; Zhang, W. D. Ainsliatrimers A and B, R.; Rosenberg, S. H.; Roth, B.; Ross, R.; Schapira, M.; Schreiber, S. L.;
the First Two Guaianolide Trimers from Ainsliaea fulvioides. Org. Shoichet, B.; Sundstrom, M.; Superti-Furga, G.; Taunton, J.; Toledo-
Lett. 2008, 10, 5517−5520. Sherman, L.; Walpole, C.; Walters, M. A.; Willson, T. M.; Workman,

K https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX
Accounts of Chemical Research pubs.acs.org/accounts Article

P.; Young, R. N.; Zuercher, W. J. The promise and peril of chemical


probes. Nat. Chem. Biol. 2015, 11, 536−541.

L https://dx.doi.org/10.1021/acs.accounts.9b00600
Acc. Chem. Res. XXXX, XXX, XXX−XXX

Вам также может понравиться