ArticlePDF AvailableLiterature Review

The roles of pyroptosis in genitourinary diseases

Authors:

Abstract and Figures

Pyroptosis, a form of programmed cell death distinct from apoptosis and necrosis, is thought to be closely associated with the pathogenesis of diseases. Recently, the association between pyroptosis and urinary diseases has attracted considerable attention, and a comprehensive review focusing on this issue is not available. In this study, we reviewed the role of pyroptosis in the development and progression of benign urinary diseases and urinary malignancies. Based on this, pyroptosis has been implicated in the development of urinary diseases. In summary, this review sheds light on future research directions and provides novel ideas for using pyroptosis as a powerful tool to fight urinary diseases.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
International Urology and Nephrology (2024) 56:1515–1523
https://doi.org/10.1007/s11255-023-03894-6
UROLOGY - REVIEW
The roles ofpyroptosis ingenitourinary diseases
HaopengLiu1· HaoranLiu1· GuoshuaiHuang1· HexingYuan1· XuefengZhang1
Received: 2 September 2023 / Accepted: 15 November 2023 / Published online: 16 December 2023
© The Author(s) 2023
Abstract
Pyroptosis, a form of programmed cell death distinct from apoptosis and necrosis, is thought to be closely associated with
the pathogenesis of diseases. Recently, the association between pyroptosis and urinary diseases has attracted considerable
attention, and a comprehensive review focusing on this issue is not available. In this study, we reviewed the role of pyropto-
sis in the development and progression of benign urinary diseases and urinary malignancies. Based on this, pyroptosis has
been implicated in the development of urinary diseases. In summary, this review sheds light on future research directions
and provides novel ideas for using pyroptosis as a powerful tool to fight urinary diseases.
Keywords Pyroptosis· Regulated cell death· Genitourinary diseases· Mechanism· Gasdermins
Abbreviations
ASC Caspase-recruitment domain
ROS Reactive oxygen species
IL-18 Interleukin-18
IL-1β Interleukin-1β
LPS Lipopolysaccharide
TAK1 TGF-β-activated kinase-1
GSDMA Gasdermin A
GSDMB Gasdermin B
GSDMC Gasdermin C
GSDMD Gasdermin D
GSDME Gasdermin E
GZMB Granzyme B
GZMA Granzyme A
USF2 Upstream stimulatory factor 2
THBS1 Thrombospondin-1
NLRP1 NOD-like receptor 1
AIM2 Absent in melanoma 2
NLRP3 NOD-like receptor 3
HMGB1 High mobility group box1
ERS Endoplasmic reticulum stress
TLR2 Toll-like receptor 2
ROCK1 Rho-associated coiled-coil containing protein
kinase-1
TXNIP Thioredoxin–interacting protein
miR-93 MicroRNA-93
PRDX3 Peroxiredoxin 3
STAT3 Transcription 3
HK2 Hexokinase 2
LDHA Lactate dehydrogenase A
ENO2 Enolase 2
USP24 Ubiquitin-specific peptidase 24
IGFBP3 Insulin-like growth factor-binding protein 3
Introduction
Pyroptosis is a newly discovered form of programmed cell
death. Cell death is usually categorized as nonprogrammed
cell death and programmed cell death (PCD) [1]. Pyroptosis
is a type of inflammatory PCD [2]. The process of pyrop-
tosis was first described in 1992, but the term was coined
in 2001 following the observation that bacterium-infected
macrophages underwent rapid lytic cell death dependent on
caspase-1 activity [3]. Recently, macrophages were shown to
regulate pyroptosis and play an important role in the devel-
opment of acute kidney injury (AKI), diabetic nephropathy
(DN) and renal fibrosis [46]. Pyroptosis is characterized
by cell membrane pore formation, cell swelling, and the
release of inflammatory intracellular contents [7, 8]. The
inflammatory factors released during cell lysis, such as
interleukin-1β (IL-1β) and interleukin-18 (IL-18), amplify
* Hexing Yuan
yuanhexing@suda.edu.cn
* Xuefeng Zhang
zhangxuefeng626@sina.com
1 Department ofUrology, The First Affiliated Hospital
ofSoochow University, 188 Shizi Road, Suzhou, China
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1516 International Urology and Nephrology (2024) 56:1515–1523
1 3
the inflammatory effects and activate immune responses [7,
8].
The underlying mechanism was only uncovered upon
the discovery of gasdermin D (GSDMD) protein. Shi etal.
found that caspase-1/11/4/5 can induce pyroptosis by cleav-
ing GSDMD to release its N-terminal domain [9]. In addi-
tion to GSDMD, the gasdermin family also includes five
other members. The human gasdermin family comprises
GSDMA, GSDMB, GSDMC, GSDMD, GSDME/DFNA5,
and PVJK/DFNB59. In mice, there are five gasdermin mem-
bers, namely, GSDMA, GSDMC, GSDMD, GSDME, and
PJVK/DFNB59, but not GSDMB [2]. All gasdermins except
DFNB59 have two conserved domains: an N-terminal effec-
tor domain and a C-terminal inhibitory domain [2].
Normally, moderate pyroptosis contributes to host
defence against pathogen infection, but excessive pyropto-
sis leads to uncontrolled inflammatory responses, massive
cell death, and serious tissue damage, causing inflammatory
or autoimmune diseases [2]. As a proinflammatory type of
cell death, pyroptosis provides a new opportunity for cancer
elimination by activating the anti-tumour immune response
[2]. An increasing number of studies have shown that pyrop-
tosis plays a crucial role in many cancers, such as breast
cancer, gastric cancer, and lung cancer [1012].
Here, we first describe the different signalling path-
ways of pyroptosis to gain an in-depth understanding of
the molecular mechanism. Finally, the role of pyroptosis in
urinary diseases is discussed, followed by suggestions for
future research directions.
Overview ofpyroptosis
Canonical pathway
The classical pyroptosis pathway is mediated by caspase-1
[13]. Inflammasomes are formed by pattern-recognition
receptors (PRRs, also known as inflammasome sensors),
apoptosis-associated speck-like protein containing a cas-
pase-recruitment domain (ASC), and inactive pro-caspase-1
[1315]. PRRs can recognize pathogen-associated molecular
patterns and danger-associated molecular patterns (PAMPs
and DAMPs) [16, 17]. PRRs include nucleotide-binding
oligomerization domain-like receptors (NLRs, includ-
ing NLRP1, NLRP3, and NLRC4), absent in melanoma
2 (AIM2), and pyrin [18, 19]. NLRs usually consist of a
leucine-rich repeat (LRR), a nucleotide-binding oligomeri-
zation domain (NACHT/NOD), and a caspase-recruitment
domain (CARD) or pyrin domain (PYD) and are divided
into NLRPs or NLRCs according to whether their N-termi-
nus contains a PYD or CARD [20]. A PYD is needed for
interaction with ASC. The NOD participates in adenosine
triphosphate (ATP)-dependent activation of the signal. The
LRR is responsible for ligand recognition and autoinhibi-
tion. The CARD participates in pro-caspase-1 recruitment
[2]. Upon receiving an activating signal, inflammasome
sensors recruit pro-caspase-1 (which has a CARD) either
directly through homotypic binding of CARD or indirectly
through the PYD by means of ASC, which contains a PYD
and a CARD [17]. Subsequently, caspase-1 activation
occurs through self-cleavage. Activated caspase-1 not only
cleaves inactive IL-1β and IL-18 precursors but also cleaves
GSDMD to form GSDMD-NT and GSDMD-CT [2124].
GSDMD-N forms pores in the plasma membrane, leading
to cell swelling and pyroptosis [25, 26] (Fig.1).
Non‑canonical pathway
Most gram-negative bacteria activate the non-canonical
inflammasome pathway [2]. The nonclassical signalling
pathway is mediated by caspase-4 and caspase-5 in humans
and by caspase-11 in mice [27, 28]. These caspases can be
activated by directly binding to lipopolysaccharide (LPS)
[28]. Activated caspase-4/5/11 cleaves GSDMD to promote
pyroptosis. However, caspase-4/5/11 cannot cleave pro-
IL-18/pro-IL-1β but can cleave GSDMD, which can cause
K+ efflux and NLRP3/caspase-1 pathway activation, eventu-
ally leading to the maturation and release of interleukin-18
(IL-18) and interleukin-1β (IL-1β) [9, 14, 29] (Fig.1).
Apoptotic caspase‑mediated pathway
In addition to inflammatory caspase-1/4/5/11, some apop-
totic caspases can also trigger pyroptosis. Chemotherapeu-
tic drugs can induce caspase-3 to cleave GSDME to form
GSDME-N termini, which cause pyroptosis [30, 31]. In
addition, pathogenic Yersinia has been shown to inhibit
TGFβ-activated kinase-1 (TAK1) via the Yersinia effec-
tor protein YopJ and induce caspase-8-related cleavage of
GSDMD to elicit pyroptosis [32, 33]. Interestingly, cas-
pase-8 induces GSDMC cleavage, thereby leading to a non-
canonical pyroptosis pathway in cancer cells [34] (Fig.1).
Granzyme‑mediated pathway
Granzyme A (GzmA) is the most abundant serine protease
of the granzyme family and has traditionally been recog-
nized as a mediator of cell death [2]. Zhou etal. found
that GZMA derived from cytotoxic T lymphocytes cleaves
GSDMB to induce pyroptosis [35]. In 2020, it was reported
that CAR-T cells activated caspase-3 by releasing granzyme
B (GzmB), subsequently leading to the activation of the cas-
pase-3/GSDME-mediated pyroptotic pathway, thus causing
pyroptosis [36]. Additionally, Zhang etal. found that GzmB
directly cleaved GSDME and induced pyroptosis, enhancing
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1517International Urology and Nephrology (2024) 56:1515–1523
1 3
anti-tumour immunity and inhibiting tumour growth [37]
(Fig.1).
Pyroptosis inbenign urinary diseases
Pyroptosis ininterstitial cystitis
Interstitial cystitis (IC), also known as bladder pain syn-
drome (BPS), is a chronic pain disorder that most commonly
presents in the bladder, pelvis, or abdomen [38]. Pyroptosis
plays an important role in the development of IC. A study
showed that the NLRP3 inflammasome is a crucial player
in the development of bladder disease [39]. Some results
have demonstrated that the expression levels of NLRP3, cas-
pase-1, and GSDMD in patients with IC are elevated [40,
41]. Wang etal. found that the NLRP3/GSDMD-N pathway
was activated and played a role in the development of IC
[42]. Wang etal. showed that aster tataricus extract (ATE)
can be used as an inhibitor of NLRP3 in treating IC [43].
The discovery of NLRP3/caspase-1/GSDMD-N as a new
pathway provides a new direction for IC research.
Pyroptosis inBPH
Benign prostatic hyperplasia (BPH) is characterized by the
nonmalignant overgrowth of prostatic tissue surrounding the
urethra, ultimately constricting the urethral opening and giv-
ing rise to associated lower urinary tract symptoms (LUTS)
such as urgency, frequency, nocturia, incomplete bladder
emptying, and a weak urine stream [44]. There is much
evidence to suggest that inflammation plays an important
role in BPH. It has been reported that the expression lev-
els of NLRP1 and caspase-1, IL-18 and IL-1β are elevated
in BPH [45]. Therefore, the NLRP1/caspase-1 pathway
is activated and participates in the development of BPH.
Fig. 1 Molecular mechanisms of the canonical pathway, non-canon-
ical pathway, apoptotic caspase-mediated pathway and granzyme-
mediated pathway in pyroptosis. In the canonical pathway, patho-
gen-associated molecular patterns or damage-associated molecular
patterns (such as ROS, ATP, viruses, bacteria, or toxins) stimulate
inflammasomes, which then activate caspase-1. Activated cas-
pase-1 not only cleaves inactive IL-1β and IL-18 precursors but also
cleaves GSDMD, which forms pores and induces pyroptosis. In the
non-canonical pathway, LPS from Gram-negative bacteria activates
caspase-4/5/11, and activated caspase-4/5/11 cleaves GSDMD to
promote pyroptosis. In the apoptotic caspase-mediated pathway, cas-
pase-3/GSDME, caspase-8/GSDMD and caspase-8/GSDMC mecha-
nisms can promote pyroptosis. In the granzyme-mediated pathway,
GZMA or GZMB derived from CAR-T cells cleaves GSDMB or
GSDME, respectively, to induce pyroptosis. ASC caspase-recruit-
ment domain, ROS reactive oxygen species, IL-18 interleukin-18,
IL-1β interleukin-1β, LPS lipopolysaccharide, TAKI TGF-β-activated
kinase-1, GSDMD gasdermin D, GSDME gasdermin E, GSDMB gas-
dermin B, GZMB granzyme B, GZMA granzyme A
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1518 International Urology and Nephrology (2024) 56:1515–1523
1 3
Jiang etal. found that peroxiredoxin 3 (PRDX3) suppressed
autophagy flux and activated pyroptosis to induce inflam-
matory responses and stimulate the overgrowth of prostate
tissues [46]. Emerging results indicate that steady-state
levels of AIM2 mRNA are higher in BPH tissue than in
normal prostate tissue [47]. AIM2 recruits ASC and pro-
caspase-1 to assemble the AIM2 inflammasome, leading to
cell swelling and pyroptosis. These studies have facilitated
the identification of potential BPH treatment targets. The
signalling pathways regulating pyroptosis in BPH are dis-
played in Fig.3.
Pyroptosis inAKI
Acute kidney injury (AKI) is defined by a rapid increase
in serum creatinine, a decrease in urine output, or both
[48]. Recent advances have revealed a role for pyroptosis in
AKI. Sun etal. found that thrombospondin-1 (THBS1) and
upstream stimulatory factor 2 (USF2) were highly expressed
in patients with sepsis-induced AKI and that USF2 upregu-
lated THBS1 expression to activate the TGF-β/Smad3/
NLRP3/caspase-1 signalling pathway and stimulate pyrop-
tosis, ultimately exacerbating sepsis-induced AKI [49]. Miao
etal. found that the expression of GSDMD was significantly
increased in both cisplatin-induced and ischaemiareper-
fusion (I/R) models [50]. The knockout of caspase-11 or
GSDMD alleviated kidney damage in mice with cisplatin-
induced AKI. A study published in 2020 showed that the
protein levels of high mobility group box1 (HMGB1),
IL-1β, IL-18, NLRP3, and GSDMD were elevated in an
AKI model [6]. Therefore, we hypothesize that the HMGB1/
NLRP3/GSDMD signalling pathway plays a pivotal role in
the pathogenesis of AKI. In addition, Li etal. demonstrated
that the ROS/NLRP3/caspase-1/GSDMD pathway medi-
ated contrast-induced AKI (CI-AKI) via pyroptosis and
that baicalin treatment alleviated the associated inflamma-
tion and oxidation levels [51]. Studies have also shown that
macrophage-derived exosomal miRNAs play important roles
in AKI [52, 53]. Xia etal. found that the levels of GSDME-
N and IL-1β were elevated in cisplatin-induced AKI [54].
The inhibition of caspase-3 blocked GSDME-N cleavage
and attenuated cisplatin-induced pyroptosis and kidney dys-
function. Therefore, caspase-3/GSDME-triggered pyroptosis
plays an important role in AKI. Juan etal. found that the
exosomal miR-93/thioredoxin–interacting protein (TXNIP)
signalling pathway plays a crucial role in the progression of
sepsis-induced AKI and that M1 exosomes promote pyrop-
tosis and M2 exosomes inhibit pyroptosis [55]. It has been
well established that Rho-associated coiled-coil containing
protein kinase-1 (ROCK1) plays an important role in a series
of pathological processes, including pyroptosis, inflamma-
tion, and endoplasmic reticulum stress (ERS) [56, 57]. Wang
etal. found that ROCK1 regulates LPS-induced kidney cell
pyroptosis via Toll-like receptor 2 (TLR2)-mediated ERS,
thereby accelerating sepsis-induced AKI progression [58].
The signalling pathways regulating pyroptosis in AKI are
displayed in Fig.2.
Pyroptosis inDN
Diabetic nephropathy (DN), or diabetic kidney disease
(DKD), is a frequent and severe long-term microvascular
complication resulting from lesions in the renal glomeruli
and tubules [59]. Growing evidence has demonstrated that
chronic inflammation promotes the pathogenesis of DN
[60]. The role of pyroptosis signalling pathways in DN
progression has attracted the attention of researchers and
clinicians. In 2020, it was reported that the TXNIP/NLRP3
axis is an important pathway that regulates DN induced by
pyroptosis [61]. Interestingly, Ke etal. found that the ERS-
related factor IRE1α upregulated TXNIP/NLRP3 inflamma-
some-induced pyroptosis in DN rats [62]. Li etal. found
that NLRP3/caspase-1/GSDMD signalling was strikingly
upregulated and the secretion of IL-1β and IL-18 dramati-
cally increased in DN mice [63]; in addition, they also con-
firmed that SYR inhibited the NLRP3/caspase-1/GSDMD
pyroptosis pathway by upregulating NRF2 signalling in
DN. Li etal. found that the expression of p-NF-κB, ASC,
cleaved-IL-1β, NLRP3, cleaved-caspase-1, and GSDMD-N
was elevated in a DN mouse model [64]; in addition, they
confirmed that geniposide (GE) may inhibit the develop-
ment of DN via the APMK/SIRT1/NF-κB pathway [64].
The APMK/SIRT1/NF-κB axis may become a new signal-
ling pathway for the treatment of DN. In addition, NLRP3
inflammasome activation is related to the pathogenesis of
DN. Wang etal. revealed that the expression of NLRC4,
IL-1β, and IL-18 was increased under high glucose condi-
tions, inducing pyroptosis in renal tubular epithelial cells
[65]. Komada etal. demonstrated that the activation of the
AIM2 inflammasome by DNA from necrotic cells drives
pyroptosis, which contributes to chronic kidney injury [66].
Cheng etal. demonstrated that caspase-11/4- and GSDMD-
mediated pyroptosis was activated in a DN mouse model
and involved in the development of DN [67]. In summary,
these findings confirm that pyroptosis and inflammasomes
play important roles in renal injury, ultimately affecting the
pathogenesis of DN.
Pyroptosis inurinary malignancies
Pyroptosis inbladder cancer
Bladder cancer (BCa) is the most common malignancy of
the urinary tract [68]. Recent advances have revealed an
important role of pyroptosis in bladder cancer. He etal.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1519International Urology and Nephrology (2024) 56:1515–1523
1 3
found that GSDMB binds to signal transducer and activator
of transcription 3 (STAT3) and increases the phosphoryla-
tion of STAT3, which increases the expression of hexoki-
nase 2 (HK2), lactate dehydrogenase A (LDHA), enolase
2 (ENO2), and insulin-like growth factor-binding protein 3
(IGFBP3) to enhance glycolysis in BCa cells and promote
cancer cell proliferation [69]; in addition, they also demon-
strated that ubiquitin-specific peptidase 24 (USP24) inter-
acts with GSDMB and prevents GSDMB degradation in
BCa cells [69]. Therefore, the USP24/GSDMB/STAT3 axis
may become a new targetable signalling pathway for blad-
der cancer treatment. Chen etal. showed, based on KM
Fig. 2 Signalling pathways regulating pyroptosis in AKI. THBS1
is upregulated by USF2 and activates the TGF-β/Smad3/NLRP3/
caspase-1 signalling pathway, thus inducing pyroptosis. NLRP3 is
upregulated by HMGB1 and activates the expression of GSDMD.
ROS induce pyroptosis via the NLRP3/caspase-1/GSDMD signalling
axis. Cisplatin induces pyroptosis via the caspase-3/GSDME signal
axis. miR-93 targets TXN2P and thus induces pyroptosis. ROCK1
regulates LPS-induced pyroptosis via TLR2-mediated ERS. USF2
upstream stimulatory factor 2, ROS reactive oxygen species, THBS1
thrombospondin-1, TGF-β transforming growth factor-β, NLRP3
NOD-like receptor 3, HMGB1 high mobility group box 1, LPS
lipopolysaccharide, ERS endoplasmic reticulum stress, TLR2 toll-
like receptor 2, ROCK1 Rho-associated coiled-coil containing pro-
tein kinase-1, TXNIP thioredoxin–interacting protein, miR-93 micro-
RNA-93, GSDMD gasdermin D, GSDME gasdermin E
Fig. 3 Signalling pathways
regulating pyroptosis in BPH.
NLRP1/caspase-1 induces
pyroptosis to promote the
development of BPH. PRDX3
suppresses autophagy flux
and activates pyroptosis to
promote the development of
BPH. AIM2/caspase-1 induces
pyroptosis to promote the
development of BPH. PRDX3
peroxiredoxin 3, NLRP1 NOD-
like receptor 1, AIM2 absent
in melanoma 2, BPH benign
prostatic hyperplasia
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1520 International Urology and Nephrology (2024) 56:1515–1523
1 3
curves, that GSDMB and CASP6 are associated with better
prognoses for patients with BCa [70]; they also found that
many tumours with high GSDMB and CASP6 expression
were immune-inflamed tumours and that many tumours with
low GSDMB and CASP6 expression were immune-desert
tumours. Then, they demonstrated that GSDMB and CASP6
play important roles in immune infiltration [70]. The results
from El-Gamal etal. showed that the expression level of
GSDMD in muscle-invasive bladder cancer (MIBC) was sig-
nificantly higher than that in non-muscle-invasive bladder
cancer (NMIBC) and that the expression level in NMIBC
was higher than that in the control group [71]. These results
show that GSDMD is involved in the pathogenesis of BCa
and muscle invasion. In addition, the expression of GSDMD
in tissue can be used as a useful tool for predicting local
tumour recurrence [71]. Peng etal. found that CD147 pro-
moted cell proliferation in BCa by upregulating the expres-
sion of GSDMD [72].
Pyroptosis inprostate cancer
Prostate cancer (PCa) is a major disease that affects men’s
health worldwide. It is the second most common form of
cancer in men, surpassed only by nonmelanoma skin cancers
such as basal and squamous cell carcinomas [73]. Pyroptosis
is also involved in PCa development. As a classical pyropto-
sis pathway, the caspase-1 pathway plays an important role
in PCa. NLRP3 participates in physiological and pathologi-
cal processes, including tumour progression. In 2021, Xu
etal. found that the expression of NLRP3 in PCa tissues
and cell lines was elevated and was positively correlated
with that of caspase-1 [74]. Their results revealed that the
NLRP3 inflammasome exerted a tumour-promoting effect by
activating caspase-1 in PCa [74]. Karan etal. reported that
the expression of NLRP12 was significantly higher in PCa
tissue than in adjacent benign tissue and that NLRP12 may
play an important role in activating NF-κB and IL-1β signal-
ling and its association with the pathogenesis and progres-
sion of PCa [75]; they indicated that NLRP12 can upregu-
late caspase-1, IL-1 β, and IL-18 to promote the occurrence
and progression of PCa. Many studies have shown that LPS
participates in the proliferation, migration, and invasion of
PCa cells [7678]. It has been shown that LPS activates the
caspase-4/5/11 pathway to induce pyroptosis [28]. However,
LPS-mediated pyroptosis is still being investigated in PCa.
Pyroptosis inrenal cell carcinoma
Renal cell carcinoma (RCC) accounts for 2–3% of all malig-
nant diseases in adults [79]. It is the seventh most com-
mon cancer in men and the ninth most common in women
[79]. The most common RCC is clear cell RCC (ccRCC)
(70–90%), followed by papillary RCC (10–15%) and
chromophobe RCC (3–5%) [80]. In recent years, research-
ers have found that pyroptosis is inextricably linked to the
development of RCC. Cui etal. found that GSDMB expres-
sion was significantly more upregulated in ccRCC tissues
than in surrounding normal tissues [81]; in addition, they
confirmed that the upregulation of GSDMB is significantly
related to immune infiltrates and poor survival in ccRCC
[81]. GSDMB has the potential to become a biomarker for
poor prognosis and a potential target for immune therapy
in ccRCC. Liver X receptors [LXRs; nuclear receptor
subfamily 1, group H, member 2 (NR1H2, also known as
LXRB) and nuclear receptor subfamily 1, group H, member
3 (NR1H3, also known as LXRA)] belong to the nuclear
receptor superfamily and are expressed in various cells [82].
Wang etal. found that the expression levels of NLRP3 in
ccRCC tissue were significantly lower than those in normal
kidney tissue and that LXRα promoted tumour metastasis
by downregulating the NLRP3 inflammasome in ccRCC
[83]. In addition, bromodomain-containing 4 (BRD4) inhi-
bition was shown to prevent cell proliferation and epithe-
lial–mesenchymal transition (EMT) and play an anti-tumour
role in RCC by activating the NF-κB–NLRP3–caspase-1
pyroptosis signalling pathway [84]. Zhang etal. found that
the expression of most pyroptosis regulatory genes is posi-
tively correlated and plays an important prognostic role in
ccRCC [85]. AIM2 plays a crucial role in the development
of various tumours. Recent studies have shown that AIM2 is
highly expressed in ccRCC and promotes tumour develop-
ment through immune activation pathways [86]. Tang etal.
found that lncRNA FOXD2 adjacent opposite strand RNA
1 (FOXD2-AS1) affects GSDMB and NLRP1 [87]; interest-
ingly, they also found that downregulating the expression
of FOXD2-AS1 reduced the proliferation and migration of
ccRCC cells [87]. This indicates that FOXD2-AS1 may pro-
vide a new direction for research on the treatment of RCC.
Conclusion
In conclusion, pyroptosis is a newly identified form of cell
death mediated by gasdermin proteins, which are often
activated by caspases. It plays a crucial role in the occur-
rence, development, and progression of urologic diseases.
The molecular mechanism of pyroptosis is shown in Fig.1.
The signalling pathways regulating pyroptosis in AKI
are shown in Fig.2. The signalling pathways regulating
pyroptosis in BPH are shown in Fig.3. Future in-depth
research on pyroptosis in urological diseases will help us
better understand the diagnosis and treatment of urinary
diseases. Future studies are urgently needed to develop
more clinical trials to explore the potential application of
pyroptosis in urinary diseases.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1521International Urology and Nephrology (2024) 56:1515–1523
1 3
Acknowledgements The authors would like to thank their families
for their constant support and encouragement throughout this review.
Author contributions HL, HY, and XZ conceived the study and drafted
the article. HL was a major contributor in writing the manuscript.
HL and GH provided suggestions to improve it. All authors read and
approved the final manuscript.
Funding The authors did not receive support from any organization
for the submitted work.
Declarations
Conflict of interest The authors declare that the research was con-
ducted in the absence of any commercial or financial relationships that
could be construed as a potential conflict of interest.
Informed consent Not applicable.
Ethics approval and consent to participate Not applicable.
Open Access This article is licensed under a Creative Commons Attri-
bution 4.0 International License, which permits use, sharing, adapta-
tion, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are
included in the article’s Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in
the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a
copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
References
1. Xia X, Wang X, Cheng Z etal (2019) The role of pyroptosis in
cancer: pro-cancer or pro-“host”? Cell Death Dis 10:650. https://
doi. org/ 10. 1038/ s41419- 019- 1883-8
2. Rao Z, Zhu Y, Yang P etal (2022) Pyroptosis in inflammatory
diseases and cancer. Theranostics 12:4310–4329. https:// doi. org/
10. 7150/ thno. 71086
3. Frank D, Vince JE (2019) Pyroptosis versus necroptosis: simi-
larities, differences, and crosstalk. Cell Death Differ 26:99–114.
https:// doi. org/ 10. 1038/ s41418- 018- 0212-6
4. Ding X, Jing N, Shen A etal (2021) MiR-21-5p in macrophage-
derived extracellular vesicles affects podocyte pyroptosis in
diabetic nephropathy by regulating A20. J Endocrinol Invest
44:1175–1184. https:// doi. org/ 10. 1007/ s40618- 020- 01401-7
5. Fu H, Gu YH, Tan J etal (2022) CircACTR2 in macrophages
promotes renal fibrosis by activating macrophage inflammation
and epithelial-mesenchymal transition of renal tubular epithe-
lial cells. Cell Mol Life Sci 79:253. https:// doi. org/ 10. 1007/
s00018- 022- 04247-9
6. Wang Y, Zhang H, Chen Q etal (2020) TNF-α/HMGB1 inflam-
mation signalling pathway regulates pyroptosis during liver failure
and acute kidney injury. Cell Prolif 53:e12829. https:// doi. org/ 10.
1111/ cpr. 12829
7. Fink SL, Cookson BT (2006) Caspase-1-dependent pore forma-
tion during pyroptosis leads to osmotic lysis of infected host mac-
rophages. Cell Microbiol 8:1812–1825. https:// doi. org/ 10. 1111/j.
1462- 5822. 2006. 00751.x
8. Fink SL, Cookson BT (2007) Pyroptosis and host cell death
responses during Salmonella infection. Cell Microbiol 9:2562–
2570. https:// doi. org/ 10. 1111/j. 1462- 5822. 2007. 01036.x
9. Shi J, Zhao Y, Wang K etal (2015) Cleavage of GSDMD by
inflammatory caspases determines pyroptotic cell death. Nature
526:660–665. https:// doi. org/ 10. 1038/ natur e15514
10. Tan Y, Sun R, Liu L etal (2021) Tumor suppressor DRD2 facili-
tates M1 macrophages and restricts NF-κB signaling to trigger
pyroptosis in breast cancer. Theranostics 11:5214–5231. https://
doi. org/ 10. 7150/ thno. 58322
11. Teng JF, Mei QB, Zhou XG etal (2020) Polyphyllin VI induces
caspase-1-mediated pyroptosis via the induction of ROS/NF-κB/
NLRP3/GSDMD signal axis in non-small cell lung cancer. Can-
cers (Basel). https:// doi. org/ 10. 3390/ cance rs120 10193
12. Wang Y, Yin B, Li D etal (2018) GSDME mediates caspase-
3-dependent pyroptosis in gastric cancer. Biochem Biophys Res
Commun 495:1418–1425. https:// doi. org/ 10. 1016/j. bbrc. 2017. 11.
156
13. Schneider KS, Groß CJ, Dreier RF etal (2017) The inflamma-
some drives GSDMD-independent secondary pyroptosis and IL-1
release in the absence of caspase-1 protease activity. Cell Rep
21:3846–3859. https:// doi. org/ 10. 1016/j. celrep. 2017. 12. 018
14. Shi J, Gao W, Shao F (2017) Pyroptosis: gasdermin-mediated
programmed necrotic cell death. Trends Biochem Sci 42:245–254.
https:// doi. org/ 10. 1016/j. tibs. 2016. 10. 004
15. Thi HTH, Hong S (2017) Inflammasome as a therapeutic target for
cancer prevention and treatment. J Cancer Prev 22:62–73. https://
doi. org/ 10. 15430/ jcp. 2017. 22.2. 62
16. Liston A, Masters SL (2017) Homeostasis-altering molecular
processes as mechanisms of inflammasome activation. Nat Rev
Immunol 17:208–214. https:// doi. org/ 10. 1038/ nri. 2016. 151
17. Strowig T, Henao-Mejia J, Elinav E etal (2012) Inflammasomes in
health and disease. Nature 481:278–286. https:// doi. org/ 10. 1038/
natur e10759
18. Rathinam VA, Vanaja SK, Fitzgerald KA (2012) Regulation of
inflammasome signaling. Nat Immunol 13:333–342. https:// doi.
org/ 10. 1038/ ni. 2237
19. Wu J, Fernandes-Alnemri T, Alnemri ES (2010) Involvement of
the AIM2, NLRC4, and NLRP3 inflammasomes in caspase-1 acti-
vation by Listeria monocytogenes. J Clin Immunol 30:693–702.
https:// doi. org/ 10. 1007/ s10875- 010- 9425-2
20. Yu P, Zhang X, Liu N etal (2021) Pyroptosis: mechanisms and
diseases. Signal Transduct Target Ther 6:128. https:// doi. org/ 10.
1038/ s41392- 021- 00507-5
21. Kayagaki N, Lee BL, Stowe IB etal (2019) IRF2 transcriptionally
induces GSDMD expression for pyroptosis. Sci Signal. https:// doi.
org/ 10. 1126/ scisi gnal. aax49 17
22. Lamkanfi M, Dixit VM (2014) Mechanisms and functions of
inflammasomes. Cell 157:1013–1022. https:// doi. org/ 10. 1016/j.
cell. 2014. 04. 007
23. Miao EA, Leaf IA, Treuting PM etal (2010) Caspase-1-induced
pyroptosis is an innate immune effector mechanism against intra-
cellular bacteria. Nat Immunol 11:1136–1142. https:// doi. org/ 10.
1038/ ni. 1960
24. Ramos-Junior ES, Morandini AC (2017) Gasdermin: a new player
to the inflammasome game. Biomed J 40:313–316. https:// doi. org/
10. 1016/j. bj. 2017. 10. 002
25. Chen X, He WT, Hu L etal (2016) Pyroptosis is driven by non-
selective gasdermin-D pore and its morphology is different from
MLKL channel-mediated necroptosis. Cell Res 26:1007–1020.
https:// doi. org/ 10. 1038/ cr. 2016. 100
26. Sborgi L, Rühl S, Mulvihill E etal (2016) GSDMD membrane
pore formation constitutes the mechanism of pyroptotic cell death.
Embo J 35:1766–1778. https:// doi. org/ 10. 15252/ embj. 20169 4696
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1522 International Urology and Nephrology (2024) 56:1515–1523
1 3
27. Kayagaki N, Warming S, Lamkanfi M etal (2011) Non-canonical
inflammasome activation targets caspase-11. Nature 479:117–121.
https:// doi. org/ 10. 1038/ natur e10558
28. Shi J, Zhao Y, Wang Y etal (2014) Inflammatory caspases are
innate immune receptors for intracellular LPS. Nature 514:187–
192. https:// doi. org/ 10. 1038/ natur e13683
29. Rühl S, Broz P (2015) Caspase-11 activates a canonical NLRP3
inflammasome by promoting K(+) efflux. Eur J Immunol
45:2927–2936. https:// doi. org/ 10. 1002/ eji. 20154 5772
30. Rogers C, Fernandes-Alnemri T, Mayes L etal (2017) Cleavage
of DFNA5 by caspase-3 during apoptosis mediates progression to
secondary necrotic/pyroptotic cell death. Nat Commun 8:14128.
https:// doi. org/ 10. 1038/ ncomm s14128
31. Wang Y, Gao W, Shi X etal (2017) Chemotherapy drugs induce
pyroptosis through caspase-3 cleavage of a gasdermin. Nature
547:99–103. https:// doi. org/ 10. 1038/ natur e22393
32. Orning P, Weng D, Starheim K etal (2018) Pathogen blockade
of TAK1 triggers caspase-8-dependent cleavage of gasdermin D
and cell death. Science 362:1064–1069. https:// doi. org/ 10. 1126/
scien ce. aau28 18
33. Sarhan J, Liu BC, Muendlein HI etal (2018) Caspase-8 induces
cleavage of gasdermin D to elicit pyroptosis during Yersinia infec-
tion. Proc Natl Acad Sci U S A 115:E10888-e10897. https:// doi.
org/ 10. 1073/ pnas. 18095 48115
34. Hou J, Zhao R, Xia W etal (2020) PD-L1-mediated gasdermin
C expression switches apoptosis to pyroptosis in cancer cells and
facilitates tumour necrosis. Nat Cell Biol 22:1264–1275. https://
doi. org/ 10. 1038/ s41556- 020- 0575-z
35. Zhou Z, He H, Wang K etal (2020) Granzyme A from cytotoxic
lymphocytes cleaves GSDMB to trigger pyroptosis in target cells.
Science. https:// doi. org/ 10. 1126/ scien ce. aaz75 48
36. Liu Y, Fang Y, Chen X etal (2020) Gasdermin E-mediated tar-
get cell pyroptosis by CAR T cells triggers cytokine release syn-
drome. Sci Immunol. https:// doi. org/ 10. 1126/ sciim munol. aax79
69
37. Zhang Z, Zhang Y, Xia S etal (2020) Gasdermin E suppresses
tumour growth by activating anti-tumour immunity. Nature
579:415–420. https:// doi. org/ 10. 1038/ s41586- 020- 2071-9
38. Daniels AM, Schulte AR, Herndon CM (2018) Interstitial cystitis:
an update on the disease process and treatment. J Pain Palliat Care
Pharmacother 32:49–58. https:// doi. org/ 10. 1080/ 15360 288. 2018.
14764 33
39. Tudrej KB, Piecha T, Kozłowska-Wojciechowska M (2019) Role
of NLRP3 inflammasome in the development of bladder pain syn-
drome interstitial cystitis. Ther Adv Urol 11:1756287218818030.
https:// doi. org/ 10. 1177/ 17562 87218 818030
40. Cui D, Liu S, Tang M etal (2020) Phloretin ameliorates hyper-
uricemia-induced chronic renal dysfunction through inhibiting
NLRP3 inflammasome and uric acid reabsorption. Phytomedicine
66:153111. https:// doi. org/ 10. 1016/j. phymed. 2019. 153111
41. Deng W, Yang Z, Yue H etal (2020) Disulfiram suppresses
NLRP3 inflammasome activation to treat peritoneal and gouty
inflammation. Free Radic Biol Med 152:8–17. https:// doi. org/ 10.
1016/j. freer adbio med. 2020. 03. 007
42. Wang X, Yin H, Fan L etal (2021) Shionone alleviates NLRP3
inflammasome mediated pyroptosis in interstitial cystitis injury.
Int Immunopharmacol 90:107132. https:// doi. org/ 10. 1016/j.
intimp. 2020. 107132
43. Wang X, Fan L, Yin H etal (2020) Protective effect of Aster tatari-
cus extract on NLRP3-mediated pyroptosis of bladder urothelial
cells. J Cell Mol Med 24:13336–13345. https:// doi. org/ 10. 1111/
jcmm. 15952
44. McVary KT (2006) BPH: epidemiology and comorbidities. Am J
Manag Care 12:S122-128
45. Kashyap M, Pore S, Wang Z etal (2015) Inflammasomes are
important mediators of prostatic inflammation associated
with BPH. J Inflamm (Lond) 12:37. https:// doi. org/ 10. 1186/
s12950- 015- 0082-3
46. Jiang MY, Han ZD, Li W etal (2017) Mitochondrion-associated
protein peroxiredoxin 3 promotes benign prostatic hyperplasia
through autophagy suppression and pyroptosis activation. Onco-
target 8:80295–80302. https:// doi. org/ 10. 18632/ oncot arget. 17927
47. Ponomareva L, Liu H, Duan X etal (2013) AIM2, an IFN-induc-
ible cytosolic DNA sensor, in the development of benign prostate
hyperplasia and prostate cancer. Mol Cancer Res 11:1193–1202.
https:// doi. org/ 10. 1158/ 1541- 7786. Mcr- 13- 0145
48. Ronco C, Bellomo R, Kellum JA (2019) Acute kidney injury.
Lancet 394:1949–1964. https:// doi. org/ 10. 1016/ s0140- 6736(19)
32563-2
49. Sun J, Ge X, Wang Y etal (2022) USF2 knockdown downregu-
lates THBS1 to inhibit the TGF-β signaling pathway and reduce
pyroptosis in sepsis-induced acute kidney injury. Pharmacol Res
176:105962. https:// doi. org/ 10. 1016/j. phrs. 2021. 105962
50. Miao N, Yin F, Xie H etal (2019) The cleavage of gasdermin D by
caspase-11 promotes tubular epithelial cell pyroptosis and urinary
IL-18 excretion in acute kidney injury. Kidney Int 96:1105–1120.
https:// doi. org/ 10. 1016/j. kint. 2019. 04. 035
51. Li Y, Wang J, Huang D etal (2022) Baicalin alleviates contrast-
induced acute kidney injury through ROS/NLRP3/Caspase-1/
GSDMD pathway-mediated proptosis invitro. Drug Des Devel
Ther 16:3353–3364. https:// doi. org/ 10. 2147/ dddt. S3796 29
52. Ying W, Riopel M, Bandyopadhyay G etal (2017) Adipose tissue
macrophage-derived exosomal miRNAs can modulate invivo and
invitro insulin sensitivity. Cell 171:372-384.e312. https:// doi. org/
10. 1016/j. cell. 2017. 08. 035
53. Zheng P, Luo Q, Wang W etal (2018) Tumor-associated mac-
rophages-derived exosomes promote the migration of gastric can-
cer cells by transfer of functional Apolipoprotein E. Cell Death
Dis 9:434. https:// doi. org/ 10. 1038/ s41419- 018- 0465-5
54. Xia W, Li Y, Wu M et al (2021) Gasdermin E deficiency
attenuates acute kidney injury by inhibiting pyroptosis and
inflammation. Cell Death Dis 12:139. https:// doi. org/ 10. 1038/
s41419- 021- 03431-2
55. Juan CX, Mao Y, Cao Q etal (2021) Exosome-mediated pyrop-
tosis of miR-93-TXNIP-NLRP3 leads to functional difference
between M1 and M2 macrophages in sepsis-induced acute kidney
injury. J Cell Mol Med 25:4786–4799. https:// doi. org/ 10. 1111/
jcmm. 16449
56. Liu Y, Minze LJ, Mumma L etal (2016) Mouse macrophage
polarity and ROCK1 activity depend on RhoA and non-apoptotic
Caspase 3. Exp Cell Res 341:225–236. https:// doi. org/ 10. 1016/j.
yexcr. 2016. 02. 004
57. Su D, Guan L, Gao Q etal (2017) ROCK1/p53/NOXA signaling
mediates cardiomyocyte apoptosis in response to high glucose
invitro and vivo. Biochim Biophys Acta Mol Basis Dis 1863:936–
946. https:// doi. org/ 10. 1016/j. bbadis. 2017. 01. 021
58. Wang QL, Xing W, Yu C etal (2021) ROCK1 regulates sepsis-
induced acute kidney injury via TLR2-mediated endoplasmic
reticulum stress/pyroptosis axis. Mol Immunol 138:99–109.
https:// doi. org/ 10. 1016/j. molimm. 2021. 07. 022
59. Papadopoulou-Marketou N, Paschou SA, Marketos N etal (2018)
Diabetic nephropathy in type 1 diabetes. Minerva Med 109:218–
228. https:// doi. org/ 10. 23736/ s0026- 4806. 17. 05496-9
60. Moreno JA, Gomez-Guerrero C, Mas S etal (2018) Targeting
inflammation in diabetic nephropathy: a tale of hope. Expert Opin
Investig Drugs 27:917–930. https:// doi. org/ 10. 1080/ 13543 784.
2018. 15383 52
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1523International Urology and Nephrology (2024) 56:1515–1523
1 3
61. An X, Zhang Y, Cao Y etal (2020) Punicalagin protects diabetic
nephropathy by inhibiting pyroptosis based on TXNIP/NLRP3
pathway. Nutrients. https:// doi. org/ 10. 3390/ nu120 51516
62. Ke R, Wang Y, Hong S etal (2020) Endoplasmic reticulum stress
related factor IRE1α regulates TXNIP/NLRP3-mediated pyrop-
tosis in diabetic nephropathy. Exp Cell Res 396:112293. https://
doi. org/ 10. 1016/j. yexcr. 2020. 112293
63. Li G, Liu C, Yang L etal (2023) Syringaresinol protects against
diabetic nephropathy by inhibiting pyroptosis via NRF2-mediated
antioxidant pathway. Cell Biol Toxicol. https:// doi. org/ 10. 1007/
s10565- 023- 09790-0
64. Li F, Chen Y, Li Y etal (2020) Geniposide alleviates diabetic
nephropathy of mice through AMPK/SIRT1/NF-κB pathway. Eur
J Pharmacol 886:173449. https:// doi. org/ 10. 1016/j. ejphar. 2020.
173449
65. Wang Y, Gou R, Yu L etal (2021) Activation of the NLRC4
inflammasome in renal tubular epithelial cell injury in diabetic
nephropathy. Exp Ther Med 22:814. https:// doi. org/ 10. 3892/ etm.
2021. 10246
66. Komada T, Chung H, Lau A etal (2018) Macrophage uptake of
necrotic cell DNA activates the AIM2 inflammasome to regu-
late a proinflammatory phenotype in CKD. J Am Soc Nephrol
29:1165–1181. https:// doi. org/ 10. 1681/ asn. 20170 80863
67. Cheng Q, Pan J, Zhou ZL etal (2021) Caspase-11/4 and gas-
dermin D-mediated pyroptosis contributes to podocyte injury in
mouse diabetic nephropathy. Acta Pharmacol Sin 42:954–963.
https:// doi. org/ 10. 1038/ s41401- 020- 00525-z
68. Dobruch J, Oszczudłowski M (2021) Bladder cancer: current chal-
lenges and future directions. Medicina (Kaunas). https:// doi. org/
10. 3390/ medic ina57 080749
69. He H, Yi L, Zhang B etal (2021) USP24-GSDMB complex pro-
motes bladder cancer proliferation via activation of the STAT3
pathway. Int J Biol Sci 17:2417–2429. https:// doi. org/ 10. 7150/
ijbs. 54442
70. Chen X, Chen H, Yao H etal (2021) Turning up the heat on non-
immunoreactive tumors: pyroptosis influences the tumor immune
microenvironment in bladder cancer. Oncogene 40:6381–6393.
https:// doi. org/ 10. 1038/ s41388- 021- 02024-9
71. El-Gamal R, Abdelrahim M, El-Sherbiny M etal (2022) Gas-
dermin D: a potential mediator and prognostic marker of bladder
cancer. Front Mol Biosci 9:972087. https:// doi. or g/ 10. 3 389/ fmolb.
2022. 972087
72. Peng J, Jiang H, Guo J etal (2020) CD147 expression is associated
with tumor proliferation in bladder cancer via GSDMD. Biomed
Res Int 2020:7638975. https:// doi. org/ 10. 1155/ 2020/ 76389 75
73. Nguyen-Nielsen M, Borre M (2016) Diagnostic and therapeu-
tic strategies for prostate cancer. Semin Nucl Med 46:484–490.
https:// doi. org/ 10. 1053/j. semnu clmed. 2016. 07. 002
74. Xu Z, Wang H, Qin Z etal (2021) NLRP3 inflammasome pro-
moted the malignant progression of prostate cancer via the acti-
vation of caspase-1. Cell Death Discov 7:399. https:// doi. org/ 10.
1038/ s41420- 021- 00766-9
75. Karan D, Tawfik O, Dubey S (2017) Expression analysis of
inflammasome sensors and implication of NLRP12 inflamma-
some in prostate cancer. Sci Rep 7:4378. https:// doi. org/ 10. 1038/
s41598- 017- 04286-4
76. Jain S, Dash P, Minz AP etal (2019) Lipopolysaccharide (LPS)
enhances prostate cancer metastasis potentially through NF-κB
activation and recurrent dexamethasone administration fails to
suppress it invivo. Prostate 79:168–182. https:// doi. org/ 10. 1002/
pros. 23722
77. Tang S, Lian X, Cheng H etal (2021) Bacterial lipopolysaccharide
augmented malignant transformation and promoted the stemness
in prostate cancer epithelial cells. J Inflamm Res 14:5849–5862.
https:// doi. org/ 10. 2147/ jir. S3329 43
78. Xing WY, Zhang ZH, Xu S etal (2020) Calcitriol inhibits lipopol-
ysaccharide-induced proliferation, migration and invasion of pros-
tate cancer cells through suppressing STAT3 signal activation. Int
Immunopharmacol 82:106346. https:// doi. org/ 10. 1016/j. intimp.
2020. 106346
79. Rini BI, Campbell SC, Escudier B (2009) Renal cell carcinoma.
Lancet 373:1119–1132. https:// doi. org/ 10. 1016/ s0140- 6736(09)
60229-4
80. Warren AY, Harrison D (2018) WHO/ISUP classification, grad-
ing and pathological staging of renal cell carcinoma: standards
and controversies. World J Urol 36:1913–1926. https:// doi. org/
10. 1007/ s00345- 018- 2447-8
81. Cui Y, Zhou Z, Chai Y etal (2021) Upregulated GSDMB in clear
cell renal cell carcinoma is associated with immune infiltrates and
poor prognosis. J Immunol Res 2021:7753553. https:// doi. org/ 10.
1155/ 2021/ 77535 53
82. Bobin-Dubigeon C, Chauvin A, Brillaud-Meflah V etal (2017)
Liver X receptor (LXR)-regulated genes of cholesterol trafficking
and breast cancer severity. Anticancer Res 37:5495–5498. https://
doi. org/ 10. 21873/ antic anres. 11979
83. Wang K, Xu T, Ruan H etal (2019) LXRα promotes cell metas-
tasis by regulating the NLRP3 inflammasome in renal cell
carcinoma. Cell Death Dis 10:159. https:// doi. org/ 10. 1038/
s41419- 019- 1345-3
84. Tan YF, Wang M, Chen ZY etal (2020) Inhibition of BRD4
prevents proliferation and epithelial-mesenchymal transition
in renal cell carcinoma via NLRP3 inflammasome-induced
pyroptosis. Cell Death Dis 11:239. https:// doi. org/ 10. 1038/
s41419- 020- 2431-2
85. Zhang Y, Chen X, Fu Q etal (2021) Comprehensive analysis
of pyroptosis regulators and tumor immune microenvironment in
clear cell renal cell carcinoma. Cancer Cell Int 21:667. https:// doi.
org/ 10. 1186/ s12935- 021- 02384-y
86. Zhang X, Wei X, Wang Y etal (2021) Pyroptosis regulators and
tumor microenvironment infiltration characterization in clear cell
renal cell carcinoma. Front Oncol 11:774279. https:// doi. org/ 10.
3389/ fonc. 2021. 774279
87. Tang X, Zhang A, Feng Y etal (2021) A novel pyroptosis-related
lncrnas signature for predicting the prognosis of kidney renal
clear cell carcinoma and its associations with immunity. J Oncol
2021:9997185. https:// doi. org/ 10. 1155/ 2021/ 99971 85
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The current study probed into how tumor cell-derived exosomes (Exos) mediated hsa_circ_0001739/lncRNA AC159540.1 to manipulate microRNA (miR)-218-5p/FTO-N6-methyladenosine (m6A)/MYC signal axis in liver metastasis in colorectal cancer (CRC). hsa_circ_0001739 and lncRNA AC159540.1 were identified as the upstream regulator of miR-218-5p using ENCORI and LncBase databases. Expression patterns of miR-218-5p, hsa_circ_0001739, lncRNA AC159540.1, FTO, and MYC were detected, accompanied by loss-and-gain-of function assays to examine their effects on CRC cell biological functions. SW480 cells-derived Exos were purified, followed by in vitro studies to uncover the effect of hsa_circ_0001739/lncRNA AC159540. miR-218-5p was downregulated while hsa_circ_0001739/lncRNA AC159540.1 was upregulated in CRC tissues and cells. Silencing of hsa_circ_0001739/lncRNA AC159540.1 restrained the malignant phenotypes of CRC cells. Exos-mediated hsa_circ_0001739/lncRNA AC159540.1 competitively inhibited miR-218-5p to elevate FTO and MYC. The inducing role of Exos-mediated hsa_circ_0001739/lncRNA AC159540.1 in CRC was also validated in vivo. Conclusively, Exos-mediated circ_0001739/lncRNA AC159540.1 regulatory network is critical for CRC, offering a theoretical basis for CRC treatment.
Article
Full-text available
Diabetic nephropathy (DN) is one of the serious complications of diabetes that has limited treatment options. As a lytic inflammatory cell death, pyroptosis plays an important role in the pathogenesis of DN. Syringaresinol (SYR) possesses anti-inflammatory and antioxidant properties. However, the therapeutic effects and the underlying mechanism of SYR in DN remain unclear. Herein, we showed that SYR treatment ameliorated renal hypertrophy, fibrosis, mesangial expansion, glomerular basement membrane thickening, and podocyte foot process effacement in streptozotocin (STZ)-induced diabetic mice. Mechanistically, SYR prevented the abundance of pyroptosis-related proteins such as NOD-like receptor family pyrin domain containing 3 (NLRP3), cysteinyl aspartate-specific proteinase 1 (Caspase-1), and gasdermin D (GSDMD), and the biosynthesis of inflammatory cytokines interleukin 1β (IL-1β) and interleukin 18 (IL-18). In addition, SYR promoted the nuclear translocation of nuclear factor E2-related factor 2 (NRF2) and enhanced the downstream antioxidant enzymes heme oxygenase 1 (HO-1) and manganese superoxide dismutase (MnSOD), thereby effectively decreasing excess reactive oxygen species (ROS). Most importantly, knockout of NRF2 abolished SYR-mediated renoprotection and anti-pyroptotic activities in NRF2-KO diabetic mice. Collectively, SYR inhibited the NLRP3/Caspase-1/GSDMD pyroptosis pathway by upregulating NRF2 signaling in DN. These findings suggested that SYR may be promising a therapeutic option for DN.
Article
Full-text available
Purpose To investigate the effect of baicalin on the reactive oxygen species (ROS)/ NOD-like receptor protein 3 (NLRP3)/Caspase-1/gasdermin-D (GSDMD) inflammasome pathway and its related mechanism in regulating pyroptosis of human renal tubular epithelial cells (HK-2) induced by contrast media. Methods Iohexol was used to act on HK-2 cells to establish a renal tubular cell pyroptosis model; and the signal pathway genes were silenced, cytokines were detected by enzyme-linked immunosorbent assay (ELISA), and cell viability, gene expression, and protein expression were evaluated by double fluorescence staining and flow cytometry. To assess the cytotoxicity caused by the contrast agent; cells were pretreated with different concentrations of baicalin; and then the cells were exposed to iohexol again, and the relevant indicators were tested again. Results After HK-2 cells were exposed to iohexol, the NLRP3 inflammasome pathway markers NLRP3, interleukin (IL)-1β, and IL-18 mRNA levels as well as the protein expression levels of NLRP3, ASC, Caspase-1, and GSDMD were up-regulated. In addition, the effect also significantly increased the IL-18, IL-1β, lactate dehydrogenase (LDH), superoxide dismutase (SOD), malondialdehyde (MDA) release, and cellular ROS levels. The results of Annexin V-FITC/PI flow cytometry showed that the level of apoptosis was increased. However, after the intervention of baicalin, the changes in the above indexes caused by iohexol stimulation of HK-2 cells were inhibited. Conclusion Exposure to iohexol can induce pyroptosis of HK-2 cells through the ROS/NLRP3/Caspase-1/GSDMD signaling pathway. Baicalin ameliorated iohexol-induced pyroptosis in HK-2 cells by regulating the NLRP3 inflammasome pathway.
Article
Full-text available
Background: Bladder cancer is considered one of the commonest widespread cancers, its presentation ranges from non-muscle invasive form to being muscle-invasive. The gasdermin family of proteins consists of six proteins. Members of gasdermin family are involved in pyroptosis; which is considered as type of inflammatory apoptosis via participation of gasdermin D and inflammatory caspases. Purpose: The goal of this research was to look into the potential involvement of gasdermin D in pathogenesis of bladder cancer, In addition, to investigate its potential role as a prognostic marker of bladder cancer. Methods: Gasdermin D gene and protein expression was examined in fresh frozen 80 bladder cancer specimens (30 NMIBC, and 50 MIBC) and the matching 80 control tissue samples utilizing real-time polymerase chain reaction and western blotting. Furthermore, the immunoreactivity of gasdermin D protein was also detected by immunohistochemistry. Results: Gasdermin D gene and protein expression showed a highly significant difference between the control and the two bladder cancer groups ( p < 0.001), as demonstrated by real-time PCR, western blotting and immunohistochemistry. Cox proportional hazards regression models showed that lower gasdermin D gene expression in cancer patients (≤1.58-fold), and younger age (≤53 years) were linked with a higher risk of local tumor recurrence. Moreover, higher gasdermin D gene expression (>2.18-fold), and lymph nodes’ involvement were associated with an increased mortality. Conclusion: Gasdermin D is involved in the pathogenesis of bladder cancer and muscle invasion, in addition, tissue gasdermin D expression may be used as useful tool to predict local tumor recurrence.
Article
Full-text available
Pyroptosis is a lytic and inflammatory type of programmed cell death that is usually triggered by inflammasomes and executed by gasdermin proteins. The main characteristics of pyroptosis are cell swelling, membrane perforation, and the release of cell contents. In normal physiology, pyroptosis plays a critical role in host defense against pathogen infection. However, excessive pyroptosis may cause immoderate and continuous inflammatory responses that involves in the occurrence of inflammatory diseases. Attractively, as immunogenic cell death, pyroptosis can serve as a new strategy for cancer elimination by inducing pyroptotic cell death and activating intensely antitumor immunity. To make good use of this double-edged sword, the molecular mechanisms, and therapeutic implications of pyroptosis in related diseases need to be fully elucidated. In this review, we first systematically summarize the signaling pathways of pyroptosis and then present the available evidences indicating the role of pyroptosis in inflammatory diseases and cancer. Based on this, we focus on the recent progress in strategies that inhibit pyroptosis for treatment of inflammatory diseases, and those that induce pyroptosis for cancer therapy. Overall, this should shed light on future directions and provide novel ideas for using pyroptosis as a powerful tool to fight inflammatory diseases and cancer.
Article
Full-text available
The crosstalk between macrophages and tubular epithelial cells (TECs) actively regulates the progression of renal fibrosis. In the present study, we revealed the significance of circular RNA ACTR2 (circACTR2) in regulating macrophage inflammation, epithelial–mesenchymal transition (EMT) of TECs, and the development of renal fibrosis. Our results showed UUO-induced renal fibrosis was associated with increased inflammation and EMT, hypertrophy of contralateral kidney, up-regulations of circACTR2 and NLRP3, and the down-regulation of miR-561. CircACTR2 sufficiently and essentially promoted the activation of NLRP3 inflammasome, pyroptosis, and inflammation in macrophages, and through paracrine effect, stimulated EMT and fibrosis of TECs. Mechanistically, circACTR2 sponged miR-561 and up-regulated NLRP3 expression level to induce the secretion of IL-1β. In TECs, IL-1β induced renal fibrosis via up-regulating fascin-1. Knocking down circACTR2 or elevating miR-561 potently alleviated renal fibrosis in vivo. In summary, circACTR2, by sponging miR-561, activated NLRP3 inflammasome, promoted macrophage inflammation, and stimulated macrophage-induced EMT and fibrosis of TECs. Knocking down circACTR2 and overexpressing miR-561 may, thus, benefit the treatment of renal fibrosis. Graphical abstract
Article
Full-text available
Background It is well known that chronic inflammation can promote the occurrence and progression of cancer. As a type of proinflammatory death, pyroptosis can recast a suitable microenvironment to promote tumor growth. However, the potential role of pyroptosis in clear cell renal cell carcinoma (ccRCC) remains unclear. Methods The transcriptome expression profile and mutation profile data of ccRCC with clinical characteristics included in this study were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Consensus clustering was used for clustering. Gene set enrichment analysis (GSEA) analysis were applied to evaluate the biological mechanisms. Single sample gene set enrichment analysis (ssGSEA) was applied for evaluating the proportion of various immune infiltrating cells. The ESTIMATE algorithm was involved to compute the immune microenvironment scores. Results Among the 17 pyroptosis regulators, a total of 15 pyroptosis regulators were differential expressed between tumor and normal tissues, in which 12 of them emerged strong correlations with prognoses. According to the pyroptosis components, the ccRCC patients were divided into four pyroptosis subtypes with different clinical, molecular, and pathway characteristics. Compared with other clusters, cluster B showed the pyroptosis heat phenotype, while cluster D represented the pyroptosis cold phenotype with poor overall survival. In addition, we performed principal component analysis (PCA) on the differential genes between clusters to construct the pyroptosis index. Furthermore, the pyroptosis index was significantly correlated with survival in different tumor mutation statuses and different grades and stages. Besides, the expression of pyroptosis-related regulators was related to the infiltration of immune cells and the expression of immune checkpoints, among which AIM2 was considered as the most significant immune-related pyroptosis regulator. Ultimately, we found that AIM2 was related to the immune activation pathway and was significantly overexpressed in tumor tissues. Conclusion This study revealed that pyroptosis regulators and pyroptosis index played an important role in the development and prognoses of ccRCC. Moreover, AIM2 can be used as a predictor of the response of immunotherapy. Assessing the pyroptosis patterns may help evaluate the tumor status and guide immunotherapy strategies.
Article
Full-text available
It is widely accepted that inflammation is an important risk for the development of prostate cancer (PCa). The objective of this study was designed to investigate the potential molecular mechanism of NLR family, pyrin domain-containing protein 3 (NLRP3) inflammasome in the malignant progression of PCa. The expression level of NLRP3 was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR) and fluorescence in situ hybridization. The effects of NLRP3 in the development of PCa by applying gain- and loss-of-function assays in LNCaP and PC3 cell lines were detected by CCK-8, TUNEL, and Transwell migration assays. The underlying mechanism of NLRP3 and caspase-1 in PCa was examined by the rescue experiments, western blotting, and qRT-PCR assays. In addition, the promoting effect of NLRP3 inflammasome was performed with an animal subcutaneous tumorigenesis experiment in vivo. The upregulation of NLRP3 was confirmed in PCa tissues and cell lines. Functionally, using CCK-8, TUNEL, and Transwell migration assays, these results showed that activation of NLRP3/caspase-1 inflammasome by LPS + ATP could enhance the ability of proliferation and migration; and decrease the apoptosis of LNCaP and PC3 cell lines. Western blotting assay showed that the activation of caspase-1 would increase after the stimulation of NLRP3 inflammasome by LPS + ATP. Moreover, the overexpression of NLRP3 promoted, while the knockdown of NLRP3 inhibited the malignant progression in PCa cell lines by positively regulating caspase-1. In addition, the rescue experiments revealed the association among NLRP3 and caspase-1, which showed that the overexpression vectors/inhibitors of caspase-1 could reverse the effect of knockdown/overexpression of NLRP3 in PCa cell lines in vitro. Finally, In in vivo experiment, the suppression of NLRP3 knockdown impaired tumor growth of PCa. Collectively, these results indicated that NLRP3 inflammasome played a vital role in promoting the malignant progression of PCa via the activation of caspase-1. Together, our findings provided insight into the mechanisms of NLRP3/caspase-1 inflammasome and revealed an alternative and potential target for the clinical diagnosis and treatment of PCa.
Article
Full-text available
Gasdermin B (GSDMB) is part of the gasdermin (GSDM) family, and they use varying means of domain interactions in molecules to adjust their pore-forming and lipid-binding actions. The GSDM family has roles in the regulation of cell differentiation and proliferation, particularly in the process of pyroptosis. Nonetheless, the correlation of GSDMB with immune infiltrates and its prognostic values in clear cell renal cell carcinoma (ccRCC) are still undefined. Therefore, we assessed the correlation of GSDMB with immune infiltrates and its prognostic role in ccRCC. The transcriptional expression profiles of GSDMB in ccRCC tissues in addition to normal tissues were retrieved from The Cancer Genome Atlas (TCGA) and additionally verified in a different independent cohort, which was obtained from the Gene Expression Omnibus (GEO) database. The Human Protein Atlas and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) were used to assess the protein expression of GSDMB. To assess the effectiveness of GSDMB in distinguishing ccRCC from normal samples, the receiver operating characteristic (ROC) curve analysis was performed. Relationships between GSDMB expression, clinicopathological variables, and overall survival (OS) were evaluated with multivariate methods as well as Kaplan-Meier survival curves. Protein-protein interaction (PPI) networks were created with STRING. Functional enrichment analyses were conducted by utilizing the “ClusterProfiler” package. The Tumor Immune Estimation Resource (TIMER) and tumor-immune system interaction database (TISIDB) were utilized to determine the association between the mRNA expression of GSDMB and immune infiltrates. GSDMB expression was significantly more upregulated in ccRCC tissues compared to surrounding normal tissues. An increase in the mRNA expression of GSDMB was related to the high pathologic stage and advanced TNM stage. The analysis of the ROC curve indicated that GSDMB had an AUC value of 0.820 to distinguish between ccRCC tissues and adjacent normal controls. Kaplan-Meier survival analysis indicated that ccRCC patients with high GSDMB had a poorer prognosis compared to those with low GSDMB (). Correlation analysis showed that the mRNA expression of GSDMB was associated with immune infiltrates and the purity of the tumor. Upregulation of GSDMB is significantly related to immune infiltrates and poor survival in ccRCC. The results of this study indicate that GSDMB could be regarded as a biomarker for the detection of poor prognosis and potential target of immune treatment in ccRCC. 1. Introduction Renal cell carcinoma’s (RCC) incidence has been growing on a global scale in the last few decenniums, and RCC has the highest mortality rate annually among urological carcinomas [1]. RCC is a heterogeneous type of carcinoma, of which the most common form is clear cell RCC (ccRCC) which makes up 75-80% of RCCs [2]. Due to the resistance to chemotherapy and radiotherapy, the current treatment of ccRCC patients is still unsatisfactory. Therefore, resecting the tumor is the most optimal choice as treatment of ccRCC patients, which is regarded as the sole type of treatment that could lead to complete curation [3]. Generally, the majority of ccRCC patients are diagnosed in an advanced stage, as a result of an occult onset and rapid progression [4]. Although targeted therapy has shown a positive effect on extending the duration of patients’ survival time, the drug resistance associated with long-term use was still a problem that has not been settled [5]. Immune therapy, in particular immune checkpoint inhibitors, is a type of treatment for ccRCC patients that is very promising [6]. However, not every patient can benefit from it since research has shown that the objective response rate to anti PD-L1 therapy is merely 20% approximately. The patients who did have a positive response to immune checkpoint inhibitors did not exhibit long-term remission [7]. The proliferation mechanism of ccRCC has a complex and multifactorial nature, consisting of an elaborate network of different genetic backgrounds and multiple carcinogens that result in changes in oncogenes or tumor suppressors [8]. Thus, it is a necessity to determine the molecular mechanisms that are related to the progression of ccRCC, which is valuable for diagnosis and treatment. A new kind of programmed cell death known as pyroptosis has vital functions in both immune defense and septic shock [9]. It is also known as programmed cell death mediated by gasdermin. It is known that the gasdermin (GSDM) family has different functions in the regulation of both cell proliferation and differentiation containing GSDMA, GSDMB, GSDMC, GSDMD, GSDME, and DFNB59 [10]. GSDMB and GSDMA genes are found in chromosome 17q2, and GSDMC and GSDMD can be found in chromosome 8q24 [10]. Except for DFNB59, the other family members of the gene share an approximate 45% of sequence homology; in addition, each GSDM has two domains that can bind one another and are attached via a long type of flexible linker [11]. With the exception of DFNB59, other known members of the GSDM family have comparable 3D structures as indicated by the sequence homology [12]. The gasdermin-N domain allows the majority of GSDM members to serve as a novel kind of pore-forming protein. While they are executing their function as pore-forming proteins, multiple GSDM family members may use varying processes of interactions between intramolecular domains that modify their pore-forming and lipid-binding actions, possibly inducing pyroptosis-like qualities in these cells. In GSDMB, one of the members of the GSDM family, pyroptosis-like features have also been observed, and several studies have suggested that overexpression of GSDMB exists in multiple types of carcinomas, in which it could be correlated with the progression of cancer and metastasis. However, the value of GSDMB in prognosis and its relation with immune infiltrates in ccRCC are yet to be completely elucidated. In this article, we downloaded data and evaluated the association between GSDMB expression, clinical data, and overall survival (OS) in patients with ccRCC by using the different databases TCGA, GEO, and Human Protein Atlas. Then, the TIMER and GEPIA databases were used in order to identify the correlation between GSDMB expression and immune cells that have infiltrated and their equivalent sets of gene markers. Besides, we used the STRING website to explore the GSDMB-interacted protein network. Results demonstrated that high GSDMB level was correlated with poor prognosis and related to an inadequate infiltration of immune cells in ccRCC. Hence, there is a strong possibility that GSDMB overexpression may undermine the antitumor effects of the immune system in ccRCC. 2. Method 2.1. The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) Database TCGA database (https://genome-cancer.ucsc.edu/) is a freely accessible data portal of a large-scale project on cancer genomes that consists of pathological as well as clinical data of over 30 different kinds of cancer and is intended for research. TCGA tool cancer browser was used to obtain data of ccRCC patients, including RNA-Seq expression and corresponding data on clinical pathology. The GEO database, an extensive gene expression library, can be found in the National Center of Biotechnology Information (NCBI) (https://www.ncbi.nlm.nih.gov/geo/). Since this database is available to the public by means of open access, authorization from the local ethics committee was not necessary. 2.2. The Human Protein Atlas (HPA), UALCAN, and Clinical Proteomic Tumor Analysis Consortium (CPTAC) The HPA consists of a large amount of information on proteomics and transcription of individual human samples containing tissue, cell, and a pathology atlas. The online database contains information of cell-specific locations for over 40 different healthy tissues as well as 20 most common categories of carcinomas. Furthermore, data on protein immunohistochemistry in human tumor tissues and normal tissues is also available on the HPA website. UALCAN (http://ualcan.path.uab.edu/) is a convenient and simple-to-use online resource that can be used for analyzing publicly available data on cancer. Using proteomics technologies, CPTAC (http://ualcan.path.uab.edu/analysis-prot.html) evaluates tumor biospecimens by mass spectrometry, which identifies and quantifies the characterizing proteome and constituent proteins of every tumor sample. In the present report, we used UALCAN to perform a throughput analysis of GSDMB protein expression obtained from CPTAC. 2.3. Univariate and Multivariate Logistic Regression Analyses In order to identify the impact of the expression of GSDMB in ccRCC patients, univariate Cox regression analysis was conducted to calculate the relation between GSDMB’s expression level and OS of patients across two different cohorts. Then, multivariate analysis was conducted to evaluate whether GSDMB is a distinct prognostic factor of survival in ccRCC patients. GSDMB was considered statistically significant in the Cox regression analysis when . 2.4. Protein-Protein Interaction (PPI) Networks and Functional Enrichment Analysis The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) website (https://string-db.org/) is another online tool. On this website, a large collection of integrating and consolidated PPI data is hosted. The PPI network information could be obtained after importing the GSDMB into STRING. A confidence score of >0.7 was regarded as significant. The “ClusterProfiler” package was used to perform Gene Ontology (GO) enrichment as well as Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of coexpression genes, which were visualized with the “ggplot2” package [13]. 2.5. Tumor Immune Estimation Resource (TIMER) Database TIMER (https://cistrome.shinyapps.io/timer/) is an extensive web-based resource that can be used for systematic analysis of immune infiltrates in various kinds of cancers. In the present study, we applied TIMER to establish the association among GSDMB’s expression in ccRCC and six different types of immune infiltrates (B cells, CD4-positive T cells, CD8-positive T cells, macrophages, neutrophils, and dendritic cells). 2.6. The Gene Expression Profiling Interactive Analysis (GEPIA) Analysis The GEPIA (http://gepia.cancer-pku.cn/index.html) is a database, which can be accessed online and is comprised of 8587 normal and 9736 tumors samples from GTEx and TCGA data. The database is dedicated to different types of analyses regarding the expression of RNA sequencing. We used it to analyze the association among GSDMB expression and various immune cell markers. The -axis in the graph represented the amount of GSDMB expression, while the -axis represented other type of genes of interest. Furthermore, TIMER data was used to verify which genes had a significant association with GSDMB expression as indicated by the GEPIA website. 2.7. Tumor-Immune System Interaction Database (TISIDB) TISIDB (http://cis.hku.hk/TISIDB/) is an integrated repository web portal, accessible online, for information on the correlation that exists between tumors and the innate immune system. In this article, we used the TISIDB to establish GSDMB expression and tumor-infiltrating lymphocytes (TILs) in cancers occurring in Homo sapiens. The relative abundance of TILs was deduced from the gene expression profile, which was derived through gene set variation analysis. Spearman’s test was conducted to quantify the associations between GSDMB and TILs. 2.8. Statistical Analyses All of the statistical analyses were conducted with R (V 3.6.3), and the R package ggplot2 was utilized to observe the differences in expression. The Mann-Whitney test and paired -test were conducted to establish the differences among ccRCC tissues and surrounding normal tissues. The pROC package was used to visualize the ROC curve, in which the cutoff value of GSDMB could be detected. To evaluate the effect of GSDMB on survival, log-rank and Kaplan-Meier tests were performed by using the survminer package. Correlation analysis was used by the Pearson correlation and Spearman test. 3. Result 3.1. Expression Pattern of GSDMB in Pan-Cancer Perspective The complete working set contained 33 types of cancer of which the mRNA expression pattern of GSDMB was evaluated. As shown in Figure 1, in comparison to normal tissues, GSDMB was significantly upregulated in 12 out of 33 cancer types and downregulated in 15 out of 33 cancer types. The data demonstrated that the mRNA expression of GSDMB was expressed in an abnormal way throughout different types of cancers.
Article
Full-text available
Background Increasing evidence has indicated that pyroptosis could regulate the tumor immune microenvironment (TIME) to affect the tumor development. As a highly immunogenic tumor, clear cell renal cell carcinoma (ccRCC) can benefit from immunotherapy, but related research on pyroptosis in the TIME of ccRCC is still deficient. Methods Available data derived from TCGA and GEO databases were analyzed to identify the different expression profiles of pyroptosis in ccRCC and normal tissues, and the correlation of pyroptosis regulators with TIME was evaluated in ccRCC. Results According to consensus clustering analysis, two differential expression levels of subtypes were identified to affect patient prognosis, and were related to histological tumor stage and grade. Immune cells were calculated by the CIBERSORT algorithm. Higher infiltrated levels of B cells naive, T cells CD4 memory resting, NK cells resting, monocytes, macrophages were observed in Cluster 1, while higher infiltrated levels of CD8 ⁺ T cells, T follicular helper cells, and Tregs were observed in Cluster 2. Gene set enrichment analysis indicated that Cluster 2 was enriched in multiple immune-related pathways, including the JAK-STAT signaling pathway. Moreover, overexpression of eight immune checkpoints was related to ccRCC development, especially in Cluster 2. As four potentially key pyroptosis regulators, AIM2, CASP5, NOD2, and GZMB were confirmed to be upregulated in ccRCC by RT-qPCR analysis and further verified by the HPA database. Further pan-cancer analysis suggested that these four pyroptosis regulators were differentially expressed and related to the TIME in multiple cancers. Conclusion The present study provided a comprehensive view of pyroptosis regulators in the TIME of ccRCC, which may provide potential value for immunotherapy.