You are on page 1of 18

Pharmacological Research 170 (2021) 105714

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Second-generation antipsychotics induce cardiotoxicity by disrupting


spliceosome signaling: Implications from proteomic and
transcriptomic analyses
Jing Wang a, 1, Xiaoqing Li a, 1, Zheng Liu a, Xinyi Lin a, Fan Zhong b, c, Shuhao Li a, Xinru Tang a,
Yang Zhang c, Liliang Li a, *
a
Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
b
Department of Systems Biology for Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
c
Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China

A R T I C L E I N F O A B S T R A C T

Keywords: Second-generation antipsychotics (SGAs) are first-line drugs that are prescribed for mental disorders in clinic.
Antipsychotics Severe cardiotoxicity has been widely reported and thus limits their clinical application. This study aimed to
Spliceosome identify the common mechanism underlying SGAs-induced cardiotoxicity using dual-omics analyses. Balb/C
Cardiotoxicity
mice were intraperitoneally injected with two representative SGAs, olanzapine (2.5 mg/kg) and clozapine (25
Proteome
mg/kg), at clinically comparable doses for 0, 7, 14 and 21 days. Our results showed that both SGAs induced
Transcriptome
cardiomyocyte degeneration, inflammation infiltration, and cardiac fibrosis, all of which worsened with time.
Chemical compounds studied in this article:
Olanzapine (Compound CID: 135398745)
Proteomic analysis revelaed that 22 differentially expressed (DE) proteins overlapped in olanzapine and
Clozapine (Compound CID: 135398737) clozapine-treated hearts. These proteins were significantly enriched in muscle contraction, amino acid meta­
Pladienolide B (Compound CID: 16202130) bolism and spliceosomal assembly by GO term analysis and spliceosome signaling was among the top enriched
2-Pyridylethylamine (Compound CID: 201148) pathways by KEGG analysis. Among the 22 DE proteins, three spliceosome signal proteins were validated in a
dynamic detection, and their expression significantly correlated with the extent of SGAs-induced cardiac fibrosis.
Following the spliceosome signaling dysregulation, RNA sequencing revealed that alternative splicing events in
the mouse hearts were markedly enhanced by SGAs treatments, and the production of vast transcript variants
resulted in dysregulation of multiple pathways that are critical for cardiomyocytes adaptation and cardiac
remodeling. Pladienolide B, a specific inhibitor of mRNA splicing, successfully corrected SGAs-induced alter­
native splicing and significantly attenuated the secretion of pro-inflammatory factors and cell deaths induced by
SGAs exposure. Our study concluded that the spliceosome signaling was a common pathway driving SGAs
cardiotoxicity. Pharmacological inhibition of the spliceosome signaling represents a novel therapeutic strategy
against SGAs cardiotoxicity.

1. Introduction major depressive disorders. The prescription of SGAs has increased more
than 18-fold in the past decades, helping 10 million people worldwide
Second-generation antipsychotics (SGAs) are the cornerstone of [1,2]. With increasingly wider clinical application, the toxic effects of
treating mental disorders such as schizophrenia, bipolar disorder and SGAs become a critical concern. These toxic effects range from minor

Abbreviations: AS, alternative splicing; A3SS, alternative 3′ splice site; A5SS, alternative 5′ splice site; BP, biological process; CC, cellular component; Clz, clo­
zapine; CK, creatine kinase; DE, differentially expressed; GO, gene ontology; HE, haematoxylin and eosin; IHC, immunohistochemistry; KEGG, Kyoto Encyclopedia of
Genes and Genomes; LD50, half lethal dose; LFQ, label-free quantification; MF, molecular function; MXE, mutually exclusive exons; Olz, olanzapine; PB, pladienolide
B; RI, retained intron; RNA-seq, RNA sequencing; RT-PCR, reverse transcription polymerase chain reaction; SE, skipped exon; SGA, second-generation antipsychotics;
snRNA, small nuclear RNAs; TEM, Transmission electron microscope; 2-PAE, 2-Pyridylethylamine.
* Correspondence to: Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, 131 Dongan Road, Shanghai 200032, China.
E-mail addresses: wangj16@fudan.edu.cn (J. Wang), lixq15@fudan.edu.cn (X. Li), liuz18@fudan.edu.cn (Z. Liu), linxy16@fudan.edu.cn (X. Lin), zonefan@163.
com (F. Zhong), shli15@fudan.edu.cn (S. Li), xrtang18@fudan.edu.cn (X. Tang), zhangyang@fudan.edu.cn (Y. Zhang), liliangli11@fudan.edu.cn (L. Li).
1
These authors contributed equally to this study

https://doi.org/10.1016/j.phrs.2021.105714
Received 29 January 2021; Received in revised form 18 May 2021; Accepted 2 June 2021
Available online 5 June 2021
1043-6618/© 2021 Elsevier Ltd. All rights reserved.
J. Wang et al. Pharmacological Research 170 (2021) 105714

issues (e.g., mild sedation or dry mouth) to troubling issues (e.g., weight 2. Material and methods
gain or metabolic disturbances) to even life threatening issues (e.g.,
cardiotoxicity or agranulocytosis) [3–6]. For example, cardiac electrical 2.1. Chemicals and reagents
activity can be altered by the application of antipsychotics, resulting in
QTc prolongation which increases the risk of torsades de pointes and Antipsychotic clz was purchased from Sigma-Aldrich (St. Louis, MO,
eventually sudden cardiac deaths [7]. Other serious cardiovascular USA). Clozapine was dissolved in 0.1 M HCl and pH balanced in PBS to
adverse effects include myocardial infarction, myocarditis and Brugada make a stock solution of clz (25 mg/mL, equal to 80 mM). The stock
syndrome phenotype [8]. The cardiotoxicity of SGAs strongly limits solution was then diluted on the basis of use dosage. Antipsychotic olz
their utility in clinic, even resulting in discontinuation of potentially was purchased from Selleck Chemicals (Houston, TX, USA) and prepared
successful regimens. as 5 mg/mL (equal to 16 mM) stock solution in solvents consisting of
Among SGAs, olanzapine (olz) and clozapine (clz) are the two most DMSO and PBS. The highest final concentration of DMSO in external
prescribed regimens in clinic. Unfortunately, both of them have been solution was ≤ 1%, a concentration that had no effect on the survival of
reported to associate with cardiotoxicity. According to a population- mice. Primary antibodies against Sf3b3 (Catalog: ab209402) and Snrpf
based study, current users of SGAs had higher rates of sudden cardiac (Catalog: ab154870) were purchased from Abcam (Cambridge, United
deaths than non-users, with an adjusted incidence-rate ratio of 2.26 [9]. Kingdom). A rabbit polyclonal antibody against Snrpd1 (Catalog:
SGAs enjoy a wide pharmacological profile including D2 dopaminergic, 10352-1-AP) was purchased from Proteintech Inc. (Rosemont, IL, USA).
5-HT1A, 5-HT2A, 5-HT2C serotonergic, and histaminergic receptors, all A mouse monoclonal antibody against β-actin (Catalog: sc-47778) was
of which are responsible for their pharmacological actions and adverse purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). A
effects [10]. Olanzapine, a first-line SGA, has been increasingly pre­ rabbit polyclonal antibody against HRH1 (Catalog: DF4980) was pur­
scribed for nausea and vomiting caused by cancer and chemotherapy. chased from Affinity Biosciences Inc. (Cincinnati, OH, USA). All the
Current literature has reported that olz can lead to QTc changes [11], primary antibodies were validated by commercial companies and re-
with the blockade of Human Ether-ago-go Related Gene (hERG) potas­ validated by us before use. HRP-linked secondary antibodies were pur­
sium channel being the predisposing factor [12]. Acute olz administra­ chased from Jackson laboratories Inc. (West Grove, PA, USA). Propi­
tion interfered with the cardiac energy metabolism and elevated dium Iodide (PI, Catalog: ST512) and 4′ ,6-diamidino-2-phenylindole
acetyl-CoA carboxylase phosphorylation and tissue ATP levels in iso­ (DAPI, Catalog: C1006) dyes were purchased from Beyotime Biotech­
lated rat hearts [13]. Though rarely presented, cases with dilated car­ nology (Nantong, Jiangsu, China). Mouse IL-1β (Catalog: BP-E93105)
diomyopathy after long-term exposure of olz have also been reported and mouse IL-18 (Catalog: BP-E00001) detection kits were commer­
[14]. Similar to olz in binding and functional profile [15,16], clz is the cially purchased from Boyun Biotechnology, Shanghai, China. The
most powerful antipsychotic in treating refractory schizophrenia [17, specific inhibitor of spliceosome signaling (pladienolide B, PB) was a
18]. Over two thirds of the patients who fail to respond to other anti­ generous gift from Dr. Yongbo Wang at the Department of Cellular and
psychotics may benefit from clz [19], making clz another potent Genetic Medicine, School of Basic Medical Sciences, Fudan University at
regimen. However, the high risk of cardiotoxicity is preventing clz from a stock solution of 1 mM. A specific agonist of HRH1 (2-Pyridylethyl­
becoming a first-line treatment [20]. We have demonstrated that clz is amine, 2-PAE, Catalog: GC11001) was purchased from GLPBio (Mon­
the most likely drug to trigger sudden cardiac death among all anti­ tclair, CA, USA) and prepared as 10 mM stock solution in deionized
psychotics [21]. Analog to olz, clz also blocks the hERG potassium water.
channel, leading to torsadogenic cardiotoxicity [22]. Myocarditis and
cardiomyopathy may also occur after clz administration, potentially 2.2. Animal study
leading to heart failure [23]. Therefore, olz and clz are two powerful
remedies with similar chemical structures and reported cardiotoxicity. All experimental procedures were conducted with the protocols for
Although the toxicological effects of olz and clz in hearts have been animal experiments in compliance with the regulations of the Institu­
reported [13,24,25], these studies were largely clinical reports. The tional Animal Care Committee at the School of Basic Medical Sciences,
cardiotoxic mechanisms of olz and clz have yet to be fully elucidated. To Fudan University (Approval No.: 20170223-004). The sample size for
the best of our knowledge, previous studies investigating the car­ animal studies was set as ≥ 5 unless otherwise stated. Specific pathogen-
diotoxicity of SGAs focused on single drug at a time or were mainly free (SPF) male Balb/C mice (approximately 6 weeks old at delivery)
observations of the consequences after cardiotoxicity [26]. Based on the were purchased from Shanghai Laboratory Animal Center (Shanghai,
cardiopathology (cardiomyopathy and myocarditis), arrhythmogenic China). The animals were group-housed in cages and acclimated to the
changes, and similar chemical structure shared by these antipsychotics new environment for one week. A 12-h light and dark cycle and
[15], we presumed that olz and clz share common molecular mecha­ continuous food and water supply were offered.
nisms when inducing cardiotoxicity. The shared ground for the car­ To mimic human therapeutic regimens, a daily dose of 2.5 mg/kg
diotoxicity by both drugs, however, are still largely unexplored [27], body weight of olz or 25 mg/kg body weight of clz were intraperito­
despite the notion that histamine 1 receptor (HRH1) and histamine 3 neally administered for 0, 7, 14 and 21 days, respectively. Mice were
receptor (HRH3) are believed to mediate SGAs-induced weight gain and randomly distributed into the 4 time periods (n = 5–7 mice per drug per
cardiometabolic disturbance [28,29]. Thereby, the potential toxicolog­ time period). Mice weights were recorded on a daily basis before each
ical effects shared by the representative SGAs and the causal mecha­ injection. Mice were sacrificed at each indicated time period by cervical
nisms accounting for their cardiotoxicity remain unknown. dislocation after euthanasia using 2.5% isoflurane. Hearts were trans­
In the present study, we performed dual-omics analyses integrating versely dissected and part of the heart tissue was fixed in formalin and
proteomics and transcriptomics to obtain systemic information on the embedded in paraffin for sectioning and histology. The remaining heart
common molecular alterations in representative SGAs (olz and clz)- tissues were stored at − 80 ℃ for subsequent analysis.
exposed mouse hearts. We aimed to uncover the primary mechanism
underlying cardiac toxicity shared by olz and clz which may aid in 2.3. Histological examination
providing venues for developing wide-spectrum cardioprotectants. Our
results revealed signatures of spliceosome signaling that underlined the Heart tissues were fixed in a 10% neutral formalin solution,
common causal mechanisms of SGAs cardiotoxicity. This study, to the embedded in paraffin, sectioned at a thickness of 4 µm, and stained with
best of our knowledge, represents the first to depict the molecular re­ haematoxylin and eosin (HE) to examine inflammatory infiltrates.
sponses to chronic SGAs exposure in the mouse heart. Fibrotic tissues within the hearts were examined by Masson’s trichrome
staining. After Masson’s trichrome staining, five fields were randomly

2
J. Wang et al. Pharmacological Research 170 (2021) 105714

selected for each slide and the area of fibrosis was measured by Image J monitored on a daily basis. On each monitored day, an aliquot of 10 μL
software (National Institutes of Health, Bethesda, MD, USA). The CCK-8 solution (Beyotime, Nantong, China) was added into the culture
percent of fibrosis in each slide was then averaged over the 5 fields. medium. Cells were then re-incubated at 37 ℃ for additional 2 h before
measurement of the optical density at a wavelength of 450 nm on a
2.4. Immunohistochemistry (IHC) analysis microplate reader (BioTek, Winooski, VT, USA). All assays were inde­
pendently repeated for three times.
IHC staining protocols were in accordance with our previous publi­
cation [5]. Briefly, slides were antigen-retrieved by heating in a micro­ 2.8. Label-free proteome analysis
wave at 100 ℃ for 10 min in 0.1 M citric acid buffer (pH= 6.0) and were
then rinsed in 3% hydrogen peroxidase for 10 min. After that, slides At the time of sacrifice, the mouse heart tissues after 21-day drug
were incubated with corresponding antibodies at 4 ℃ overnight. After treatment (n = 3/group) were individually homogenized in lysis buffer
secondary antibody incubation at room temperature for 1 h, the (4% SDS, 0.1 M DTT, 100 mM Tris/HCl, pH7.6) and boiled for 15 min.
immunoreactivity was developed in 0.05% diaminobenzidine. Haema­ Label-free proteome analysis was performed at the Shanghai Applied
toxylin was then co-stained for 3 min to visualize nuclei. For the Protein Technology Co. Ltd. (Shanghai, China). Protease and phospha­
quantification of protein positivity in nuclei, five fields were randomly tase inhibitors were used to limit potential protein degradation. The
selected for each slide and the nucleus with brown signal was counted as lysed protein samples were then quantified using a BCA kit (Thermo
positive expression. Then the protein-positive rate was calculated as the Scientific, CA, USA). Filter-aided sample preparation (FASP) was used to
number of positive nuclei in proportion to the total number of nuclei prepare the protein samples with mild modification. Briefly, each pro­
within the whole-scale field. The results from the five fields were then tein sample was mixed with 200 μL of urea (8 M) in 150 mM Tris-HCl
averaged. (pH 8.0) (UA buffer) and centrifuged at 14,000 g for 15 min at 20 ℃
to remove SDS. A second washing step with UA buffer was taken to
2.5. Determination of creatine kinase (CK) levels and pro-inflammatory exchange the remaining SDS with urea. The proteins were alkylated with
factor contents 100 μL of 50 mM iodoacetamide for 39 min at room temperature in the
dark, followed by repeated washing with 100 μL of UA buffer and 100 μL
To determine the activity of a myocardial injury marker CK, cell aliquots of 50 mM NH4HCO3. Then, trypsin was added to proteins at a
supernatants were collected from 5 independent assays for detection substrate/enzyme ratio of 50:1 (w/w) at 37 ℃ overnight in a thermo­
using a diagnostic kit (Catalog: YB-CK-Mu, YuBo Bioengineering Insti­ mixer (Eppendorf, Mittelsachsen, Saxony, Germany). After digestion,
tute, Shanghai, China). The detection protocol was in accordance with the peptides were eluted and collected by centrifugation. The acidified
the manufacturers’ instructions. The absorbance at a wavelength of 450 tryptic peptides were sampled on a SC001 traps 20 mm × 150 µm
nm was measured spectrophotometrically by a microplate spectropho­ reverse phase C18 trap column and then separated on a SC200 100 mm
tometer (BioTek Epoch, VT, USA) to calculate CK levels. To detect the × 150 µm reverse phase C18 nanocolumn (Easy-nLC1000 system,
contents of pro-inflammatory factors IL-1β and IL-18, cell supernatants Thermo Scientific) for 120 min 0.1% formic acid in 2% acetonitrile and
were collected after 24-hour exposure to SGAs and subject to enzyme- 0.1% formic acid in 84% acetonitrile was used for mobile phases A and
linked immunosorbent assays (ELISA) using commercial kits. The pro­ B, respectively, with the flow rate set at 300 nL/min. Orbitrap Q Exac­
tocol for each ELISA test was in accordance with the manufacturers’ tive mass spectrometry (Thermo Scientific) was coupled to the liquid
instructions. chromatography system (Thermo Scientific). The range of the precursor
ion in the MS scan was from 300 to 1800 m/z. After a full scan, the top 20
2.6. Transmission electron microscope (TEM) MS/MS events were acquired. Then, MaxQuant (Ver. 1.5.3.17) was used
for analyzing all MS/MS data from the mouse heart samples. Main
Balb/C mice were exposed to saline (ctrl), olz (2.5 mg/kg) or clz (25 search was set to 6 ppm, MS/MS tolerance was set to 20 ppm, and two
mg/kg) for 21 days. Immediately after sacrifice, the heart tissues were missed trypsin cleavages were allowed. Cysteine carbamidomethylation
immersed in 2.5% glutaraldehyde for overnight fixation at 4 ℃. Then and methionine oxidation were specified as the fixed and variable
samples were postfixed in 2% osmium tetraoxide, dehydrated through modifications, respectively. The false discovery rate of the protein level
graded concentrations of ethanol, infiltrated and embedded in Epon 618 and peptide level were set to 0.05. Label-free quantification (LFQ) in­
at 60 ℃ for 48 h. Ultrathin sections were cut with a diamond knife, tensity was performed in MaxQuant, as described previously [33].
mounted on formvar-coated slot grids, and subsequently stained with Proteins identified more than twice in the three biological replicates
1% uranyl acetate and lead citrate. The images were taken with a CM- were defined as quantifiable proteins. The LFQ intensity of each protein
120 model FEI/PHILIPS TEM (Philips Electron Optics B.V., Eindhoven, was then statistically compared between olz and ctrl groups, or between
The Netherlands). clz and ctrl groups using Microsoft Excel 2016 software (Microsoft,
Seatle, WA, USA). In our study, proteins with p values of less than 0.05
2.7. Cell viability assessment and fold differences greater than 1.5 were classified as significantly
differential proteins. The proteins that were undetectable in specific
To assess the effects of olz and clz on mouse cardiomyocyte viability, group but quantifiable in their counterparts were considered as specif­
a cardiac-derived HL-1 cell line (Millipore, MA, USA, Cat# SCC065; ically expressed proteins.
RRID: CVCL_0303) was seeded into 96-well plates in quadruplicates. The Both the significantly differential proteins and specific proteins were
HL-1 cells were derived from atrial cardiac muscle cells obtained from termed as differentially expressed (DE) proteins which were clustered
transgenic mice [30]. Previous studies indicate that HL-1 cells retain for bioinformatical analysis. Briefly, Gene Ontology (GO) and annota­
phenotypic characteristics of the adult cardiomyocyte, exhibit adult tion for proteins were performed using Blast2GO (Version 2.8.0) (https:
cardiomyocyte-like gene expression profiles, possess intercalated discs, //www.blast2go.com/), while the Kyoto Encyclopedia of Genes and
maintain the ability to contract, retain differentiated cardiac morpho­ Genomes (KEGG) analysis was performed using KEGG Automatic
logical, electro-physiological characteristics and display the pharmaco­ Annotation Server (KAAS). All proteins were matched to the Swiss-Prot
logical properties of primary cardiac myocytes [31,32]. Therefore, the database (http://www.ebi.ac.uk/swissprot/). p value of pathway
cells were utilized and treated with either DMSO (ctrl), olz (64 μM), or enrichment was calculated using the following formula:
clz (20 μM) for consecutive days. To assess the effects of a HRH1 agonist
on SGAs-induced cellular toxicity, HL-1 cells were primed with 2-PAE at
the indicated dose 1 h prior to SGA treatments. Cell viability was

3
J. Wang et al. Pharmacological Research 170 (2021) 105714

( )( )
M N− M spliceosome protein expression, HL-1 cells were primed with indicated

m− 1
i n− i doses of 2-PAE 1 h prior to SGA treatments. Protein was quantified by a
p = 1− ( )
N BCA kit (Thermo Scientific, CA, USA) according to the manufacturer’s
i=0
n protocol. An equal amount of 20 ng protein was loaded on a 12% SDS-
PAGE gel and transferred onto a polyvinylidene fluoride membrane.
N is the number of all identified proteins that can be connected with The blots were blocked by 5% skim milk for 1 h at room temperature and
GO or KEGG Pathway analysis information. incubated with primary antibodies at 4 ℃ overnight. The primary an­
n is the number of differential proteins in N. tibodies were used at a final dilution of 1:1000 with tris-buffered saline
M is the number of proteins that can be connected with a certain GO containing 0.1% Tween-80. Membranes were then incubated with cor­
term or pathway. responding secondary antibodies for 1 h at room temperature. The
m is the number of differential proteins with certain GO term or immunoreactivity was detected using an enhanced chemiluminescent
KEGG pathway. autoradiography kit (Amersham, Pittsburgh, PA, USA). β-actin was
If a p value was less than 0.05, this GO term or pathway was regarded synchronously developed as an internal control.
as a significant enrichment.
2.12. Reverse transcription polymerase chain reaction (RT-PCR)
2.9. Protein-protein interaction analysis
Total RNA was extracted from the mouse cardiac HL-1 cells using the
The protein-protein interaction network analysis was performed by Trizol solution (Vazyme Biotechnology, Nanjing, China) according to
using the STRING tool (version 11.0) (https://string-db.org/) to identify the manufacturer’s instructions. An equal amount of total RNA (500 ng)
interactions between the DE proteins shared by olz-treated and clz- was reverse-transcribed and amplified using the RT-PCR kit (Vazyme)
treated groups. according to the manufacturer’s instructions. The specificity of primers
was assessed using the NCBI BLAST tool (https://blast.ncbi.nlm.nih.
2.10. Transcriptome analysis gov/Blast.cgi) and validated by electrophoresis. Primer sequences are
listed as follows:
Total RNAs were isolated from the hearts of ctrl, olz, and clz at the Npr2, forward 5′ -CAAGAGAATGGGCAGCCCTA-3′ .
21-day treatment groups (n = 3/group) using Trizol solution (Vazyme, reverse 5′ -TACTTGGGTATGAGTGGGAGGT-3′ ;.
Nanjing, China) following the manufacturer’s instructions. The purity Ubp1, forward 5′ - TCAACAGGCCGTGGAAATGAA-3′ ,
was assessed by an Agilent 2100 Bioanalyzer using the RNA 6000 Nano reverse 5′ - GATGCACAATTTGAGCCTGGT-3′ .
Chip (Agilent Technologies, Santa Clara, CA, USA). RNA quantification PCR products were separated by electrophoresis using 2% agarose
was performed using Thermo scientific Multiskan GO microplate reader gels containing nucleic acid red stain (Beyotime).
with μDrop™ Plate (Waltham, MA, USA).
To construct the RNA-seq library, 2 μg of total RNA per sample was 2.13. Assessment of cell death rate
exposed to poly-T oligo attached magnetic beads to isolate poly-A mRNA
following mRNA fragmentation. The cleaved RNA fragments were Mouse cardiac HL-1 cells were seeded on the coverslips in a 24-well
transcribed into double-stranded cDNAs which were then purified using plate and treated with DMSO (ctrl), olz, or clz with or without PB co-
AMPure XP beads to remove all reaction components. Reads containing treatment in distinct concentrations (0.1 nM, 1 nM and 10 nM). After
ploy-N, reads containing adapters, and low-quality reads from the raw 48 h incubation, PI dye was added to the culture medium, and cells were
data were removed to obtain clean high-quality data for subsequent further incubated for 30 min under 37 ℃. The culture medium was then
analysis. replaced by washing three times with cold PBS, and cells were fixed with
Transcript quantification of RNA-seq reads was performed with pure acetone for 10 min, followed by co-staining with DAPI for 10 min.
Genomic Alignments (ver.1.20.1) by reads aligned to Ensemble Mus The coverslips were then mounted and photographed under a fluores­
musculus transcriptome annotation (GRCm.38. p5). The FPKM values cence microscope (Zeiss, Oberkochen, Germany). Five fields were
were calculated using ‘fpkm’ function from DESeq2 (ver. 1.24.0) that randomly imaged for each group. Thereafter, the PI-positive cell
were processed with the robust median ratio method and transcript numbers and DAPI-positive cell numbers were counted, respectively.
reads were normalized by the ‘voom’ function from Limma (ver. 3.40.6). The cell death rate (%) was defined as the PI-positive cell numbers in
To assess whether a transcript was differentially expressed, EdgeR (ver. proportion to the DAPI-positive cell numbers.
3.24.3) calculates the results based on the normalized counts from entire
sequence alignments. Significantly differential transcripts were defined 2.14. Statistical analysis
as a fold change of raw FPKM value > 2 and adjusted p value < 0.01 and
specifically expressed transcripts were defined as the transcripts that Data are presented as mean ± standard error of mean (SEM). A
were undetectable in specific group but quantifiable in their counter­ Student’s t-test was used for comparisons between 2 groups. A one-way
parts. Both the significantly differential transcripts and specific tran­ or two-way analysis of variance (ANOVA), when necessary, was used to
scripts were pooled as DE transcripts which were clustered for GO term compare 3 or more groups, followed by Bonferroni post-hoc tests. A
and KEGG pathway enrichment analyses on the free online MajorBio i- linear regression analysis was performed to analyze the correlation of
Sanger Cloud Platform (www.i-sanger.com). The cutoff p value of protein expression with extents of cardiac fibrosis. Unless elsewhere
pathway enrichment was set as 0.05 and the formula for calculating stated, all statistical analysis was performed with the GraphPad Prism 8
pathway enrichment p values was as stated in the Label-free proteomics. (GraphPad Software Inc., San Diego, CA, USA). A p value of less than
0.05 was considered statistically significant.
2.11. Western blot analysis
3. Results
To validate the results of proteomic analysis, total proteins were
extracted from SGAs-exposed cardiomyocytes with RIPA buffer (Beyo­ 3.1. Second-generation antipsychotics (SGAs) induced chronic
time, Nantong, China) mixed with protease/phosphatase inhibitor cardiotoxicity in mice
cocktail (Cell Signal Technology, Boston, MA, USA). Briefly, the cardio-
derived HL-1 cells were treated with olz or clz for 0 h, 2 h, 4 h, 8 h, 12 h To mimic the clinical scenario, the maintenance doses of olz and clz
and 24 h before harvest. To assess the effects of a HRH1 agonist on used in clinic were converted to appropriate doses in mice. Therefore,

4
J. Wang et al. Pharmacological Research 170 (2021) 105714

mice were treated with the clinically comparable doses of olz (2.5 mg/ they consistently inhibited cell proliferation completely since day 3
kg) and clz (25 mg/kg) for respective 0, 7, 14 and 21 days. The histo­ onwards (Fig. 1e). Cell supernatant from HL-1 cells were collected on the
logical analyses (HE staining) showed that, compared to the untreated 48 h to test levels of the myocardial injury marker CK in different groups
control groups, the continuous olz and clz treatments resulted in sig­ (Fig. 1f). After olz or clz treatment, CK levels in antipsychotic groups
nificant inflammatory infiltration in a time-dependent manner (Fig. 1a, were significantly higher than that of the control group. We further
Fig. 1b). Besides inflammation, antipsychotic-induced fibrosis was also assessed the ultrastructure of cardiomyocytes in heart sections using
detected in both perivascular and interstitial areas by Masson’s tri­ transmission electron microscopy (Fig. 1g). It was observed that olz or
chrome staining (Fig. 1a, Fig. 1b). Quantification of the fibrotic area clz-treated hearts consistently presented with swollen cytoplasm, dis­
showed that it increased with prolonged duration of olz (Fig. 1c) and clz arrayed myofibril, and disrupted intercalated disc, which were in great
administration (Fig. 1d). In the in vitro cell viability assays, it was found contrast with control heart. Moreover, both olz and clz-treated car­
that both olz and clz dose-dependently inhibited cell proliferation with diomyocytes displayed sparse vacuoles (asterisks, Fig. 1g), electron-
the calculated LD50 being 83.437 μM for olz (Supplementary Fig. 1a) lucent spaces (arrowheads, Fig. 1g), and accumulation of autophagic
and being 26.660 μM for clz (Supplementary Fig. 1b). Furthermore, vacuoles (arrows, Fig. 1g) in the cytoplasm. Altogether, these results
when olz and clz were added at a dose proximal to their respective LD50, confirmed that both olz and clz have a time-dependent cardiotoxic effect

Fig. 1. Time effects of SGAs cardiotoxicity. a, b Mice were treated with olz at 2.5 mg/kg or clz at 25 mg/kg for 0, 7, 14 and 21 consecutive days. Then mouse hearts
were harvested for haematoxylin and eosin (HE) staining and Masson’s trichrome staining. Magnification: 400 × . c, d The percentage of fibrotic lesion was
calculated by Image J software, n = 7/group. e Mouse cardiac HL-1 cells were incubated with olz (64 μM) or clz (20 μM) for serial days (1, 2, 3, 4, and 5 days) and
cell viability was determined using a CCK-8 kit. Each value represented the average of quadruplicates. f The supernatants from mouse cardiac HL-1 cells under ctrl
(dmso) or antipsychotic treatments were collected after 48 h′ exposure and were determined of the myocardial injury marker creatine kinase (CK) activity, n = 5/
group. g Transmission electron microscopy (TEM) revealed that cardiomyocytes from the SGAs-treated mice hearts displayed degeneration and myofibril disarray.
Asterisks indicate vacuoles in the cytoplasm. Arrowheads indicate electron-lucent spaces. Arrows indicate accumulation of autophagic vacuoles at various stages of
autophagy flux. *p < 0.05, **p < 0.01, ***p < 0.001 vs. Ctrl. Ctrl, control. CK, creatine kinase.

5
J. Wang et al. Pharmacological Research 170 (2021) 105714

and these SGAs shared common pathological changes, indicating po­ Table 1
tential common molecular basis behind their cardiotoxicity. Summary of differentially expressed (DE) proteins by proteomic analysis.
Comparisons Upregulated proteins Downregulated proteins Total
3.2. Proteomic profiling of the SGAs-exposed hearts Significant Specific Significant Specific

olz vs. ctrl 10 21 21 62 114


Since the continuous administration for 21 days was sufficient to clz vs. ctrl 17 32 13 28 90
induce cardiotoxicity, heart tissues at this time point were used for Overlapped 1 7 1 11 22
subsequent dual-omics analyses to better explore the molecular alter­ Olz, olanzapine. Clz, clozapine. Ctrl, control (Saline).
ations during SGAs exposure. In the proteomic profiling, LC-ESI-QqTOF
MS/MS identified a total of 2217, 2190, and 2215 proteins in the saline
line with the proteomic data, the HRH1 was observed to be
(ctrl), olz-exposed and clz-exposed mouse hearts, respectively. The de­
membrane-positive at mouse hearts from control, but remarkably
tails of comparative proteomics were documented in the Supplementary
decreased after different days of SGAs exposure (Supplementary
Data 1. In the olz-exposed group, compared to the control group, 114
Fig. 2a). The time-dependent decrease of HRH1 was also identified in
differentially expressed (DE) proteins were detected, out of which 31
the western blotting analysis (Supplementary Fig. 2b), further rein­
were up-regulated and 83 were down-regulated. In the DE proteins
forcing the reliability of our proteomic data. Thereafter, to better un­
induced by the clz treatment, there were 49 up-regulated proteins and
derstand the molecular mechanism shared by olz/clz-induced
41 down-regulated proteins (Fig. 2a). Some of the DE proteins were
cardiotoxicity, we focused on the overlapped DE proteins by olz and clz
quantifiable with certified fold-changes as compared with the control
treatments. Twenty-two protein (10 up-regulated and 12
group; others were detectable in only one group (control or olz/clz-
down-regulated proteins) were found to be shared by both drug treat­
exposed group), which were defined as specifically expressed proteins
ments (Table 1 and Fig. 2d). Details of the 22 proteins were documented
(Table 1). Volcano plots showed the global distribution of proteins in olz
in Table 2.
vs. ctrl and clz vs. ctrl comparisons (Fig. 2b). A hierarchical clustering
heat-map further depicted these significantly altered proteins in detail
(Fig. 2c). Specifically, HRH1, a histamine receptor reported to associate 3.3. Spliceosome signaling was significantly enriched in the proteomic
with antipsychotics-induced metabolic disorders [34,35], was also analysis
identified in the mouse hearts of control group (Supplementary Data 1),
suggesting the reliability of our proteomic data. Of note, this receptor Based on the 22 DE proteins, GO and KEGG pathway analyses were
was undetectable in clz-treated mouse hearts and only minimally conducted. The cellular roles of identified proteins were classified in
expressed in one olz-treated mouse heart (Supplementary Data 1). In terms of cellular component (CC), biological process (BP) and molecular

Fig. 2. Proteomic profiling of mouse hearts after 21-day SGA treatments. a 114 and 90 differentially expressed (DE) proteins were detected in olz and clz-exposed
mouse hearts, respectively. b Volcano plot depicted the DE proteins in olz or clz-treated group as compared to the control. Red and green dots represented significant
upregulation and downregulation, respectively (fold change ≥1.5 and p value <0.05). c Hierarchically clustered heatmap of proteins. d Interactive analysis of the
proteomic changes after olz and clz exposure. Twenty-two proteins overlapped by olz and clz treatments.

6
J. Wang et al. Pharmacological Research 170 (2021) 105714

function (MF). As shown in Fig. 3 and Supplementary Data 2, the

1.57 (0.042)

0.63 (0.011)
enriched cellular components were mainly U12-type spliceosomal
olz vs. ctrl complex, precatalytic spliceosome, U2-type precatalyctic spliceosome,
U2-type spliceosomal complex and U1 snRNP, all of which are closely

NA
NA

NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
Fold-change (p value)

related to activities of RNA splicing. Changes in biological processes


highlighted the spliceosomal snRNP assembly process and fundamental

1.67 (0.0049)
1.63 (0.0066)
processes such as muscle contraction and metabolic/catabolic processes.

1.56 (0.001)
0.60 (0.018)
clz vs. ctrl

Changes in molecular function were annotated to the activity of de­


hydrogenases and transaminases, which are fundamental adaptive re­

NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA

sponses to drug toxicity. KEGG analysis was performed to determine the


significantly enriched biological pathways in the 22 overlapped DE
proteins. Consistent with the GO analysis, spliceosome signaling was the

28,770.33 ± 4429.10
most enriched pathway (Fig. 4a and Supplementary Data 3). Other

272.19 ± 58.72

500.70 ± 79.78
240.66 ± 67.61
312.18 ± 91.42

160.49 ± 10.37
enriched pathways were beta-alanine metabolism, vitamin B6 meta­

286.39 ± 45.6
176.80 ± 6.58

244.07 ± 2.98
bolism, valine, leucine and isoleucine degradation, and ascorbate and
olz-treated

aldarate metabolism (Fig. 4a and Supplementary Data 3). A map of the


spliceosome pathway was shown in Fig. 4b, where the expression levels

ND
ND
ND
ND
ND
ND
ND
ND
ND
ND
ND
ND
ND

of several proteins that play core roles in RNA splicing were altered
31,051.33 ± 12,676.53 under SGAs exposure. Moreover, three of the 22 DE proteins (Sf3b3,
Snrpf, and Snrpd1) were from the spliceosome signaling and they were
shown to play hub roles in linking other proteins as per protein-protein
407.48 ± 166.35
469.74 ± 191.77
483.48 ± 197.38
187.79 ± 12.81

242.07 ± 13.84
285.97 ± 57.17
305.15 ± 51.83
300.21 ± 41.64
218.43 ± 3.28
213.23 ± 9.08

interaction (PPI) network analysis using the STRING tool (https://strin


LFQ intensity (Mean ± SEM, ×105)

clz-treated

g-db.org/) (Fig. 5). These data indicated that SGAs exposure disrupted
the RNA splicing signaling in mouse heart.
ND
ND
ND
ND
ND
ND
ND
ND
ND
ND
ND

3.4. Expression validation of the spliceosome signal proteins and their


pathological relevance
18,615 ± 2032.54
844.32 ± 222.29

250.48 ± 15.81

797.95 ± 48.76
229.71 ± 14.98
246.34 ± 96.75
120.73 ± 33.08
116.57 ± 34.46
210.75 ± 72.79
269.56 ± 12.86
233.25 ± 59.88

300.84 ± 2.65
191.74 ± 21.9
135.46 ± 2.93

To validate the spliceosome signal proteins, we detected the


88.7 ± 2.92

expression of these proteins in situ on heart sections by IHC staining.


Dynamic detection showed that Sf3b3 and Snrpf were maximally nuclei-
ctrl

ND
ND
ND
ND
ND
ND
ND

resided in drug-free heart but gradually decreased in olz or clz-treated


hearts (Fig. 6a). Unlike Sf3b3 and Snrpf, Snrpd1 was barely detected
Swiss-Prot ID

in drug-free hearts but increased dramatically while extending drug


Details of the 22 differentially expressed (DE) proteins that overlapped in olz and clz-treated mouse hearts.

exposure periods (Fig. 6a). Indeed, quantification of the IHC staining


Q9DCG9
Q921M3

Q9D1R1
Q9CZR2

Q9DBF1

Q9CR63

Q921U8
Q9CR26
Q91YR9

Q9D958

Q9ER88
Q8K274

Q6Y685

Q91XF0

Q6P1F6
O09110
O35127

P61922

P09528
P62315
P62307
P97379

showed that the nuclei-positive rates of Sf3b3 dropped from 6% in SGAs-


free hearts to approximate 3% after 21 days’ SGAs exposure (Fig. 6b,
upper panels). Similarly, the nuclei-positive rates of Snrpf decreased
ND, not detectable. NA, not applicable. LFQ, label-free quantification. Ctrl, control (Saline).
Tmem126b

linearly from 9% to approximate 3% by the treatment end, making up of


Trmt112
Aldh7a1

Ppp2r2a
Naalad2

Map2k3

over 60% drop after 21 days’ SGAs treatments (Fig. 6b, middle panels).
Symbol

Snrpd1
Fn3krp
Grcc10

G3bp2

Cox16
Tacc1
Sf3b3

Spcs1
Ptgr1
Snrpf

Dap3
Smtn
Pnpo

Abat
Vta1

Fth1

Snrpd1, the elevated spliceosome signal protein in the proteomic


profiling, was indeed increased by continuous SGAs treatments, partic­
ularly on the time window of 21 days (Fig. 6b, lower panels). To further
Serine/threonine-protein phosphatase2A 55 kDa regulatory subunit B alpha isoform

validate the expression dynamics of the spliceosome signal proteins, we


cultured HL-1 cells (a mouse cardiac-derived cell line) with extended
SGAs exposure hours. Western blot analysis showed that Sf3b3 and Snrpf
time-dependently decreased while Snrpd1 increased after 24-hour
exposure (Fig. 6c). Quantification of the blot intensity further sup­
ported that all of the three spliceosome signal proteins significantly
Multifunctional methyltransferase subunit TRM112-like protein

altered in response to SGAs treatments (Fig. 6d). These in vivo and in


Vacuolar protein sorting-associated protein VTA1 homolog

Dual specificity mitogen-activated protein kinase kinase 3

vitro data validated the expression of spliceosome signal proteins.


Cytochrome c oxidase assembly protein COX16 homolog
Complex I assembly factor TMEM126B, mitochondrial

To evaluate whether the spliceosome signal dysregulation was


Transforming acidic coiled-coil-containing protein 1

4-aminobutyrate aminotransferase, mitochondrial

receptor-dependent, we used a specific agonist 2-PAE to re-activate


Alpha-aminoadipic semialdehyde dehydrogenase
Ras GTPase-activating protein-binding protein 2

N-acetylated-alpha-linked acidic dipeptidase 2

HRH1. Our results showed that, when 2-PAE was pre-treated with HL-
28S ribosomal protein S29, mitochondrial

1 cells, the antipsychotic-induced decrease of Sf3b3 was successfully


Small nuclear ribonucleoprotein Sm D1

blunted (Supplementary Fig. 3a). However, the pre-agonizing of HRH1


Signal peptidase complex subunit 1

failed to rescue olz or clz-disturbed Snrpf and Snrpd1 expression, even at


Small nuclear ribonucleoprotein F

Pyridoxine-5-phosphate oxidase

higher doses of 2-PAE (Supplementary Fig. 3a). Consistently, olz or clz-


Splicing factor 3B subunit 3

inhibited cell viability was significantly recovered in early periods of 2-


Prostaglandin reductase 1

PAE pretreatment (289% increase, olz+2-PAE 100 μM vs. olz on day 3;


Ketosamine-3-kinase

Ferritin heavy chain

228% increase, clz+2-PAE 100 μM vs. clz on day 3). 2-PAE, again, failed
to maintain a continuous promotion on cell viability at even high doses,
Protein name

evidenced by the less minimal recovery of cell viability on day 4 (only


Protein C10

Smoothelin

148% increase, olz+2-PAE 100 μM vs. olz; only 110% increase, clz+2-
Table 2

PAE 100 μM vs. clz) when comparing with day 3 (Supplementary


Fig. 3b). These data implied that SGAs might dysregulate the

7
J. Wang et al. Pharmacological Research 170 (2021) 105714

Fig. 3. Gene ontology (GO) analysis of the 22 overlapped proteins. The color intensity of bar charts corresponded to the statistical significance of the GO Term. BP,
biological process. MF, molecular function. CC, cellular component.

spliceosome signaling in a HRH1 receptor-independent manner. obtained (Supplementary Data 4). A total of 69,734 transcripts were
To further study the spliceosome signal relevance with cardiac pa­ identified, including 52,795, 53,495, and 54,428 transcripts for ctrl, olz,
thology, we quantified the extent of cardiac fibrosis after 7, 14, and 21 and clz groups, respectively. The principal component analysis (PCA)
days’ SGAs exposure and correlated the cardiac fibrosis rate with the showed that there was a large variance between control and
spliceosome protein expression (Fig. 6e). In the olz treatments, it was antipsychotic-treated groups (Fig. 7a). Volcano plots were constructed
shown that the expression of Sf3b3, Snrpf, and Snrpd1 correlated line­ by integrating the p value and fold change of each transcript (p <0.01
arly with the fibrosis rate (goodness of fit: R = 0.5726 and p val­ and fold change ≥2), to filter the DE transcripts for the olz and clz
ue = 0.0054 for Sf3b3, R = 0.6180 and p = 0.0022 for Snrpf, exposure groups compared to the control group (Figs. 7b and 7c). A total
R = 0.5061 and p = 0.0162 for Snrpd1, respectively, Fig. 6e, upper number of 26,152 transcripts were differentially expressed in olz groups
panels). Similar results were also observed in the clz treatments, where as compared with the control hearts, of which 13,869 were up-regulated
Sf3b3 and Snrpf negatively correlated and Snrpd1 positively correlated and 12,283 were down-regulated (Fig. 7b and Supplementary Data 4).
with the extent of cardiac fibrosis (goodness of fit: R = 0.4286 and p Similarly, a total of 21,985 transcripts were differentially expressed in
value = 0.0466 for Sf3b3, R=0.4357 and p = 0.0427 for Snrpf, the clz vs. ctrl comparisons, of which 11,768 were upregulated and
R = 0.7056 and p = 0.0002 for Snrpd1, respectively, Fig. 6e, lower 10,217 were down-regulated (Fig. 7c and Supplementary Data 4). After
panels). Altogether, these results suggested that SGAs exposure interactive analysis, a total of 12,124 DE transcripts overlapped by olz
disturbed the spliceosome signaling. and clz groups which made up of 46.4% of the total DE transcripts after
olz treatment, and 55.1% of the DE transcripts after clz treatments
(Fig. 7d).
3.5. Alternative splicing events were markedly enhanced in the SGAs- After analysis of the AS events, it turned out that five major types of
exposed hearts AS events, termed retained intron (RI), skipped exon (SE), alternative 5′
splice site (A5SS), alternative 3′ splice site (A3SS), and mutually
To assess whether spliceosome signaling dysregulation results in exclusive exons (MXE) were all observed after olz or clz treatments
biological outcomes, we performed RNA sequencing (RNA-seq) to (Fig. 8a). Skipped exon (SE) was the most common AS event in both
analyze alternative splicing (AS) events and transcript variants, two treatments (Figs. 8a and 8b), and substantial retained intron (RI) events
indices lying directly downstream of the spliceosome signaling. Total were also observed for both treatments (Figs. 8a and 8b). In olz-treated
RNAs were isolated from saline (ctrl), olz, and clz-treated hearts. After hearts, 14,720 (67.36%) SE events and 2069 (9.47%) RI events were
filtering the RNA-seq data, a comprehensive list of transcripts was

8
J. Wang et al. Pharmacological Research 170 (2021) 105714

Fig. 4. Spliceosome was significantly enriched by the KEGG pathway analysis. a KEGG pathway analysis of the 22 overlapped proteins was performed and the top
KEGG pathways were shown. The p value of each enriched pathway was reflected by the intensity of bar color. b A map of the spliceosome. Red boxes represent the
altered molecules in the pathway. Sm, short for Sm-D1 (or Snrpd1).

observed (Fig. 8b). Similar to the olz-exposed group, 19,214 (70.36%) of multiple transcript variants that consistently altered by both olz and
SE events and 2092 (7.66%) RI events were detected after clz exposure clz treatments (Fig. 8d). Natriuretic peptide receptor 2 (Npr2), a critical
(Fig. 8b). Interestingly, 8 of the 22 DE proteins (36.4%) from the pro­ gene for cardiac hypertrophic growth and cardiac remodeling [38],
teomic screening were detected to have transcript variants and partic­ displayed intron retain after SGAs treatment (Fig. 8e). To investigate the
ularly, to show high proportions of novel AS after either olz or clz dose and time effects of AS events, Npr2 and upstream binding protein 1
treatments (Fig. 8c). In particular, multiple functional genes were (Ubp1), a gene controlling nuclear pre-mRNA maturation [39], were
observed to be alternatively spliced. Malate dehydrogenase 1 (Mdh1), amplified using primers targeting the retained introns (Figs. 8f and 8g).
an enzyme in the citric acid cycle whose activity may increase during At the increasing SGAs doses, it was observed that the intron inbetween
myocardial injury [36], and lysine (K)-specific methyltransferase 2B the exons 10 and 11 of Npr2 was retained dose-dependently. The intron
(Kmt2b), an enzyme involved in mRNA production [37], were detected between exons 5 and 6 of Ubp1 was barely detectable in drug-free

9
J. Wang et al. Pharmacological Research 170 (2021) 105714

Fig. 5. Protein-protein interactions network of the 22 proteins overlapped by olz- and clz-treated mouse hearts using the STRING tool (https://string-db.org/). The
three spliceosome signal proteins were boxed in red for highlight.

cardiomyocytes but was intensively retained after SGAs exposure Supplementary Data 6). The maladaptive pathways such as neuroactive
(Fig. 8f). With different time windows, it was also observed that the RI ligand-receptor interaction, cytokine-cytokine receptor interaction,
events of Npr2 and Ubp1 were markedly enhanced at extended hours of linoleic acid metabolism, steroid hormone biosynthesis, and cAMP
SGAs treatments (Fig. 8g). signaling were also enriched (Fig. 9b and Supplementary Data 6), mir­
Subsequently, the 12,124 DE transcripts that shared by both SGAs roring the cell fate toward pathological cardiac remodeling. Taken
treatments were subject to bioinformatics analysis. The GO analysis of together, the transcriptomic analysis showed direct evidence of
biological processes showed significant overrepresentation of tran­ enhanced alterative splicing in response to SGAs-induced dysregulation
scripts attributed to ribosome biogenesis, ribonucleoprotein complex of spliceosome signaling.
assembly, and cell differentiation. The molecular function analysis
showed overrepresentation of transcripts attributed to transmembrane
3.6. Pharmacological blockade of spliceosome pathway rescued SGAs-
transporter activity, cytoskeletal protein binding, and RNA binding.
induced cardiotoxicity
Ribosome and other subcellular components such as the plasma mem­
brane, extracellular region, and nucleus were also significantly enriched
In view that dysregulated spliceosome signaling resulted in vast
in the cellular component analysis (Fig. 9a and Supplementary Data 5).
alternative splicing and consequently led to multiple biological path­
In addition, KEGG pathway analysis showed that RNA transport, mRNA
ways imbalance, we next blocked the spliceosome pathway using a
surveillance, and spliceosome were significantly enriched (Fig. 9b and
specific pharmacologic inhibitor pladienolide B (PB) to assess whether

10
J. Wang et al. Pharmacological Research 170 (2021) 105714

Fig. 6. Expression validation of the spli­


ceosome signal proteins in mouse hearts
and cardiac cells. a Dynamic detection of
Sf3b3, Snrpf, and Snrpd1 in mouse heart
sections using immunohistochemistry
(IHC) staining. b Quantitative analysis of
the IHC staining results, n = 5–6/group. c
Representative western blotting analysis of
Sf3b3, Snrpf, and Snrpd1 in a cardiac cell
line (HL-1 cells) under olz or clz treat­
ments with distinct hours. d Quantitative
analysis of the western blotting analysis,
n = 3. e Cardiac fibrosis was quantified
and the extent of fibrosis was correlated
with the expression levels of the three
spliceosome signal proteins (Sf3b3, Snrpf,
and Snrpd1) at different drug exposure
time windows. Black dots represent mouse
hearts receiving saline treatments (n = 6);
Red, green, and blue dots represent hearts
receiving olz or clz treatments for 7 days
(n = 6), 14 days (n = 5), and 21 days
(n = 5), respectively. The linear correla­
tion coefficiency was plotted. For consis­
tency, we used positive numbers of R value
for all the regression analysis, while it
should be the negative number when
yielding negative correlations. ns, not sig­
nificant; *p < 0.05, **p < 0.01,
***p < 0.001 as indicated.

11
J. Wang et al. Pharmacological Research 170 (2021) 105714

Fig. 7. Transcriptome profiling of mouse hearts after 21-day antipsychotic treatments. a Principle component analysis of RNA-seq data. The ctrl, olz, and clz
treatment groups were marked as indicated. b, c Volcano plot showed differentially expressed (DE) transcripts in olz vs. ctrl and clz vs. ctrl comparisons (p<0.01 and
fold change ≥2). Red and blue dots represent significantly up-regulated and down-regulated transcripts, respectively. The number of DE transcripts, including
significantly differential transcripts (left) and specific transcripts (right), were denoted in the corresponding quadrants. d Overlapping transcripts after olz and clz
exposure. A total of 12,124 DE transcripts were overlapped by both drugs.

the SGAs-induced cellular toxicity could be rescued. At a molecular exposure to olz (Fig. 10c) or clz (Fig. 10d) displayed marked PI uptake,
level, we observed that the retained intron of Npr2 or Ubp1 by SGAs an indicator of cytomembrane rupture and cell death. However, when
exposure was corrected with PB co-treatment, particularly at a relatively cells were co-treated with PB, the PI uptake induced by SGAs exposure
higher dose of PB (10 nM) (Fig. 10a). Further, we detected the pro- markedly attenuated (Figs. 10c and 10d). Cell death rates, calculated as
inflammatory factors in a mouse cardiac HL-1 cell line. It was found the PI-ingested cells in proportion to the DAPI-stained cells, reached
that both SGAs significantly promoted the secretion of IL-18 and IL-1β approximate 50% for both SGAs, but dose-dependently declined with PB
(Fig. 10b), a supportive evidence of the inflammatory infiltration after co-treatments, particularly at a dose of 10 nM (Fig. 10e). These data
SGAs exposure in mouse hearts. More importantly, with the co- suggested that spliceosome signaling was the causal mechanisms of both
treatment of PB ranging from 0.1 nM to 10 nM, the contents of IL-18 SGAs cardiotoxicity. Spliceosome signaling has functional significance
and IL-1β significantly declined as compared with SGA single treat­ with regard to SGAs cardiotoxicity.
ment. In particular, when PB was co-treated at 10 nM, the olz or clz-
induced production of pro-inflammatory factors dramatically dropped 4. Discussion
to levels comparable to the control group (Fig. 10b). Moreover, at the
cellular levels, it was observed that mouse cardiomyocytes after As classical SGAs, olz and clz are commonly prescribed drugs for

12
J. Wang et al. Pharmacological Research 170 (2021) 105714

Fig. 8. Alternative splicing (AS) in the


heart was enhanced by SGAs exposure.
a Schematic illustration of the five
types of AS events. The number of AS
events in each comparison was anno­
tated at the right side. b Frequencies of
different AS events were shown. c
Novel AS events for the 22 differen­
tially expressed (DE) proteins that
were identified from the proteomic
analysis were tabulated. d Effects of
SGAs exposure on the splicing of
Kmt2b and Mdh1, two representative
genes controlling pathophysiological
processes. Red line: significantly up-
regulated transcripts in olz/clz-
exposed groups; blue line: signifi­
cantly down-regulated transcripts in
olz/clz-treated groups; black line: no
significant changes. e A schematic di­
agram of RI event of Npr2 in SGAs-
treated mouse hearts. f, g Assessment
of the dose and time effects of SGAs
exposure on the intron retain of Npr2
and Ubp1.

13
J. Wang et al. Pharmacological Research 170 (2021) 105714

Fig. 9. Bioinformatics analysis of the differentially expressed (DE) transcripts revealed biological impact by SGAs exposure. a Enriched GO terms of the 12,124 DE
transcripts shared by olz and clz were depicted. Multiple critical biological processes, molecular functions, and cellular components were disrupted. b Enriched KEGG
pathways of the 12,124 transcripts shared by olz and clz were depicted. Multiple pathways that control cell adaptation and cardiac remodeling were imbalanced.

mental disorders. Pathologically, antipsychotic-induced cardiomyopa­ clozapine [43]. It was also reported that during treatment windows of 7,
thy presented with cell hypertrophy, myocardial inflammatory infiltra­ 14, and 28 days, antipsychotic-induced cardiotoxicity was significantly
tion, and fibrotic remodeling. With long term or high dose exposure, higher among those with short-term use [44]. These reports indicated
myocardial remodeling and fibrosis can be detected, clinically diag­ the time effect of SGA cardiotoxicity. Consistent with previous clinical
nosed as myocarditis and cardiac hypertrophy [40–42]. The onset of reports, our data showed that both olz and clz induced time-dependent
severe cardiotoxicity may develop as early as 8 days after starting inflammatory infiltration and cardiac fibrosis, as well as increases in the

14
J. Wang et al. Pharmacological Research 170 (2021) 105714

Fig. 10. Pharmacological blockade of the spliceosome pathway rescued SGAs-induced cardiotoxicity. a Effects of pladienolide B (PB, a specific mRNA splicing
inhibitor) on SGAs-induced Npr2 and Ubp1 alternative splicing (intron retain) were assessed using RT-PCR. b Effects of pladienolide B on SGAs-induced secretion of
pro-inflammatory factors were assessed using ELISA kits targeting IL-18 and IL-1β in cell supernatants, n = 3/group. c, d Immunofluorescence analysis of the PI
uptake in mouse cardiac cells under SGAs exposure with or without PB co-treatments. The dye PI is used for visualization of dead cells that only ingested PI when
membrane ruptured. DAPI was co-stained to reveal nuclei. Magnification: 400× . e Quantification of cell death rates in the immunofluorescence assays, n = 5/group.
*p < 0.05, **p < 0.01, ***p < 0.001 as indicated.

15
J. Wang et al. Pharmacological Research 170 (2021) 105714

CK activity of the SGAs-insulted cardiac cells. At the ultrastructure Interestingly, among the wide range of SGAs-targeted pharmaco­
levels, both olz and clz treatment led to cytoplasmic edema, autopha­ logical receptors, HRH1 was the only identified receptor in the present
gosome and autolysosome formation, and rupture of intercalated discs cardiac proteomic study. This might be due to technical limitation of the
as evidenced by electron microscopy. This comparative evidence not proteomic profiling as it only identifies proteins of considerable abun­
only suggested the time-dependent cardiotoxic effects of SGAs, but also dance. HRH1, a receptor believed to be responsible for SGAs-induced
implicated there might be shared mechanisms underlying SGAs metabolic disturbance [28], was indeed decreased by olz and clz treat­
cardiotoxicity. ments in validations. However, the selective agonist of HRH1, 2-PAE,
To our knowledge, most of the previous studies focused on single only rescued the antipsychotics-dampened Sf3b3 expression, leaving
antipsychotic toxicity and those reports were mainly observations of the Snrpf and Snrpd1 expression unchanged even at its higher doses. Olz or
consequences after cardiotoxicity. The causal mechanisms accounting clz-inhibited cell viability was also only partially recovered by 2-PAE at
for the cardiotoxicity of clz and olz remain to be elucidated. Therefore, early periods, suggesting that SGAs dysregulated the spliceosome
the present study aimed to probe the common causal mechanisms that signaling in a H1 receptor-independent manner. Further genetic ap­
drive SGAs cardiotoxicity. To this end, olz and clz, two SGAs that share proaches that edit the Hrh1 gene expression might aid in supporting our
similar chemical structures and receptor affinity, were selected as pharmacological results. Another possible explanation might be that
representative SGAs. To better mimic clinical scenario, the clinical aside from the H1 receptor, other pharmacological targets, such as
comparable dose of each drug was adopted when establishing mouse histamine H3 receptor which protects hearts by promoting the expres­
models. After dual-omics analyses, our study provided multiple lines of sion of anti-inflammatory genes in renal-heart syndrome [52] and other
evidence that the spliceosome signaling is a previously unappreciated potential G protein-coupled receptors (i.e. cannabinoid CB1 and CB2
mechanism that underlined the similar cardiotoxicity induced by the receptors that oppositely regulated clz and quetiapine cardiotoxicity [5,
representative SGAs. In the proteomic analysis, RNA splicing was among 41]) may involve in the regulation of the spliceosome signal proteins.
the top enriched pathways in mouse hearts following olz or clz exposure. Investigation of other potential receptors would deserve further study.
Sf3b3, Snrpf, and Snrpd1, the three spliceosome signal proteins identi­ The identification of the spliceosome signaling as a common driver of
fied by proteomic profiling, were validated to be time-dependently SGAs cardiotoxicity is of paramount importance. First, the mechanisms
altered by olz and clz exposure in mouse hearts and in the cultured of SGAs-induced life-threatening cardiac effects remain largely un­
cardiomyocytes. Importantly, the alteration of these proteins signifi­ known. A recent report demonstrated that acute olz administration
cantly correlated with the extent of cardiac fibrosis, a supportive evi­ interfered with the cardiac energy metabolism and elevated acetyl-CoA
dence of the biological importance of the spliceosome in SGAs carboxylase phosphorylation and tissue ATP levels [13]. The car­
cardiotoxicity. As a direct evidence of spliceosome pathway dysregula­ diotoxicity of clz was associated with an increase in myocardial oxida­
tion, RNA sequencing showed that a vast of AS events occurred. As a tive stress, inflammatory cytokines, DNA damage and apoptosis with
consequence, various transcript variants yielded and thereby disrupting attenuation in antioxidant defenses [40,42,53]. Elevated catechol­
multiple pathways that are critical for maintaining cell adaptation and amines were also responsible for cardiac damage after clz exposure [54].
cardiac remodeling. Particular examples of the unwelcome splicing were These reports, however, were mainly observations of the consequences
the Npr2 and Ubp1 genes, which control cardiac remodeling [38] and of drug toxicity. As we have provided solid data that blockade of the
pre-mRNA maturation [39] under physiological conditions but were spliceosome signaling rescued SGAs toxicity in multiple perspectives,
incorrectly spliced by olz and clz exposure in a dose and time-dependent including the incorrect splicing of critical genes, secretion of
manner. The involvement of spliceosome signaling in SGAs cardiotox­ pro-inflammatory factors, and the cell deaths, we believe the spliceo­
icity was further reinforced when PB, a specific inhibitor of mRNA some signaling represents the primary and causal mechanisms under­
splicing [45], corrected olz or clz-induced intron retain of Npr2 and lying the SGAs cardiotoxicity. Second, previous studies have shown that
Ubp1 and significantly attenuated the drugs-induced secretion of PB, an inhibitor of the spliceosome signaling, evokes apoptosis and cell
pro-inflammatory factors and cell death rates. These lines of evidence death in several carcinoma cell lines [50,55]. We tested various con­
therefore strongly suggested that the spliceosome signaling was a crit­ centrations of PB and found that under a low dose ranging from 0.1 to
ical pathway driving the SGAs cardiotoxic effects. 10 nM, the PB has resulted in beneficial effects, suggesting that low
In particular, the three spliceosome signal proteins (Sf3b3, Snrpf, and doses of spliceosome signaling inhibitors might confer therapeutic,
Snrpd1) have been previously characterized in cancer biology but instead of additional toxic, outcomes. Third and more importantly, we
received minor attention in heart diseases. Spliceosome, the key ma­ aimed to identify the common pathways that shared by representative
chinery in RNA splicing, is a large and dynamic ribonucleoprotein SGAs. The identification of the spliceosome signaling as a shared
complex made up of proteins and small nuclear RNAs (snRNAs) [46]. mechanism driving SGAs cardiotoxicity would shed insights into the
Ribonucleoproteins are composed of 1 or 2 small nuclear RNAs (snRNPs; development of wide-spectrum cardioprotectants that may avoid
U1, U2, U4, U5, U6) and various complex-specific proteins [47]. discontinuation of on-the-market potent drug.
Changes in expression levels or mutations of snRNP and splice factors One limitation of the present study is that the number of replicates in
can produce aberrant mRNA splicing patterns on a genome-wide scale. omics studies is relatively restricted (n = 3). In further studies, more
Generally, Snrpd1 and Snrpf are small nuclear ribonucleoproteins biological replicates can be applied to validate the altered spliceosome
participating in the whole process of splicing and the splicing factor signaling after SGAs exposure.
Sf3b3 is part of the U2 snRNP and the minor U12-type spliceosome. In all, this study investigated the primary mechanism of cardiotox­
According to previous studies, Snrpd1 and Snrpf were reported to be icity induced by both olz, a first-line SGA regimen, and clz, another
biomarkers for breast cancer [48] and systemic lupus erythematosus powerful SGA for mental disorders. The dual-omics study integrating
[49]. Differential expression of Sf3b3 was detected in diffuse malignant proteome and transcriptome analyses identified the spliceosome
peritoneal mesothelioma [50] and chronic lymphocyte leukemia [51]. signaling as the common mechanism behind SGAs cardiotoxicity.
The data obtained from our omics analyses suggested that in addition to Pharmacological blockade of the spliceosome signaling under low doses
cancer biology, these proteins might also critically dominate the car­ successfully rescued SGAs-induced cardiotoxic effects. Our study
diotoxic effects by SGAs or other pharmacological compounds. Of note, depicted the molecular responses to chronic SGAs exposure and may
these proteins displayed different expression patterns, mirroring func­ provide venues for developing wide-spectrum cardioprotectants against
tional differences when executing SGAs cardiotoxicity. Hence, our SGAs cardiotoxicity.
findings broadened the functional importance of these spliceosome
signal proteins and it merits further investigation of the detailed roles
played by individual protein.

16
J. Wang et al. Pharmacological Research 170 (2021) 105714

Funding [10] C. Carnovale, E. Lucenteforte, V. Battini, F. Mazhar, M. Fornili, E. Invernizzi,


G. Mosini, M. Gringeri, A. Capuano, C. Scavone, M. Nobile, C. Vantaggiato,
S. Pisano, C. Bravaccio, S. Radice, E. Clementi, M. Pozzi, Association between the
This work was financially supported by the National Key Research glyco-metabolic adverse effects of antipsychotic drugs and their chemical and
and Development Program of China (No. 2018YFC0910700 and pharmacological profile: a network meta-analysis and regression, Psychol. Med.
2016YFB0201702), the National Natural Science Foundation of China (2021) 1–13.
[11] S. Shoja Shafti, P. Fallah Jahromi, Olanzapine induced Q-Tc shortening, Ther. Adv.
(No. 81701861, 81871527, and 82070285), the Shanghai Health Com­ Psychopharmacol. 4 (2014) 240–246.
mittee Research Foundation (20194Y0066), the Wang-Dao Research [12] H.J. Lee, J.S. Choi, S.J. Hahn, Mechanism of inhibition by olanzapine of cloned
Program of Fudan University (No. 20017), The Qing-Feng Scholar hERG potassium channels, Neurosci. Lett. 609 (2015) 97–102.
[13] P. Gulac, M. Arnold, M. Grman, T. Carrel, S. Longnus, T. Stankovicova,
Foundation of Shanghai Medical College, Fudan University (No. L. Tomasova, Olanzapine-mediated cardiotoxicity is associated with altered energy
QF2007), the Zhengyi Scholar Foundation of School of Basic Medical metabolism in isolated rat hearts, Acta Biochim Pol. 67 (2020) 15–23.
Sciences, Fudan University (No. S24-4). [14] B. Puttegowda, J. Theodore, R. Basappa, M.C. Nanjappa, Olanzapine induced
dilated cardiomyopathy, Malays. J. Med. Sci. 23 (2016) 82–84.
[15] F.P. Bymaster, K. Rasmussen, D.O. Calligaro, D.L. Nelson, N.W. DeLapp, D.
Authors contribution T. Wong, N.A. Moore, In vitro and in vivo biochemistry of olanzapine: a novel,
atypical antipsychotic drug, J. Clin. Psychiatry 58 (Suppl 10) (1997) 28–36.
[16] M.E. Stockton, K. Rasmussen, Electrophysiological effects of olanzapine, a novel
JW and XL performed animal experiments and analyzed data. ZL and atypical antipsychotic, on A9 and A10 dopamine neurons,
XL helped in animal experiments. SL and XT performed cell culture. FZ Neuropsychopharmacology 14 (1996) 97–105.
and YZ analyzed the dual-omics data. JW drafted the manuscript. LL [17] D. Siskind, L. McCartney, R. Goldschlager, S. Kisely, Clozapine v. first- and second-
generation antipsychotics in treatment-refractory schizophrenia: systematic review
conceived, designed the study and edited the manuscript. All authors and meta-analysis, Br. J. Psychiatry 209 (2016) 385–392.
read and approved the final version of the manuscript. [18] D. De Berardis, G. Rapini, L. Olivieri, D. Di Nicola, C. Tomasetti, A. Valchera,
M. Fornaro, F. Di Fabio, G. Perna, M. Di Nicola, G. Serafini, A. Carano, M. Pompili,
F. Vellante, L. Orsolini, G. Martinotti, M. Di Giannantonio, Safety of antipsychotics
Declaration of Competing Interest for the treatment of schizophrenia: a focus on the adverse effects of clozapine,
Ther. Adv. Drug Saf. 9 (2018) 237–256.
[19] A. Essali, N. Al-Haj Haasan, C. Li, J. Rathbone, Clozapine versus typical neuroleptic
The authors declare that they have no known competing financial medication for schizophrenia, Cochrane Database Syst. Rev. 2009 (2009), 000059.
interests or personal relationships that could have appeared to influence [20] M. Wiciński, M.M. Węclewicz, Clozapine-induced agranulocytosis/
granulocytopenia: mechanisms and monitoring, Curr. Opin. Hematol. 25 (2018)
the work reported in this paper.
22–28.
[21] L. Li, X. Ye, Z. Zhao, P. Gao, Y. Jiang, Overlooked fatal infectious diseases after
Acknowledgment long-term antipsychotic use in patients with psychiatric illness, Schizophr. Res. 195
(2018) 258–259.
[22] S.Y. Lee, Y.J. Kim, K.T. Kim, H. Choe, S.H. Jo, Blockade of HERG human K+
The authors would like to thank professor Yongbo Wang from the channels and IKr of guinea-pig cardiomyocytes by the antipsychotic drug
Department of Cellular and Genetic Medicine, School of Basic Medical clozapine, Br. J. Pharmacol. 148 (2006) 499–509.
[23] D.L. Youssef, P. Narayanan, N. Gill, Incidence and risk factors for clozapine-
Sciences, Fudan University for his generous gift of the spliceosome
induced myocarditis and cardiomyopathy at a regional mental health service in
signaling inhibitor (pladienolide B) and professor L. D. Timmie Top­ Australia, Australas. Psychiatry 24 (2016) 176–180.
oleski from the Mechanical Engineering Department at the University of [24] R. Ahangari, S. Khezri, A. Jahedsani, S. Bakhshii, A. Salimi, Ellagic acid alleviates
clozapine‑induced oxidative stress and mitochondrial dysfunction in
Maryland, Baltimore County, MD, USA, for his thoughtful comments and
cardiomyocytes, Drug Chem. Toxicol. (2020) 1–9.
careful editing. [25] G. Kanniah, S. Kumar, Clozapine associated cardiotoxicity: issues, challenges and
way forward, Asian J. Psychiatr. 50 (2020), 101950.
[26] M. Beauchemin, R. Geguchadze, A.R. Guntur, K. Nevola, P.T. Le, D. Barlow,
Appendix A. Supporting information M. Rue, C.P.H. Vary, C.W. Lary, K.J. Motyl, K.L. Houseknecht, Exploring
mechanisms of increased cardiovascular disease risk with antipsychotic
Supplementary data associated with this article can be found in the medications: risperidone alters the cardiac proteomic signature in mice,
Pharmacol. Res 152 (2020), 104589.
online version at doi:10.1016/j.phrs.2021.105714.
[27] S. Aringhieri, M. Carli, S. Kolachalam, V. Verdesca, E. Cini, M. Rossi, P.
J. McCormick, G.U. Corsini, R. Maggio, M. Scarselli, Molecular targets of atypical
References antipsychotics: from mechanism of action to clinical differences, Pharmacol. Ther.
192 (2018) 20–41.
[28] A.K. Tiwari, D. Zhang, J.G. Pouget, C.C. Zai, N.I. Chowdhury, E.J. Brandl, L. Qin,
[1] S.U. Lee, V. Ryu, M. Soh, C.E. Kim, S. Park, S. Roh, I.H. Oh, H.Y. Lee, S. Choi,
N. Freeman, J.A. Lieberman, H.Y. Meltzer, J.L. Kennedy, D.J. Müller, Impact of
Changes in antipsychotic drug usage and factors affecting the use of typical drugs
histamine receptors H1 and H3 polymorphisms on antipsychotic-induced weight
based on nationwide health insurance data in South Korea, BMJ Open 8 (2018),
gain, World J. Biol. Psychiatry 19 (2018) S97–s105.
020280.
[29] C.U. Correll, T. Lencz, A.K. Malhotra, Antipsychotic drugs and obesity, Trends Mol.
[2] R. Ronsley, D. Scott, W.P. Warburton, R.D. Hamdi, D.C. Louie, J. Davidson,
Med. 17 (2011) 97–107.
C. Panagiotopoulos, A population-based study of antipsychotic prescription trends
[30] L.J. Field, Atrial natriuretic factor-SV40 T antigen transgenes produce tumors and
in children and adolescents in British Columbia, from 1996 to 2011, Can. J.
cardiac arrhythmias in mice, Science 239 (1988) 1029–1033.
Psychiatry 58 (2013) 361–369.
[31] W.C. Claycomb, N.A. Lanson Jr., B.S. Stallworth, D.B. Egeland, J.B. Delcarpio,
[3] M. De Hert, J. Detraux, R. van Winkel, W. Yu, C.U. Correll, Metabolic and
A. Bahinski, N.J. Izzo Jr., HL-1 cells: a cardiac muscle cell line that contracts and
cardiovascular adverse effects associated with antipsychotic drugs, Nat. Rev.
retains phenotypic characteristics of the adult cardiomyocyte, Proc. Natl. Acad. Sci.
Endocrinol. 8 (2011) 114–126.
USA 95 (1998) 2979–2984.
[4] T.S. Stroup, N. Gray, Management of common adverse effects of antipsychotic
[32] S.M. White, P.E. Constantin, W.C. Claycomb, Cardiac physiology at the cellular
medications, World Psychiatry 17 (2018) 341–356.
level: use of cultured HL-1 cardiomyocytes for studies of cardiac muscle cell
[5] X. Li, Z. Peng, Y. Zhou, J. Wang, X. Lin, X. Dong, X. Liu, J. Jiang, Y. Jiang, L. Li,
structure and function, Am. J. Physiol. Heart Circ. Physiol. 286 (2004)
Quetiapine induces myocardial necroptotic cell death through bidirectional
H823–H829.
regulation of cannabinoid receptors, Toxicol. Lett. 313 (2019) 77–90.
[33] M. Wilhelm, J. Schlegl, H. Hahne, A.M. Gholami, M. Lieberenz, M.M. Savitski,
[6] J. Zhu, W. Hou, Y. Xu, F. Ji, G. Wang, C. Chen, C. Lin, X. Lin, J. Li, C. Zhuo,
E. Ziegler, L. Butzmann, S. Gessulat, H. Marx, T. Mathieson, S. Lemeer,
M. Shao, Antipsychotic drugs and sudden cardiac death: a literature review of the
K. Schnatbaum, U. Reimer, H. Wenschuh, M. Mollenhauer, J. Slotta-Huspenina, J.
challenges in the prediction, management, and future steps, Psychiatry Res. 281
H. Boese, M. Bantscheff, A. Gerstmair, F. Faerber, B. Kuster, Mass-spectrometry-
(2019), 112598.
based draft of the human proteome, Nature 509 (2014) 582–587.
[7] K.G. Jensen, K. Juul, A. Fink-Jensen, C.U. Correll, A.K. Pagsberg, Corrected QT
[34] A.R. Abela, X.D. Ji, Z. Li, A.D. Lê, P.J. Fletcher, Clozapine reliably increases the
changes during antipsychotic treatment of children and adolescents: a systematic
motivation for food: parsing the role of the 5-HT(2c) and H(1) receptors,
review and meta-analysis of clinical trials, J. Am. Acad. Child Adolesc. Psychiatry
Psychopharmacology 237 (2020) 957–966.
54 (2015) 25–36.
[35] X. Chen, Y. Yu, P. Zheng, T. Jin, M. He, M. Zheng, X. Song, A. Jones, X.F. Huang,
[8] C. Polcwiartek, K. Kragholm, O. Schjerning, C. Graff, J. Nielsen, Cardiovascular
Olanzapine increases AMPK-NPY orexigenic signaling by disrupting H1R-GHSR1a
safety of antipsychotics: a clinical overview, Expert Opin. Drug Saf. 15 (2016)
interaction in the hypothalamic neurons of mice, Psychoneuroendocrinology 114
679–688.
(2020), 104594.
[9] W.A. Ray, C.P. Chung, K.T. Murray, K. Hall, C.M. Stein, Atypical antipsychotic
drugs and the risk of sudden cardiac death, N. Engl. J. Med. 360 (2009) 225–235.

17
J. Wang et al. Pharmacological Research 170 (2021) 105714

[36] A.S. Lo, C.T. Liew, S.M. Ngai, S.K. Tsui, K.P. Fung, C.Y. Lee, M.M. Waye, [46] M.C. Wahl, C.L. Will, R. Lührmann, The spliceosome: design principles of a
Developmental regulation and cellular distribution of human cytosolic malate dynamic RNP machine, Cell 136 (2009) 701–718.
dehydrogenase (MDH1), J. Cell. Biochem. 94 (2005) 763–773. [47] M.S. Jurica, M.J. Moore, Pre-mRNA splicing: awash in a sea of proteins, Mol. Cell
[37] V. Ladopoulos, H. Hofemeister, M. Hoogenkamp, A.D. Riggs, A.F. Stewart, 12 (2003) 5–14.
C. Bonifer, The histone methyltransferase KMT2B is required for RNA polymerase [48] V. Quidville, S. Alsafadi, A. Goubar, F. Commo, V. Scott, C. Pioche-Durieu,
II association and protection from DNA methylation at the MagohB CpG island I. Girault, S. Baconnais, E. Le Cam, V. Lazar, S. Delaloge, M. Saghatchian,
promoter, Mol. Cell. Biol. 33 (2013) 1383–1393. P. Pautier, P. Morice, P. Dessen, S. Vagner, F. André, Targeting the deregulated
[38] S. Manivasagam, V. Subramanian, A. Tumala, E. Vellaichamy, Differential spliceosome core machinery in cancer cells triggers mTOR blockade and
expression and regulation of anti-hypertrophic genes Npr1 and Npr2 during autophagy, Cancer Res. 73 (2013) 2247–2258.
β-adrenergic receptor activation-induced hypertrophic growth in rats, Mol. Cell. [49] J.B. Engler, R. Undeutsch, L. Kloke, S. Rosenberger, M. Backhaus, U. Schneider,
Endocrinol. 433 (2016) 117–129. K. Egerer, D. Dragun, J. Hofmann, D. Huscher, G.R. Burmester, J.Y. Humrich,
[39] H. Koutnikova, M. Laakso, L. Lu, R. Combe, J. Paananen, T. Kuulasmaa, P. Enghard, G. Riemekasten, Unmasking of autoreactive CD4 T cells by depletion of
J. Kuusisto, H.U. Häring, T. Hansen, O. Pedersen, U. Smith, M. Hanefeld, R. CD25 regulatory T cells in systemic lupus erythematosus, Ann. Rheum. Dis. 70
W. Williams, J. Auwerx, Identification of the UBP1 locus as a critical blood (2011) 2176–2183.
pressure determinant using a combination of mouse and human genetics, PLoS [50] R. Sciarrillo, A. Wojtuszkiewicz, B. El Hassouni, N. Funel, P. Gandellini,
Genet. 5 (2009), 1000591. T. Lagerweij, S. Buonamici, M. Blijlevens, E.A. Zeeuw van der Laan, N. Zaffaroni,
[40] B.A. Abdel-Wahab, M.E. Metwally, M.M. El-khawanki, A.M. Hashim, Protective M. Deraco, S. Kusamura, T. Würdinger, G.J. Peters, C.F.M. Molthoff, G. Jansen, G.J.
effect of captopril against clozapine-induced myocarditis in rats: role of oxidative L. Kaspers, J. Cloos, E. Giovannetti, Splicing modulation as novel therapeutic
stress, proinflammatory cytokines and DNA damage, Chem. Biol. Interact. 216 strategy against diffuse malignant peritoneal mesothelioma, EBioMedicine 39
(2014) 43–52. (2019) 215–225.
[41] L. Li, X. Dong, C. Tu, X. Li, Z. Peng, Y. Zhou, D. Zhang, J. Jiang, A. Burke, Z. Zhao, [51] A. Foy, M.F. McMullin, Somatic SF3B1 mutations in myelodysplastic syndrome
L. Jin, Y. Jiang, Opposite effects of cannabinoid CB(1) and CB(2) receptors on with ring sideroblasts and chronic lymphocytic leukaemia, J. Clin. Pathol. 72
antipsychotic clozapine-induced cardiotoxicity, Br. J. Pharmacol. 176 (2019) (2019) 778–782.
890–905. [52] K. Noguchi, J. Ishida, J.D. Kim, N. Muromachi, K. Kako, H. Mizukami, W. Lu,
[42] B.A. Abdel-Wahab, M.E. Metwally, Clozapine-induced cardiotoxicity in rats: T. Ishimaru, S. Kawasaki, S. Kaneko, J. Usui, H. Ohtsu, K. Yamagata, A. Fukamizu,
Involvement of tumour necrosis factor alpha, NF-κβ and caspase-3, Toxicol. Rep. 1 Histamine receptor agonist alleviates severe cardiorenal damages by eliciting anti-
(2014) 1213–1223. inflammatory programming, Proc. Natl. Acad. Sci. USA 117 (2020) 3150–3156.
[43] J.G. Kilian, K. Kerr, C. Lawrence, D.S. Celermajer, Myocarditis and cardiomyopathy [53] B.A. Abdel-Wahab, M.E. Metwally, Clozapine-induced cardiotoxicity: role of
associated with clozapine, Lancet 354 (1999) 1841–1845. oxidative stress, tumour necrosis factor alpha and NF-κβ, Cardiovasc. Toxicol. 15
[44] C.S. Wu, Y.T. Tsai, H.J. Tsai, Antipsychotic drugs and the risk of ventricular (2015) 355–365.
arrhythmia and/or sudden cardiac death: a nation-wide case-crossover study, [54] J.F. Wang, J.Y. Min, T.G. Hampton, I. Amende, X. Yan, S. Malek, W.H. Abelmann,
J. Am. Heart Assoc. 4 (2015). A.I. Green, J. Zeind, J.P. Morgan, Clozapine-induced myocarditis: role of
[45] K.A. Effenberger, D.D. Anderson, W.M. Bray, B.E. Prichard, N. Ma, M.S. Adams, A. catecholamines in a murine model, Eur. J. Pharmacol. 592 (2008) 123–127.
K. Ghosh, M.S. Jurica, Coherence between cellular responses and in vitro splicing [55] J. Jorge, S. Petronilho, R. Alves, M. Coucelo, A.C. Gonçalves, J.M. Nascimento
inhibition for the anti-tumor drug pladienolide B and its analogs, J. Biol. Chem. Costa, A.B. Sarmento-Ribeiro, Apoptosis induction and cell cycle arrest of
289 (2014) 1938–1947. pladienolide B in erythroleukemia cell lines, Invest New Drugs 38 (2020) 369–377.

18

You might also like