Access to this full-text is provided by Springer Nature.
Content available from Scientific Reports
This content is subject to copyright. Terms and conditions apply.
1
Vol.:(0123456789)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports
MCPIP1 functions as a safeguard
of early embryonic development
Agata Lichawska‑Cieslar
1,6, Weronika Szukala
1,2,6, Tomasz K. Prajsnar
3,6,
Niedharsan Pooranachandran
3, Maria Kulecka
4,5, Michalina Dabrowska
5, Michal Mikula
5,
Krzysztof Rakus
3, Magdalena Chadzinska
3 & Jolanta Jura
1*
Monocyte chemoattractant protein‑induced protein 1 (MCPIP1), also called Regnase‑1, is an RNase
that has been described as a key negative modulator of inammation. MCPIP1 also controls numerous
tumor‑related processes, such as proliferation, apoptosis and dierentiation. In this study, we utilized
a zebrash model to investigate the role of Mcpip1 during embryogenic development. Our results
demonstrated that during embryogenesis, the expression of the zc3h12a gene encoding Mcpip1
undergoes dynamic changes. Its transcript levels gradually increase from the 2‑cell stage to the
spherical stage and then decrease rapidly. We further found that ectopic overexpression of wild‑type
Mcpip1 but not the catalytically inactive mutant form resulted in an embryonic lethal phenotype in
zebrash embryos (24 hpf). At the molecular level, transcriptomic proling revealed extensive changes
in the expression of genes encoding proteins important in the endoplasmic reticulum stress response
and in protein folding as well as involved in the formation of primary germ layer, mesendoderm and
endoderm development, heart morphogenesis and cell migration. Altogether, our results demonstrate
that the expression of zc3h12a must be tightly controlled during the rst cell divisions of zebrash
embryos and that a rapid decrease in its mRNA expression is an important factor promoting proper
embryo development.
Implantation of the blastocyst is necessary for embryonic development to occur. In mammals, uterine implanta-
tion is associated with numerous structural and molecular changes in the luminal epithelia1. Interestingly, this
process is closely correlated with the inammatory mechanism, where during the attachment of blastocysts to
the uterus, modulations in the expression of dierent growth factors, chemokines and cytokines, such as TNF,
IL-6 and prostaglandin E2 (PGE2), are observed, as well as vascularization and inltration of immune cells from
the blood to the endometrial tissue2–5. e inammatory process is essential for implantation, but during the
next stage of pregnancy, anti-inammatory factors are induced to prevent rejection of the fetus. us, proper
embryonic development is strictly dependent on optimal microenvironmental conditions. is microenviron-
ment must provide optimal temperature, oxygen tension, pH, and nutrients to ensure embryo survival. Stress
during the embryonic period may impair fetal development and result in embryo mortality6,7.
In human, monocyte chemoattractant protein-induced protein 1 (MCPIP1), also known as Regnase-1 and
encoded by the ZC3H12A gene, possesses a PilT N-terminus domain (PIN) that exerts RNAse properties. It has
been shown that MCPIP1 degrades transcripts coding for mediators of inammation: IL-1β8, IL-69, IL-12p4010
and IL-211 and its own transcript10. Further studies have indicated that MCPIP1 also plays an important role
in the suppression of miRNA activity and biogenesis via cleavage of the terminal loops of precursor miRNAs
(pre-miRNAs), counteracting Dicer, a key ribonuclease in miRNA processing12. MCPIP1 transcript expression
is induced by Toll-like receptor (TLR) ligands9,13 and proinammatory cytokines, such as IL-1β and TNFα8.
In vivo studies have shown that MCPIP1 plays a critical role in preventing autoimmune conditions. Zc3h12a
knockout mice display severe anemia, augmented serum immunoglobulin levels and autoantibody production.
In macrophages of Zc3h12a-/- mice, elevated expression levels of IL-6, IL-12p40 and IL-1β were observed9,14.
Changes in the immune response of knockout mice are not only the result of MCPIP1 involvement in mRNA
OPEN
1Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian
University, Gronostajowa 7, 30-387 Kraków, Poland. 2Doctoral School of Exact and Natural Sciences, Jagiellonian
University, Lojasiewicza 11, 30-348 Kraków, Poland. 3Department of Evolutionary Immunology, Institute of
Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Kraków,
Poland. 4Medical Center for Postgraduate Education, Department of Gastroenterology, Hepatology and Clinical
Oncology, Marymoncka 99/103, 01-813 Warsaw, Poland. 5Maria Sklodowska-Curie National Research Institute of
Oncology, Roentgena 5, 02-781 Warsaw, Poland. 6
These authors contributed equally: Agata Lichawska-Cieslar,
Weronika Szukala and Tomasz K. Prajsnar. *email: jolanta.jura@uj.edu.pl
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
Vol:.(1234567890)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
decay and miRNA biogenesis but also of its inuence on the activity of some transcription factors. To date,
MCPIP1 has been conrmed to negatively regulate the activity of NF-ĸB and AP1, which is essential in the
regulation of the synthesis of mediators controlling inammatory and immune responses14–17.
In addition, evidence indicates that MCPIP1 is involved in cell cycle arrest18, apoptosis19 and regulation of cell
dierentiation20–22. us, we hypothesized that MCPIP1 might safeguard early development and regulate crucial
processes in the early stages of embryonic development, such as cell dierentiation, cell division, apoptosis,
angiogenesis, and regulation of inammatory processes. A zebrash (Danio rerio) model was utilized, which is a
powerful vertebrate platform widely used to investigate developmental processes. First, we investigated whether
the temporal expression pattern of the zc3h12a transcript, encoding Mcpip1, is changed during embryogenesis.
Subsequently, we determined how the modulation of Mcpip1 levels aects the early development of zebrash.
Results
The zebrash genome contains four unique members of the PIN domain‑like superfamily
To begin investigating the potential function of the Mcpip1 RNase in zebrash, we performed bioinformatic
searches and alignments. According to the Ensembl and NCBI databases, the zebrash genome contains ve
members of the PIN domain-like superfamily (Fig.1a,b), also recently showed by Yang et al.23. e full-length
zebrash zc3h12a sequence (XM_021466808.1; Gene ID: 798235; NCBI) encodes a 579 amino acid Mcpip1
protein with high (82.90%) amino acid identity to common carp Mcpip1 and intermediate (~ 50%) amino acid
identity to amphibian, reptilian, bird and mammalian MCPIP1. e maximum likelihood (ML) phylogenetic
tree for MCPIP1 proteins (Supplementary Fig.S1) demonstrates that the sh species cluster together and form
a clade separate from the nonsh species. However, a direct comparison of the zebrash orthologs of the human
PIN-domain like-superfamily members indicated that zebrash and human MCPIP1 proteins possess a similar
domain structure and 87.79% identity within the PIN domain (Fig.1c, Supplementary Fig.S2). e most highly
conserved region is the PIN domain, the catalytic center of Mcpip1 (Fig.1d).
The zebrash ortholog of ZC3H12A is downregulated during early embryonic development
To investigate the temporal expression pattern of the zebrash zc3h12a gene during early embryonic develop-
ment, quantitative real-time PCR (qRT‒PCR) was performed on whole embryo-derived RNA. One-cell stage
fertilized zebrash eggs were collected and incubated at a standard temperature of 28°C as described in the
Figure1. e zebrash ortholog of human MCPIP1 shares a similar domain structure. (a) List of genes
containing the Zc3h12a-like NYN domain in the zebrash genome based on the Ensembl database. (b) List
of genes containing the Zc3h12a-like NYN domain in the zebrash genome based on the NCBI database. (c)
Domain comparison of human MCPIP1 and its zebrash ortholog Mcpip1. UBA—ubiquitin-associated domain,
PRR—proline-rich region, PIN—PilT N-terminus nuclease domain, ZF—zinc nger motif, NDR—disordered
region, CTD—C-terminal conserved domain. (d) Alignment of the amino acid sequences of human and
zebrash PIN domains. Asterisk (*) denotes an identical residue, a colon (:) denotes conserved substitutions,
and a period (.) denotes semiconserved substitutions. e arrows indicate four aspartic acid residues, which are
critical for Mcpip1/MCPIP1 catalytic activity. Black rectangle indicates position of the D112N mutation within
the PIN domain of zebrash Mcpip1.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
Vol.:(0123456789)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
methods. e embryos were monitored for cell division and collected at dierent developmental periods over
72h. As expected, the rst divisions were observed synchronously at ~ 20-min intervals (Fig.2a)24. For the isola-
tion of RNA, ~ 10 embryos were pooled at each timepoint.
We found that the zc3h12a gene follows dynamic changes in expression during early zebrash development.
e level of zc3h12a mRNA gradually increases during the rst embryonic divisions, peaking at the sphere
stage (4h post fertilization [hpf]) and then signicantly rapidly declining during transition into the shield stage
and 1-day embryo (Fig.2b and Supplementary Fig.S3a). In addition, we analyzed the global transcriptomic
RNASeq data by White etal.25, which provides information about gene expression changes during early zebrash
embryogenesis. Both approaches showed a similar trend of zc3h12a gene expression uctuations during zebrash
embryogenesis (Fig.2c).
Transient thermal shock during early zebrash development enhances the expression of
zc3h12a mRNA
We next sought to investigate whether an external shock during early development aects the expression of the
zc3h12a gene. e thermal stimulation approach was performed, due to the fact that the more commonly used
zebrash infection/inammation models could not be eciently utilized to monitor processes in early embryos
(0–24 hpf)26. Synchronous populations of 2–4 cell embryos that had been cultured at 28°C were subjected to
a 30-min thermal shock at 4°C (cold shock) or 37°C (heat shock). Control embryos were kept at a standard
temperature of 28°C. Aer a transient temperature shi, the embryos were incubated at the standard 28°C and
collected at the indicated timepoints for RNA analysis (Fig.3a).
We ensured that the developmental stages of embryos subjected to cold or heat shock followed similar timing
as the control siblings (Fig.3b) so the analysis of zc3h12a gene expression was not biased by a potential shi of
developmental stage. Transient thermal shocks did not impair the overall embryonic survival rates at the 24-h
stage (Fig.3c).
Quantitative RT‒PCR analysis revealed that the expression of zc3h12a was signicantly enhanced in response
to the 30min cold shock, reaching levels ~ 2.5-fold higher than those in the control embryos at the shield stage (6
hpf; Fig.3d). e 37°C shock suggested a similar tendency on the zc3h12a mRNA expression level at the sphere
stage (4 hpf), with a ~ 1.5-fold increase (with adj. p value 0,097; Fig.3e).
Figure2. e expression of zc3h12a is tightly controlled during early embryonic development in zebrash.
(a) One-cell fertilized eggs were incubated under standard conditions and collected at the indicated timepoints
for RNA analysis. Representative pictures of zebrash embryos at various developmental stages are shown.
(b) Real-time PCR analysis of the zc3h12a expression level at dierent developmental stages of zebrash. (c)
Graph represents the level of zc3h12a mRNA based on the RNA-Seq data (PMID: 29,144,233). For comparison
of both datasets, mean values of zc3h12a mRNA levels from qRT-PCR were also shown (normalized values of
data presented on Fig.2a). In both cases the level of zc3h12a mRNA was normalized to the expression level at
2-cell stage (0.75 hpf). hpf—hours post fertilization n = 3. Rps11 was used as a reference gene. Data represent
the mean ± SD, ****p < 0.0001 by one-way ANOVA (only comparisons with p < 0.0001 are shown). Scale
bar = approx. 250µm.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
Vol:.(1234567890)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
Overexpression of zc3h12a impairs early embryonic development in zebrash
Zebrash Mcpip1 was overexpressed by microinjection of invitro transcribed mRNA encoding the zebrash
Mcpip1-P2A-mTurquoise protein into the yolks of one-cell stage embryos. Expression of Mcpip1 was tied
to mTurquoise expression via self-cleaving P2A peptide. is strategy enabled visualization of exogenously
expressed protein. For overexpression of catalytically inactive Mcpip1, one of the four conserved aspartic acid
residues within the RNase domain (Fig.1d) was substituted with asparagine (Mcpip1-D112N, herein Mcpip1
DN). It has previously been shown that a single mutation in the catalytic center of Mcpip1 issucient to com-
pletely abolish its RNase activity9. As a control, a construct encoding only P2A-mTurquoise was used (Fig.4a).
Prior to microinjection, agarose electrophoresis was performed, which indicated high quality of invitro tran-
scribed mRNAs (see Supplementary Fig. S3b).
Embryos were sampled at 4, 6 and 24h aer microinjection. Exogenous zc3h12a mRNA was clearly detected
at the sphere (4 hpf) and shield (6 hpf) stages. We also found that the microinjection procedure itself did not
aect the kinetics of the zc3h12a mRNA expression pattern at those two developmental stages (Fig.4b).
We further conrmed that mTurquoise expressed from microinjected mRNA was detected in all modied
embryos at the shield stage (6 hpf; Fig.4c). We also observed that at this stage, overexpression of wild-type (WT)
but not the RNase-dead mutant did not aect embryo morphology (Fig.4d). However, embryonic lethality
increased by ~ 10% (Fig.4e). Consistent with this nding, 24h aer microinjection, most Mcpip1-overexpressing
embryos showed gross morphological abnormalities; thus, the overall lethality rate was signicantly higher than
that of the control embryos (Fig.4f–g).
Increased activity of Mcpip1 leads to profound transcriptomic changes in developing zebrash
embryos
Transcriptomic proling was performed on RNA isolated from viable Mcpip1 WT and Mcpip1 DN-overexpress-
ing embryos collected at the shield stage (n = 3 of ~ 10 pooled embryos per condition). Dierentially expressed
genes (DEGs) were dened with a threshold of p value < 0.05 and fold change > 1.5. Accordingly, the expressions
of 247 genes were signicantly upregulated and those of 303 genes were downregulated in zebrash embryos
overexpressing WT Mcpip1 compared to those overexpressing the Mcpip1 DN mutant (Fig.5a).
Gene Ontology (GO) enrichment analyses revealed that upregulated DEGs were signicantly enriched in
biological processes (BP) mostly related to the response to protein folding and endoplasmic reticulum stress,
examples of which are calr3a (calreticulin 3a), edem 2 (ER degradation enhancer) and hsp70 (heat shockprotein70)
family genes (Fig.5b,c).
In contrast, the downregulated DEGs were enriched in pathways related to endoderm development, le/
right pattern formation, organ morphogenesis and formation of the primary germ layer and ectodermal placode
development (Fig.5d,e).
Figure3. Eect of transient thermal shock on the expression of zebrash zc3h12a during early embryonic
development. (a) Zebrash embryos that were incubated at standard 28°C temperature for 1h were subjected
to a transient 30-min heat shock (37°C) or cold shock (4°C) as indicated on the schematic diagram. Embryos
were collected for RNA analysis at the indicated timepoint. (b) Representative pictures of zebrash embryos at
4 hpf. c. Percentages of live and dead embryos at 24 hpf. (d,e) Real-time PCR analysis of the zc3h12a expression
level in zebrash embryos subjected to each thermal shock. e arrow on the X-axis indicates the onset of the
30min thermal shock. hpf—hours post fertilization. n = 4. Rps11 was used as a reference gene. Data represent
the mean ± SD, **p < 0.01 by an unpaired t-test. Scale bar = approx. 250µm.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
Vol.:(0123456789)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
Oscillation of zc3h12a expression during early embryonic development in zebrash inversely
correlates with changes in nrarpa and rasl11b expression levels
Based on our RNASeq analysis, we next selected four signicantly downregulated DEGs in zebrash embryos
overexpressing WT Mcpip1: nrarpa (Notch-regulated ankyrin repeat-containing protein A), rasl11b (RAS-like
family 11 member B), rhov (ras homolog family member V) and foxh1 (forkhead box H1). ese DEGs were
functionally assigned to the most signicantly downregulated processes (Fig.5d,e), and their expression levels
were validated by qRT‒PCR (Fig.6a).
In the next step, the expression patterns of two transcripts, nrarpa and rasl11b, were analyzed over the course
of zebrash embryogenesis by qRT‒PCR (Fig.2a). We found that their expression levels undergo extensive
changes during the transition from 64-cell to 1-day embryos (Fig.6b). Moreover, the relative expression levels
of those genes peaked at the shield stage, which correlates with the time at which zc3h12a expression profoundly
decreases (Fig.2b).
Discussion
Zebrash (D. rerio) is a leading model for studying developmental biology because the genome is known, fertiliza-
tion occurs externally, and development is very fast. At 48h aer fertilization, zebrash embryos form complete
organ systems, including the heart, intestine and blood vessels. In addition, embryos are transparent and develop
outside the uterus, making it even easier to track developmental stages24,27.
Figure4. Overexpression of catalytically active Mcpip1 impairs early development in zebrash. (a) Schematic
diagram of control and Mcpip1 overexpression constructs. e sequences were cloned into the pCS2 expression
vector. (b) Real-time PCR analysis of zc3h12a expression levels in uninjected and microinjected control,
Mcpip1 WT or Mcpip1 DN mRNA zebrash embryos at 4 hpf and 6 hpf. n = 3. (c) Representative uorescence
(mTurquoise) images of embryos at 6 hpf. (d) Representative bright eld images of embryos at 6 hpf. (e)
Percentages of live and dead embryos at 6 hpf. (f) Representative bright eld images of embryos at 24 hpf. (g)
Graph indicating the percentages of live and dead embryos at 24 hpf. n = 3. Rps11 was used as a reference gene.
Data represent the mean ± SD, *p < 0.05, **p < 0.01, ****p < 0.0001 by an unpaired t-test (b) or one-way ANOVA
(d,f). Scale bar = approx. 250µm.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
Vol:.(1234567890)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
MCPIP1 expression is inducible, and mRNA levels change rapidly aer stimuli associated with cell dier-
entiation, stress induction or pathogen infection8,9,28. Consequently, an increase in MCPIP1 is correlated with
a decrease in the levels of many transcripts encoding regulators of inammation, regulators of cell dierentia-
tion and division, and regulators of apoptosis and angiogenesis. Here, we wanted to test the role of MCPIP1
in embryological development. Comparison of the amino acid sequences showed dierences between human
MCPIP1 and its zebrash ortholog. However, the PIN domain has high homology with the conserved codons
encoding four acidic amino acid residues that form the putative active site, which is essential for the ribonu-
cleolytic activity of MCPIP1.
We found that the expression of zc3h12a is tightly controlled within the rst cell divisions of zebrash.
Mcpip1 mRNA levels increase from the 2-cell stage up to the sphere stage (6h aer fertilization); later, its level
drops dramatically. us, starting in the sphere stage, Mcpip1-dependent transcript level regulation is silenced.
Relatively high expression of Mcpip1 during rst hours post fertilization may suggest potential role of this RNase
during the decay of maternal mRNA (which in zebrash is completed by 6 hpf29), but this hypothesis requires
further investigation.
MCPIP1 is encoded by an inducible gene that has the ability to rapidly respond to microenvironmental
changes and responds quickly to various kinds of stress8,9,28,30,31. In this study, mild external stress was induced
Figure5. Transcriptome analysis of zebrash embryos overexpressing Mcpip1. (a) Volcano plot for the RNA-
Seq dataset indicating dierentially expressed genes with p value < 0.05 and fold change (FC) > 1.5 between
embryos microinjected with Mcpip1 WT and Mcpip1 DN mRNA (6 hpf). n = 3. (b) Gene Ontology (GO)
enrichment analysis of upregulated biological processes in Mcpip1 WT. (c) Heatmap illustrating the expression
levels of selected upregulated DEGs. (d) Gene Ontology (GO) enrichment analysis of downregulated biological
processes in Mcpip1 WT. (e) Heatmap illustrating the expression levels of selected downregulated DEGs.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
Vol.:(0123456789)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
by a temporary change in temperature, which led to transient modulations of zc3h12a gene expression, with
consequent eects on the RNA prole. To determine what consequences an increase in Mcpip1 transcript levels
might have on embryonic development, we used zebrash models overexpressing WT Mcpip1 or its mutant
form containing a mutation in the active site of the PIN domain.
We found that ectopic expression of WT Mcpip1 resulted in a 10% death rate of zebrash embryos at 6h
post-fertilization and almost 90% aer 24h, while the model expressing the inactive form of Mcpip1 resembled
control models (uninjected zebrash embryos or injected with an empty vector). ese results provide evidence
for a signicant eect of Mcpip1 on the zebrash embryo transcriptome. ere are many studies in the literature
showing that Mcpip1 regulates genes involved in apoptotic processes16,32–34, so high embryonic lethality may
be a consequence of the dysregulation of transcripts involved in this process. In our model, activation of the
endoplasmic reticulum (ER) stress response pathway observed in the Mcpip1-overexpressing embryos already
at 6 hpf (Fig.5c) could have provoked the activation of apoptotic cell death, as prolonged ER-stress is a common
trigger of apoptosis35. Furthermore, Mcpip1-overexpressing embryos showed gross morphological abnormali-
ties. Transcriptome analysis revealed expression dierences in mRNA classes encoding proteins important for
embryonic development. We observed that, among the genes that were activated, the largest group comprised
genes encoding proteins important to the endoplasmic reticulum stress response and involved in protein fold-
ing. e activation of these genes may be an indirect consequence of the ectopic expression of Mcpip1 and the
accumulation of this protein in embryonic zebrash cells. However, among the many transcripts downregulated
and thus potentially directly degraded by Mcpip1 were those involved in the formation of the primary germ
layer, mesendoderm, ectoderm and endoderm development, heart morphogenesis and cell migration. us, the
change in the expression levels of such developmentally important genes may explain why such high lethality was
observed when Mcpip1 levels were elevated during early zebrash development. For validation of RNA-Seq data
Figure6. Expression pattern of nrarpa, rasl11b, rhov and foxh1 genes during zebrash embryogenesis. (a)
Real-time PCR analysis of nrarpa, rasl11b, rhov and foxh1 expression levels in zebrash embryos overexpressing
Mcpip1 WT or Mcpip1 DN (6 hpf). (b) Real-time PCR analysis of nrarpa and rasl11b expression levels during
early embryonic development in zebrash. n = 3. Rps11 was used as a reference gene. Data represent the
mean ± SD, *p < 0.05, **p < 0.01, ****p < 0.0001 by an unpaired t-test (a) or one-way ANOVA (b). For (b) only
selected developmental stages have been compared.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
Vol:.(1234567890)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
by qRT-PCR, four signicantly downregulated transcripts in zebrash embryos, nrarpa (Notch-regulated ankyrin
repeat-containing protein A), rasl11b (RAS-like family 11 member B), rhov (ras homolog family member V) and
foxh1 (forkhead box H1) were selected. We also showed that the levels of nrarpa and rasl11b inversely correlate
with uctuations in Mcpip1 mRNA levels during early stages of development (Fig.6b). We hypothesize that a
rapid decline of Mcpip1 expression level observed at shield stage (6 hpf) contributes to a profound upregulation
of nrarpa, rasl11b and possibly other developmentally important target mRNAs at this stage.
NRARP is a component of the Notch signaling pathway and participates in embryonic development in ver-
tebrates by regulating the segmentation of the body axis. e importance of this transcript in the segmentation
process has been documented in lower and higher vertebrates36–38. e rasl11b gene, on the other hand, encodes
a small GTPase protein that is highly conserved among vertebrates; it is expressed in mesendodermal cells, and
its expression is controlled by the Nodal pathway. Nodal signaling controls the expression of conserved mesendo-
dermal transcription factors39. Interestingly, it has been shown that rasl11b knockdown induces a specic “curly
tail down” phenotype in zebrash39. Similarly, ectopic overexpression of WT Mcpip1 resulted in embryo malfor-
mations, including tail malformations (Fig.4e). e third highly-downregulated transcript, rhov, is involved in
the signal transduction of the Rho pathway, which is essential for the regulation of gastrulation and neurulation,
two major developmental processes of early embryogenesis40,41. In addition, our analysis identied many other
genes whose levels are directly or indirectly dependent on Mcpip1, e.g., teratocarcinoma-derived growth factor 1
(tdgf1), described as an important regulator in the development of the cardiac tube in mouse embryogenesis42,
and forkhead box protein H1 (foxh1), essential during zebrash gastrulation and head and dorsal axis formation43.
Our RNAseq data also showed a decreasing trend of neurogenin 1 (neurog1) expression (p value = 0.077), a key
factor directing specialization of neuroectoderm44 in 6 hpf embryos overexpressing active Mcpip1 (Supplemen-
tary Fig. S3c), which additionally proves observed lethal phenotype.
In conclusion, our studies in the zebrash model showed that the Mcpip1 level is tightly regulated during
embryonic development, while even transient stress leads to rapid induction of the zc3h12a gene. Consequently,
elevated expression of the zc3h12a gene leads to abnormalities in zebrash development as a result of altered
levels of transcripts involved in processes important in embryogenesis. It can be speculated that stress leading
to induction of the gene encoding Mcpip1/MCPIP1 will have similar consequences on the developing embryo
in higher vertebrates, including humans.
Materials and methods
Phylogenetics and bioinformatics
To nd MCPIP1 orthologs in the zebrash genome (GRCz11), the Ensembl database was searched for genes
containing a Zc3h12a-like NYN domain. Sequences were retrieved from the SwissProt, EMBL and GenBank
databases using SRS and/or BLAST (Basic Local Alignment Search Tool)45. Amino acid sequence alignment
was performed using the Clustal Omega program at EMBL-EBI (https:// www. ebi. ac. uk/ Tools/ msa/ clust alo).
Phylogenetic trees were constructed on the basis of amino acid dierences by the maximum likelihood (ML)
method with 500 bootstrap replications using Molecular Evolutionary Genetics Analysis (MEGA) version 1146.
For metadata analysis of zc3h12a mRNA expression prole during zebrash development, the RNA-seq dataset
provided by the Busch-Nentwich lab and available at Expression Atlas, was used25.
Zebrash husbandry
Zebrash embryos/larvae were obtained by the natural spawning of adult zebrash (line AB/TL), which were
housed in a continuous recirculating closed-system aquarium with a light/dark cycle of 14/10h at 28°C.Larvae
were incubated in E3 medium at 28°C according to standard protocols47. e Jagiellonian University Zebrash
Core Facility (ZCF) is a licensed breeding and research facility (District Veterinary Inspectorate in Krakow
registry; Ministry of Science and Higher Education record no. 022 and 0057).
Eithics statement
All experiments were conducted in accordance with the European Community Council Directive 2010/63/EU
for the Care and Use of Laboratory Animals of Sept. 22, 2010 (Chapter1, Article 1 no.3) and National Journal
of Law act of Jan. 15, 2015 for Protection of animals used for scientic or educational purposes (Chapter1,
Article 2 no.1). All methods involving zebrash embryos/larvae were in compliance with ARRIVE guidelines.
e works with genetically modied microorganisms were authorized by the Polish Ministry of the Environ-
ment (No. 179/2021).
Cloning
For overexpression experiments, the full length mTurquoise cDNA was amplied by PCR from the p3E-p2a-
mTurquoise plasmid (a gi from David Tobin; Addgene plasmid #135213) via the StuI and XbaI sites of the pCS2
expression vector (a gi from Amro Hamdoun48; Addgene #34931) to generate pCS2-P2A-mTurquoise (control
plasmid), as presented in the schematic (Fig.4a). en, the cDNA coding for full length Mcpip1 (XM_021466808)
was amplied by PCR to introduce EcoRI and EcoRV and cloned into the EcoRI and StuI sites of the pCS2-P2A-
mTurquoise plasmid to generate the Mcpip1 WT plasmid. To clone vectors containing sequences encoding
catalytically inactive Mcpip1, site-directed mutagenesis of D112 into N112 was performed. e sequences of the
primers used for cloning are listed in Supplementary TableS1.
In vitro transcription
e pCS2 vectors (Control, Mcpip1 WT and Mcpip1 DN) were linearized with NotI digestion and cleaned
using the PCR Mini Kit (Syngen). en, the mRNAs were synthesized invitro from the SP6 promoter using the
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
Vol.:(0123456789)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
mMESSAGE mMACHINE SP6 kit (Ambion AM1340) according to the manufacturer’s protocol. Transcribed
mRNAs were puried using an RNA Clean & Concentrator kit (Zymo Research). A NanoDrop 2000 spectropho-
tometer (ermo Fisher Scientic) was used to calculate the concentration of mRNA, which was then diluted to
a concentration of 200ng/µl. e integrity of mRNA was also conrmed by denaturing agarose electrophoresis.
Microinjection of mRNA into fertilized zebrash eggs
For overexpression experiments, 3.5µl (700ng) of each mRNA was mixed with 0.5µl phenol red (Merck, P0290)
and microinjected into the yolks of approximately 100 zebrash eggs at the one-cell stage using a WPI Picopump
PV820 microinjector (2nl). e microinjected embryos were collected at 4 and 6 hpf for further RNA analysis.
RNA isolation and quantitative PCR
For RNA isolation, ~ 10 zebrash embryos were collected in fenozol (A&A Biotechnology), frozen and stored at
-80°C. en, the embryos were homogenized using a homogenizer (OMNI International), and total RNA was
extracted using the phenol–chloroform method. cDNA was synthesized using M-MLV reverse transcriptase
(Promega), and quantitative real-time PCR was performed with SYBR Green Master Mix (A&A Biotechnol-
ogy) and a QuantStudio3 thermocycler (ermo Fisher Scientic). Rps11, eef1, acbt2 and rpl13a were used as
a reference genes49,50 e primer sequences are listed in Supplementary TableS1. To validate specicity of the
qRT-PCR reaction, melt curve analysis was performed at the end of each assay. Agarose gel electrophoresis was
performed to ensure presence of a single product of predicted length at the end of the qRT-PCR reaction (Sup-
plementary Fig. S3d).
RNA sequencing
e poly(A) mRNA fraction from total RNA was isolated with a Dynabeads mRNA DIRECT Micro Kit (ermo).
e sequencing library for each RNA sample was prepared according to the protocol provided by the manu-
facturer using the Ion Total RNA-Seq Kit v2 (ermo). e libraries were generated from 1–15ng of mRNA by
fragmenting the mRNA with RNaseIII, purifying the fragmented RNA, and hybridizing and ligating it with Ion
adaptors. Subsequently, the RNA products were reverse transcribed and amplied to double-stranded cDNA and
then puried using a magnetic bead-based method. e molar concentration and size of each cDNA library were
determined using the DNA HS Kit on a Bioanalyzer 2100 (Agilent). Each library was diluted to ~ 53pM before
template preparation. Up to three barcoded libraries were mixed in equal volume and used for automatic tem-
plate preparation on the Ion Chef instrument (ermo) using reagents from the Ion PI Hi-Q 200 Kit (ermo)
and Ion PI v3 Proton Chip. All samples were sequenced on the Ion Proton System (ermo) according to the
manufacturer’s instructions.
Signal processing and base calling were performed with Torrent Suite version 5.14.0. Raw reads were mapped
to D. rerio Ensembl genome version GRCz11 using STAR (version 2.7.10a)51 and bowtie2 (version 2.4.4)52 for
unmapped reads. Gene counts were created with htseq-count53 using the Ensembl gene model. Dierential
expression was analyzed with DESeq2 version (version 1.40.1). RNA sequencing data were deposited in the GEO
repository (under accession no: GSE232220).
Functional annotation of DEGs (fold change > 1.5 and p value < 0.05) was performed using the R package
ClusterProler version 4.454. Gene lists were searched using the Entrez gene annotation (ENTREZ_GENE_ID),
with the D. rerio background dataset used for analyses. Volcano plots and dot plots were created using the
ggplot2 libraries in R.
Imaging
Each stage of zebrash embryo development was observed and photographed under an inverted microscope
(Leica DMi1 under Flexacam C1). e uorescence signal from mTurquoise-fused protein was observed under
a uorescence stereomicroscope (Zeiss Discovery V12 with a PentaFluar S lter slider equipped with a Zeiss
Axiocam 705 mono camera). e excitation wavelength was 436nm, and the emission wavelength was 480nm.
Statistics
All graphs were created using CorelDRAW 2021 (Corel Corporation), and all statistical analyses, including
unpaired t-test and one-way ANOVA followed by Tukey`s multiple comparisons test, were performed using
GraphPad Prism 8 (GraphPad Soware).
Data availability
Sequencing data have been deposited in NCBI’s Gene Expression Omnibus and are accessible through GEO
Series accession number GSE232220. Any additional data are available from the corresponding author upon
reasonable request.
Received: 25 May 2023; Accepted: 5 October 2023
References
1. Murphy, C. R. Uterine receptivity and the plasma membrane transformation. Cell Res. 14, 259–267 (2004).
2. Mor, G., Cardenas, I., Abrahams, V. & Guller, S. Inammation and pregnancy: e role of the immune system at the implantation
site. Ann. N Y Acad. Sci. 1221, 80–87 (2011).
3. Kover, K., Liang, L., Andrews, G. K. & Dey, S. K. Dierential expression and regulation of cytokine genes in the mouse uterus.
Endocrinology 136, 1666–1673 (1995).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
Vol:.(1234567890)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
4. Vilella, F. et al. PGE2 and PGF2α concentrations in human endometrial uid as biomarkers for embryonic implantation. J. Clin.
Endocrinol. Metab. 98, 4123–4132 (2013).
5. Grith, O. W. et al. Embryo implantation evolved from an ancestral inammatory attachment reaction. Proc. Natl. Acad. Sci. USA
114, E6566–E6575 (2017).
6. Hall, M. R. & Gracey, A. Y. Single-larva RNA sequencing identies markers of copper toxicity and exposure in early mytilus
californianus larvae. Front Physiol 12, 647482 (2021).
7. Kinoshita, N. et al. Mechanical stress regulates epithelial tissue integrity and stiness through the FGFR/Erk2 signaling pathway
during embryogenesis. Cell R ep. 30, 3875-3888.e3 (2020).
8. Mizgalska, D. et al. Interleukin-1-inducible MCPIP protein has structural and functional properties of RNase and participates in
degradation of IL-1β mRNA. FEBS J. 276, 7386–7399 (2009).
9. Matsushita, K. et al. Zc3h12a is an RNase essential for controlling immune responses by regulating mRNA decay. Nature 458,
1185–1190 (2009).
10. Iwasaki, H. et al. e IκB kinase complex regulates the stability of cytokine-encoding mRNA induced by TLR-IL-1R by controlling
degradation of regnase-1. Nat. Immunol. 12, 1167–1175 (2011).
11. Li, M. et al. MCPIP1 down-regulates IL-2 expression through an ARE-independent pathway. PLoS One 7, e49841 (2012).
12. Suzuki, H. I. et al. MCPIP1 ribonuclease antagonizes dicer and terminates microRNA biogenesis through precursor microRNA
degradation. Mol. Cell 44, 424–436 (2011).
13. Blazusiak, E., Florczyk, D., Jura, J., Potempa, J. & Koziel, J. Dierential regulation by toll-like receptor agonists reveals that MCPIP1
is the potent regulator of innate immunity in bacterial and viral infections. J. Innate Immun. 5, 15–23 (2013).
14. Liang, J. et al. MCP-induced protein 1 deubiquitinates TRAF proteins and negatively regulates JNK and NF-kappaB signaling. J.
Exp. Med. 207, 2959–2973 (2010).
15. Skalniak, L. et al. Regulatory feedback loop between NF-kappaB and MCP-1-induced protein 1 RNase. FEBS J. 276, 5892–5905
(2009).
16. Suk, F. M. et al. Mcpip1 enhances tnf-α-mediated apoptosis through downregulation of the nf-κb/cip axis. Biology (Basel) 10,
655 (2021).
17. Niu, J. et al. USP10 inhibits genotoxic NF-κB activation by MCPIP1-facilitated deubiquitination of NEMO. EMBO J. 32, 3206–3219
(2013).
18. Boratyn, E. et al. MCPIP1 overexpression in human neuroblastoma cell lines causes cell-cycle arrest by G1/S checkpoint block. J.
Cell Biochem. 121, 3406–3425 (2020).
19. Lu, W. et al. MCPIP1 selectively destabilizes transcripts associated with an antiapoptotic gene expression program in breast cancer
cells that can elicit complete tumor regression. Cancer Res. 76, 1429–1440 (2016).
20. Labedz-Maslowska, A. et al. Monocyte chemoattractant protein-induced protein 1 (MCPIP1) enhances angiogenic and cardio-
myogenic potential of murine bone marrow-derived mesenchymal stem cells. PLoS One 10, e0133746 (2015).
21. Roy, A., Zhang, M., Saad, Y. & Kolattukudy, P. E. Antidicer RNAse activity of monocyte chemotactic protein-induced protein-1 is
critical for inducing angiogenesis. Am. J. Physiol. Cell Physiol. 305, C1021–C1032 (2013).
22. Lipert, B. et al. Monocyte chemoattractant protein-induced protein 1 impairs adipogenesis in 3T3-L1 cells. Biochim. Biophys. Acta
Mol Cell Res 1843, 780–788 (2014).
23. Yang, S. et al. e evolution and immunomodulatory role of Zc3h12 proteins in zebrash (Danio rerio). Int. J. Biol. Macromol.
239, 124214 (2023).
24. Kimmel, C. B., Ballard, W. W., Kimmel, S. R., Ullmann, B. & Schilling, T. F. Stages of embryonic development of the zebrash. Dev.
Dyn. 203, 253–310 (1995).
25. White, R. J. et al. A high-resolution mRNA expression time course of embryonic development in zebrash. Elife 6, e30860 (2017).
26. Xie, Y., Meijer, A. H. & Schaaf, M. J. M. Modeling inammation in zebrash for the development of anti-inammatory drugs.
Front. Cell Dev. Biol. 8, 620984 (2020).
27. D’Costa, A. & Shepherd, I. T. Zebrash development and genetics: introducing undergraduates to developmental biology and
genetics in a large introductory laboratory class. Zebrash 6, 169–177 (2009).
28. Uehata, T. & Akira, S. MRNA degradation by the endoribonuclease Regnase-1/ZC3H12a/MCPIP-1. Biochimica et Biophysica
Acta - Gene Regulatory Mechanisms 1829, 708–713 (2013).
29. Mathavan, S. et al. Transcriptome analysis of zebrash embryogenesis using microarrays. PLoS Genet. 1, 260–276 (2005).
30. Qi, D. et al. Monocyte chemotactic protein-induced protein 1 (MCPIP1) suppresses stress granule formation and determines
apoptosis under stress. J. Biol. Chem. 286, 41692–41700 (2011).
31. Bugara, B. et al. MCPIP1 contributes to the inammatory response of UVB-treated keratinocytes. J. Dermatol. Sci. 87, 10–18
(2017).
32. Zhu, T. et al. e Role of MCPIP1 in ischemia/reperfusion injury-induced HUVEC migration and apoptosis. Cell Physiol Biochem
37, 577–591 (2015).
33. Oh, Y. T., Qian, G., Deng, J. & Sun, S. Y. Monocyte chemotactic protein-induced protein-1 enhances DR5 degradation and nega-
tively regulates DR5 activation-induced apoptosis through its deubiquitinase function. Oncogene 37, 3415–3425 (2018).
34. Dobosz, E. et al. MCPIP-1 restricts inammation via promoting apoptosis of neutrophils. Front. Immunol. 12, 627922 (2021).
35. Szegezdi, E., Logue, S. E., Gorman, A. M. & Samali, A. Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep.
7, 880–885 (2006).
36. Hoyle, N. & Ish-Horowicz, D. Transcript processing and export kinetics are rate-limiting steps in expressing vertebrate segmenta-
tion clock genes. Proc. Natl. Acad. Sci. USA 12, 4316–4324 (2013).
37. Ferjentsik, Z. et al. Notch is a critical component of the mouse somitogenesis oscillator and is essential for the formation of the
somites. PLoS Genet 5, e1000662 (2009).
38. Krebs, L. T. et al. e Notch-regulated ankyrin repeat protein is required for proper anterior-posterior somite patterning in mice.
Genesis 50, 366–374 (2012).
39. Pézeron, G. et al. Rasl11b knock down in zebrash suppresses one-eyed-pinhead mutant phenotype. PLoS One 3, e1434 (2008).
40. Notarnicola, C., Le Guen, L., Fort, P., Faure, S. & De Santa Barbara, P. D ynamic expression patterns of RhoV/Chp and RhoU/Wrch
during chicken embryonic development. Dev. Dyn. 237, 1165–1171 (2008).
41. Fort, P. et al. Activity of the RhoU/Wrch1 GTPase is critical for cranial neural crest cell migration. Dev. Biol. 350, 451–463 (2011).
42. N. Behrens, A. Nkx2–5 Regulates Tdgf1 (Cripto) Early During Cardiac Development. J. Clin. Exp. Cardiol. 1, 1–4 (2013).
43. Pei, W. et al. An early requirement for maternal FoxH1 during zebrash gastrulation. Dev. Biol. 310, 10–22 (2007).
44. McGraw, H. F., Nechiporuk, A. & Raible, D. W. Zebrash dorsal root ganglia neural precursor cells adopt a glial fate in the absence
of Neurogenin1. J. Neurosci. 28, 12558–12569 (2008).
45. Altschul, S. F. et al. Gapped BLAST and PSI-BLAST: A new generation of protein database search programs. Nucl. Acids Res. 25,
3389–3402 (1997).
46. Tamura, K., Stecher, G. & Kumar, S. MEGA11: Molecular evolutionary genetics analysis version 11. Mol Biol Evol 38, 3022–3027
(2021).
47. Nusslein-Volhard, C. & Dahm, R. Zebrash: A Practical Approach. (Oxford University Press, 2002).
48. Gökirmak, T. et al. Localization and substrate selectivity of sea urchin multidrug (MDR) eux transporters. J. Biol. Chem. 287,
43876–43883 (2012).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
11
Vol.:(0123456789)
Scientic Reports | (2023) 13:16944 | https://doi.org/10.1038/s41598-023-44294-1
www.nature.com/scientificreports/
49. van de Pol, I. L. E., Flik, G. & Verberk, W. C. E. P. Triploidy in zebrash larvae: Eects on gene expression, cell size and cell number,
growth, development and swimming performance. PLoS One 15, e0229468 (2020).
50. Tang, R., Dodd, A., Lai, D., McNabb, W. C. & Love, D. R. Validation of zebrash (Danio rerio) reference genes for quantitative
real-time RT-PCR normalization. Acta Biochim Biophys Sin (Shanghai) 39, 384–390 (2007).
51. Dobin, A. et al. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
52. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).
53. Anders, S., Pyl, P. T. & Huber, W. HTSeq-A Python framework to work with high-throughput sequencing data. Bioinformatics 31,
166–169 (2015).
54. Wu, T. et al. clusterProler 4.0: A universal enrichment tool for interpreting omics data. Innovation (Cambridge (Mass)) 2, 1041
(2021).
Acknowledgements
We would like to thank Dariusz Gajdzinski for his help with zebrash maintenance and breeding. We also thank
Mateusz Wilamowski for his help with cloning.
Author contributions
A.L.-C., J.J., T.K.P., K.R. and M.C. designed the study. A.L.-C. and W.S. performed the experiments and analysed
data. N.P. performed microinjection. M.K., M.D and M.M. performed RNASeq analyses. A.L.-C. and J.J. wrote
the main manuscript text; W.S., K.R., M.C. and T.K.P. edited it. W.S. prepared gures. A.L.-C., W.S. and T.K.P.
contributed equally to this work. All authors reviewed and approved the manuscript.
Funding
e research for this publication has been funded by MNS 5/2021 (to W.S.). T.K.P. was supported by Pol-
ish National Agency for Academic Exchange under Polish Returns 2019 project (Grant No.: PPN/
PPO/2019/1/00029/U/0001). Open access publication of this article was funded by the Ministry of Education
subsidy.
Competing interests
e authors declare no competing interests.
Additional information
Supplementary Information e online version contains supplementary material available at https:// doi. org/
10. 1038/ s41598- 023- 44294-1.
Correspondence and requests for materials should be addressed to J.J.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, 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. e 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:// creat iveco mmons. org/ licen ses/ by/4. 0/.
© e Author(s) 2023
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
Content uploaded by Weronika Szukała
Author content
All content in this area was uploaded by Weronika Szukała on Oct 08, 2023
Content may be subject to copyright.