Skip to main content

Multifaced roles of PLAC8 in cancer

Abstract

The role of PLAC8 in tumorigenesis has been gradually elucidated with the development of research. Although there are common molecular mechanisms that enforce cell growth, the impact of PLAC8 is varied and can, in some instances, have opposite effects on tumorigenesis. To systematically understand the role of PLAC8 in tumors, the molecular functions of PLAC8 in cancer will be discussed by focusing on how PLAC8 impacts tumorigenesis when it arises within tumor cells and how these roles can change in different stages of cancer progression with the ultimate goal of suppressing PLAC8-relevant cancer behavior and related pathologies. In addition, we highlight the diversity of PLAC8 in different tumors and its functional output beyond cancer cell growth. The comprehension of PLAC8’s molecular function might provide new target and lead to the development of novel anticancer therapies.

Introduction

Placenta specific 8 (PLAC8), also known as Onzin, C15, DGIC and PNAS-144, was first identified in genome-wide expression profiling of mid-gestation placentas and embryos using a 15,000 mouse-developmental cDNA microarray [1, 2]. PLAC8 expression is dynamic during pregnancy and placental development and accumulates in an implantation-dependent manner [1, 3]. PLAC8 has also been found to be involved in embryo development [4,5,6,7,8]. And PLAC8 is found to be highly expressed in the endometrium of pregnant cows compared to nonpregnant cows, and it is upregulated in blastocysts, resulting in calf delivery [9,10,11,12]. Subsequent research on PLAC8 was not limited to animals but also involved humans and many plants [13,14,15,16]. During the differentiation process of cytotrophoblast cells into interstitial extravillous trophoblast cells, PLAC8 is greatly induced [17]. To date, PLAC8 has been determined to be involved in organ development and tumorigenesis [18,19,20,21]. In addition, PLAC8 is a molecular marker to predict prognosis and distinguish between different cell subpopulations [17, 22]. PLAC8 also plays different roles in a cell- or tissue-type specific manner. Throughout this review, we discuss the structure of PLAC8 and how PLAC8 evokes widely different responses in tumorigenesis.

PLAC8 protein

The PLAC8 gene is located in human chromosome 4 and Mus musculus chromosome 5, which is one of the placenta-regulatory genes and belongs to the cornifelin family.

The PLAC8 protein contains five exons, coding for a mRNA species of 829 bp and an open reading frame of 115 amino acids [1], which shows a high degree of conservation (83%) between humans and mice [1, 23]. In addition, FW2.2-like (FWL) genes which are identified in plant species and PLAC8 genes, which both contain highly conserved cysteine-rich motifs, share a common ancestor before the divergence between plants and animals [24]. The first 11 amino acids of this cysteine-rich domain are reported to be required for binding of PLAC8 with Akt1 and MDM-2 protein, and then regulate the activity of Akt1 and MDM-2 [25]. This same region is also found to be required for PLAC8 transiently binds to the C/EBPβ promoter and induce its transcription [26]. In addition, this cysteine-rich domain is called the PLAC8 motif which does not conform to consensus zinc- or RING-finger domains [27, 28]. The PLAC8 motif-containing proteins form a large family and members which can be found in fungi, algae, higher plants and animals [29, 30]. In plants, AtPCR1 and AtPCR2 which contain PLAC8 motif play an important role in transport of heavy metals such as cadmium or zinc [29]. However, our knowledge about the function of PLAC8 motif-containing proteins is very limited. To some extent, although PLAC8 protein has only 115 amino acids (Fig. 1), investigation of its intact domain will help to provide a full understanding of its function and PLAC8 motif-containing proteins.

Fig. 1
figure 1

PLAC8 structure. The cysteine-rich domain of the human PLAC8 protein is located between amino acids 28 and 61

PLAC8 protein does not have an N-terminal signal peptide, indicating that this protein is not a secretory protein and functions within the cytoplasm or the nucleus [31]. And the precise cellular location of PLAC8 varies greatly depending on its specific context. For instance, the intracellular distribution of the PLAC8 protein is dynamic and regulated in an implantation-dependent manner [32]. PLAC8 is specifically expressed in the interstitial extravillous trophoblast cells on the fetomaternal interface, while its expression is hardly detectable in the endovasculare trophoblast cells [17]. PLAC8 is found exclusively at the apical domain of fully differentiated normal colonic epithelium in both colonocytes and goblet cells [33], and it localizes at the trophoblast cell periphery [17]. In addition, PLAC8 has been found in nasopharyngeal carcinoma and breast cancer cell cytoplasm and membrane [34, 35]. After breast cancer cells acquired drug resistance, PLAC8 accumulated both in nucleus and cytoplasm [36]. In pancreatic cancer cells, PLAC8 is located in the inner plasma membrane [37]. However, in pancreatic ductal adenocarcinoma, PLAC8 is mainly located in lysosomes [38]. The lysosomes contain transporters and participates in the export of molecules [39]. The location of PLAC8 in lysosomes might cause the different location of PLAC8 because of lysosomes interact with other organelles thus leading fusion or non-fusogenic contacts. And these varying localizations may result in its functional differences.

Since PLAC8 was identified 20 years ago, many studies have been performed to identify the characteristics and molecular functions of PLAC8 in cancer (Fig. 2) [40]. PLAC8 promotes the growth of tumor cells in prostate cancer cells [41] but significantly inhibits the growth of tumor cells in hepatocellular carcinoma [42]. This interesting phenomenon prompts us to explore the underlying mechanisms and regulatory network of PLAC8. Therefore, research on PLAC8 will help us to further understand the biological characteristics of tumors.

Fig. 2
figure 2

Timeline of PLAC8 research. A brief history of functional and pharmacological studies of PLAC8

Connections with cancer

As a key regulator of growth in different species, including fungi [43], plants [24, 44] and mammals [3, 30, 45, 46], PLAC8 participates in many important physiological activities in different contexts [31, 47,48,49]. Such as, the ratio of FAIM3:PLAC8 might be a diagnostic biomarker in sepsis [47]. And PLAC8 is related with septic shock [49]. To date, researchers have also found that PLAC8 acts as a tumor associated gene that is involved in many cancer processes (Fig. 3) [50,51,52,53,54,55]. We further discuss the various molecular functions of PLAC8 in cancer in our review.

Fig. 3
figure 3

Schematic overview of PLAC8 functions in cancer progression

Programed cell death

Programmed cell death, referring to apoptosis, autophagy, programmed necrosis and ferroptosis, may jointly decide the fate of malignant neoplasm cells [56,57,58]. These forms of programmed cell death balance cell death with cell survival, thus regulating cancer cell fate. Many oncogenes or tumor suppressor genes are linked with tumorigenesis through programmed cell death [59,60,61]. PLAC8, as an oncogene, promotes colorectal and prostate cancer cell growth [62,63,64]. Cancer growth is always accompanied by programmed cell death. As expected, PLAC8 regulates cell apoptosis in various cancers [65]. We found that PLAC8 inhibits breast cancer cell apoptosis, thus promoting cell proliferation [34]. PLAC8 decreases the sensitivity of lung adenocarcinoma cells to gefitinib-induced apoptosis by reducing the expression of cleaved caspase 3 and cleaved PARP [45]. The mRNA levels of PLAC8 are increased in stool, and that its increased expression correlates with colorectal cells relapse [63, 66]. PLAC8 is also upregulated in late-stage colorectal patient’s tissues and butyrate which produces microorganisms downregulated PLAC8 expression. And butyrate increased cleaved PARP fragment and then induced apoptosis in colorectal cells [62]. Exception of cancer cells, PLAC8 can also inhibits cell apoptosis of primary human and established rat fibroblasts via promoting the activation of MDM-2 and AKT1 and then inhibiting p53 [25]. Akt/MDM-2/p53 pathway serves an important role in the regulation of cell apoptosis [67]. And autophagy, is a process that delivers cytoplasmic components to the lysosomes which PLAC8 locates in [38], has opposing and context-dependent roles in cancer [68]. Autophagy induces pancreatic ductal adenocarcinoma cells growth [69]. Pancreatic ductal adenocarcinoma has signature oncogenic mutations of KRAS and the inactivation of p53 [70]. Additionally, in pancreatic ductal adenocarcinoma cell lines, PLAC8 is cooperatively induced in response to mutations in KRAS [71] and p53 [72] which are the two of the most commonly occurring mutations in cancer. And then PLAC8 promote pancreatic ductal adenocarcinoma cell lines autophagy thus promoting tumor formation [38]. The oncogenic role of PLAC8 in inducing the prosurvival function of autophagy protects cells from environmental stress and aids in the transformation of prostate epithelial cells during chronic exposure to cadmium [41]. We previously shown that PLAC8 collaborates with p62 to suppress autophagy in doxorubicin resistant breast cancer cells [36]. PLAC8 inhibits autophagy via the AKT/mTOR pathway in nasopharyngeal carcinoma cells [73]. In addition to cancer, PLAC8 also enhances autophagy in adult-onset Still’s disease [74] and promotes trophoblast cells autophagy though regulating autophagy-related markers, including LC3B I/II, ATG12 and Beclin-1 [75]. However, the relationship of PLAC8 with programmed necrosis and ferroptosis, which is a new form of cell death, is still unknown. We previously discussed that an interaction exists between ferroptosis and autophagy [76]. The crosstalk between autophagy and apoptosis regulates testicular injury induced by cadmium via PI3K and a mTOR-independent pathway [77]. Interestingly, PLAC8 regulates the PI3K pathway and interacts with AKT, which is an important kinase of the PI3K pathway [34, 42, 78]. These results strongly indicate that PLAC8 may be a core regulator in programmed cell death, affect different forms of cell death and decide cancer cell fate. This intriguing contrast in the effects of PLAC8 on cell fate in different cellular contexts presents attractive possibilities for the development of novel therapies for cancers.

Cancer stemness

Stem cells are a population of undifferentiated cells characterized by the ability of self-renewal, such as embryonic stem cells. Studies have shown that the expression of PLAC8 and several recognized stem cell markers (NANOG [79], SOX2 [80] and POU5F1 [81]) are commonly highly expressed in embryo development [82]. In POU5F1-null embryonic stem cells, PLAC8 is downregulated [83]. PLAC8 also may be upstream of KLF4 which is a stem cell marker [84] in triggering adipogenesis [51]. These studies suggest that PLAC8 may involve in stem cell progression vis interacting with stem cell markers. Consistant with stem cells, cancer stem cells (CSCs) have the potential to self-renew, and they often appear dormant and resist cancer treatments, such as radiation and chemotherapy, leading to cancer recurrence. Higher PLAC8 expression is found in the sphere-forming colorectal cancer cells than in colorectal cancer cells. And Id1 gene which can activate the Wnt/β-catenin and Shh signaling pathways promote PLAC8 expression and then maintains cell stemness in colorectal cancer [85]. In non-small cell lung cancer, PLAC8 promotes the levels of ALDH1A1 which is a putative marker for CSCs in numerous types of tumors [86,87,88]. Additionally, PLAC8 regulates the expression of POU5F1, thus increasing stemness during lung adenocarcinoma cell resistance to radiotherapy [89]. And our previous study showed that KLF4 regulates PLAC8 transcription in lung cancer cells [90]. These studies strongly indicates that the regulation loop between stem cell markers (POU5F1 and KLF4) and PLAC8 and the various roles of PLAC8 in cancer stemness. The precise association of PLAC8 with recognized stem cell markers still need further explored. Based on emerging evidence, PLAC8 may be a promising stemness related marker in tumor initiation and development.

Epithelial-mesenchymal transition

Epithelial–mesenchymal transition (EMT) is a cellular process in which cells lose their epithelial characteristics and acquire mesenchymal features that have been associated with metastasis [91]. Studies have shown that PLAC8 overexpression contributes to MAPK pathway activation and metastatic phenotypes [92] and that PLAC8 plays a role in the epithelial-mesenchymal transition [93] in different types of cancer. PLAC8 promotes trophoblast cell, non-small cell lung cancer cell, and clear cell renal cell carcinoma invasion and migration [17, 88, 94, 95]. However, PLAC8 inhibits oral squamous cell invasion [95]. PLAC8 reflects the expression of epithelial-mesenchymal related markers including E-cadherin, N-cadherin and vimentin thus involving epithelial-mesenchymal transition process. In breast cancer cells, embryonic kidney 293 T cells, colorectal cancer cells and nasopharyngeal carcinoma cells, PLAC8 downregulates the level of E-cadherin thus regulating cell migration and invasion [34, 35, 96, 97]. On the other hand, PLAC8 upregulates N-cadherin and vimentin levels in breast cancer and nasopharyngeal carcinoma cells [34, 73]. Interestingly, PLAC8 decreases E-cadherin expression but increases P-cadherin and vimentin expression; however, the level of N-cadherin is stable in colorectal cancer cells [33]. These studies demonstrate that the molecular function of PLAC8 varies in different contexts. The difference in cellular position may not be sufficient to explain this phenomenon, and in-depth research is needed in the future. In addition to cadherin family proteins, the abundant expression of PLAC8 in interstitial extravillous trophoblast cells promotes cell invasion and migration partially by upregulating the activation of RAC1 and CDC42 without change their expression [17]. PLAC8 not only promotes EMT progression but is also involved in cancer metastasis, such as bone metastasis in prostate cancer cells and lung metastasis in colorectal cancer cells in vivo [62, 64]. Taken together, PLAC8 may reflect epithelial-mesenchymal related genes thus involving EMT progression and cancer metastasis. Additionally, the expression of PLAC8 can predict of changes in EMT markers, including E-cadherin, N-cadherin and vimentin and be the hallmark of EMT progression.

Cancer immunity

PLAC8 exists in a variety of immune cells and the level of PLAC8 varies in different immune cells. PLAC8 is higher expressed by Th1 CD4 T-cells compared to Th2, Th17 and iTreg CD4 T-cells [22]. In addition, PLAC8 is relatively highly expressed in airway T helper 2 (Th2) cells which play a pathogenic role in allergies [98]. PLAC8 is robustly downregulated in CD39+ human regulatory T-cells [99]. In addition to being expressed in immune cells, PLAC8 also interacts with immune factors and regulates inflammation. For example, PLAC8 suppresses the production of the pro-inflammatory cytokines, IL-1b and IL-18, via enhancement of autophagy in adult-onset Still’s disease [74]. PLAC8 is important for suppressing IFNγ production by IL-12 stimulation in CD4 T-cell [22]. And CD4 T-cell expression of PLAC8 correlates with potent termination of Chlamydia replication and relative independence from IFNγ pretreatment of epithelial monolayers [100, 101]. And Chlamydia-specific CD8 T-cell clones do not express PLAC8 [102], but PLAC8 also promotes effector CD8 T-cell establishment through a T cell-intrinsic mechanism. In addition, PLAC8 is identified in placental functions, and PLAC8 is relatively higher in placentitis cells [103]. PLAC8 mRNA is also increased in the myometrium of adenomyosis patients, indicating the role of the immune response in the myometrium of women with adenomyosis [104]. These evidences suggest that PLAC8 may play an important role in immune system [31, 105, 106]. Determining factors that regulate PLAC8 expression in T cells may help to identify how it can be utilized therapeutically during T cell-driven inflammation, and the functions of PLAC8 in the immune system, especially in the regulation of different populations of immune cells, need to be explored further.

When referred to cancer immunity, PLAC8 is found to be most intensively expressed in the FXIII-A dim subgroup and helps to define three novel subpopulations in pediatric B-cell progenitor acute lymphoblastic leukemia [107]. And RNA sequencing data of clear cell renal cell carcinoma has shown that PLAC8 is mainly involved in immunity-related pathways [94]. With unbiased RNA sequence analysis, CXCL5, which is an inflammatory mediator, has been identified as one of the downstream targets of PLAC8 overexpression in osteosarcoma [92]. Gong et al. found that PLAC8 is abnormally overexpressed in gallbladder carcinoma cells and that its expression positively correlates with PD-L1 expression, which is the main checkpoint of the immune system [108]. However, time and more research will begin to address questions that how PLAC8 involves cancer immunity. While these findings were initially unexpected, PLAC8 is an immune-related gene and may be a targeting gene for immune reactions in cancer.

Drug resistance

In the ericoid mycorrhizal fungus, Oidiodendron maius, PLAC8-containing proteins have been reported to be involved in cadmium tolerance [28]. Additionally, specifically targeting PLAC8 may affect prostate carcinogenesis in humans, and PLAC8 activation may be used as a biomarker for the early detection of prostate cancer in cadmium-exposed populations [41]. These findings indicate that the expression of PLAC8 might be altered upon exposure to certain drugs. Drug resistance is one of the main reasons for the failure of tumor therapy, which greatly limits the choice and use of cancer drugs. Researchers have demonstrated that PLAC8 is related to multidrug resistance in various cancers. In nasopharyngeal carcinoma cells, knockout of PLAC8 radiosensitizes nasopharyngeal carcinoma cells by activating the PI3K/AKT/GSK3β pathway [78]. Our study found that overexpression of PLAC8 can promote tamoxifen resistance in breast cancer and that the expression of PLAC8 can be reduced by curcumin [96]. In addition to endocrine resistance, PLAC8 regulates RAC1 levels, and another study has reported that RAC1 promotes breast cancer chemoresistance by influencing DNA damage repair [17, 109]. These findings indicate that PLAC8 may predict multidrug resistance in breast cancer. In non-small cell lung cancer, overexpression of PLAC8 in parental cells markedly decreases osimertinib sensitivity [88]. Enhanced sensitivity to cisplatin treatment following silencing of PLAC8 in clear cell renal cell carcinoma cells suggests a potential therapeutic target of PLAC8 [94]. PLAC8 overexpression decreases sensitivity to gemcitabine and oxaliplatin in gallbladder carcinoma cells [108]. Overexpression of PLAC8 significantly decreases the sensitivity of lung adenocarcinoma to gefitinib [45]. Taken together, these results suggest that PLAC8 may predict drug resistance in various cancer cells and be a promising therapeutic target.

Other diseases

In addition to its important role in tumors, PLAC8 also participates in other disease processes, such as respiratory diseases and some infectious diseases [98, 102, 110]. For example, PLAC8 is upregulated in activated monocytes and in monocytes isolated from active ASD patients [74]. In addition, many studies have shown that PLAC8 is related to glucose metabolism [26]. However, animal models have shown that PLAC8 is expressed at different levels in F344-fa and F344-fa-nidd2 rats and is closely related to obesity and glucose loading [15]. The AIM3:PLAC8 ratio is a candidate biomarker that can be used to assist in the rapid diagnosis of CAP on ICU admission [111]. The study of PLAC8 in different systemic diseases in humans may help to further understand the function of this gene.

Overview of the PLAC8-regulated network

There is mounting evidence of the potential role of PLAC8-regulated network in cancer (Fig. 4) [104, 111, 112]. PLAC8 can be regulated at the transcriptional level. For example, PLAC8 is involved in pro-mesonephros regulation, and PAX2 regulates the transcription of PLAC8 [113]. PLAC8 is upregulated by IFNT [114], and the expression of PLAC8 is upregulated under hypoxia [17]. PLAC8 acts as a transcription factor involved in the expression of different genes. In CD4 T cells, PLAC8 suppresses IL-12-induced IFNγ production at the transcriptional level [22]. PLAC8 binds to the C/EBPβ promoter to induce its transcription [26]. PLAC8 activates the Akt/MDM-2 pathway, ultimately leading to an inability to upregulate p53. In addition, PLAC8 directly interacts with MDM-2 and Akt, thereby influencing the localization of both proteins [25]. In functional extravillous trophoblasts, PLAC8 colocalizes with p53 and regulates p53 expression at the posttranslational level [75]. In addition, the expression of PLAC8 can be reduced by curcumin in tamoxifen resistant breast cancer [96]. And butyrate reduced the expression of PLAC8 in colorectal cancer cells [62]. In acute myeloid leukemic cell lines, all-trans retinoic acid (ATRA) and phorbol 12-myristate 13-acetate (PMA) downregulate PLAC8 expression though PKCɛ-ERK2 signaling pathway [50]. As shown in Fig. 3, PLAC8 interacts with tumor-related genes both at the transcriptional and posttranscriptional levels, thereby playing a functional role in cancer progression.

Fig. 4
figure 4

Signaling pathways and genes controlling PLAC8 expression and its regulatory system. PLAC8 regulation is driven by different factors in both the nucleus and cytoplasm. It is important to point out that published mechanisms of PLAC8 regulation are not yet completely understood. Studies have shown that growth-related signaling pathways, such as the AKT, MAPK and TGF-β/Smad pathways, interact with PLAC8. Some drugs, such as curcumin and PAM, directly and indirectly affect PLAC8 levels. In addition, PLAC8, as a transcription factor, promotes C/EBPβ transcription and inhibits PU.1 transcription. The dashed lines depict mechanisms that are not completely understood. C/EBPβ, enhancer-binding protein β; ALDH1A1, aldehyde dehydrogenase 1 family member A1; CDC42, cell division control protein 42; POU5F1, POU Class 5 homeobox 1; RAC1, ras-related C3 botulinum toxin substrate 1; KLF4, Kruppel-like factor-4; PLAC8, placenta-specific gene 8; PU.1, Spi-1 proto-oncogene; CD98, ectonucleoside triphosphate diphosphohydrolase 1; ID1, inhibitor of differentiationId-1; PKCɛ, protein kinase C ɛ; ERK2, extracellular regulated protein kinases 2; c-Myc, cellular myelocytomatosis viral oncogene; CXCL5, C-X-C motif chemokine 5; DUSP6, dual specificity phosphatase 6; MDM-2, murine double minute 2; p53, tumor protein 53

Conclusion and perspectives

Our understanding of the molecular mechanisms of PLAC8 has expanded over the last decade, and this knowledge has been used to build better models that allow us to unravel the complicated role of the PLAC8 gene in human diseases. Furthermore, these studies have led to the identification of putative therapeutics to target PLAC8. While PLAC8 accumulates in most tumor cells, it tends to contribute to tumor progression by inducing tumorigenesis, immune reactions, chemoresistance and metastasis. As discussed above, PLAC8 has been identified in breast cancer, prostate cancer, lung cancer gallbladder cancer and nasopharyngeal cancer (Fig. 5). The molecular functions of PLAC8 in the brain, gastric carcinoma and osteocarcinoma remain unknown and need to be explored. Based on these studies, we suggest that PLAC8 may be a promising marker and predictor for clinical drug selection, immunotherapy response and tumor prognosis. The precise roles of PLAC8 in different cancers vary, and its underlying mechanisms should be determined in the future. In addition, the relative network related to PLAC8 is still not clear. Therefore, the mechanisms by which PLAC8 selects its downstream partners and is reflected by other genes may reveal new players and mechanisms by which PLAC8 orchestrates cancer cell behavior, thereby suggesting new targets for therapy. Another aspect that deserves attention is to understand the functional structure of each region of the PLAC8 protein, which will help to comprehend the related molecular mechanism of the protein. Further characterization of the PLAC8 protein in different cell types is paramount not only to enrich our understanding of this gene in normal physiology but also to enhance our ability to target it to reduce cancer progression. Thus, the precise roles of PLAC8 in different forms of programmed cancer death need to be discovered in the future.

Fig. 5
figure 5

Epidemiological data and functional evidence of PLAC8 in tumor types

Availability of data and materials

Not applicable.

References

  1. Galaviz-Hernandez C, Stagg C, de Ridder G, Tanaka TS, Ko MSH, Schlessinger D, et al. Plac8 and Plac9, novel placental-enriched genes identified through microarray analysis. Gene. 2003;309(2):81–9. https://doi.org/10.1016/S0378-1119(03)00508-0.

    Article  CAS  PubMed  Google Scholar 

  2. Tanaka TS, Jaradat SA, Lim MK, et al. Genome-wide expression profiling of mid-gestation placenta and embryo using a 15,000 mouse developmental cDNA microarray. Proc Natl Acad Sci U S A. 2000;97(16):9127–32.

    Article  Google Scholar 

  3. El-Sheikh Ali H, Scoggin K, Linhares Boakari Y, et al. Kinetics of placenta-specific 8 (PLAC8) in equine placenta during pregnancy and placentitis. Theriogenology. 2021;160:81–9. https://doi.org/10.1016/j.theriogenology.2020.10.041.

    Article  CAS  PubMed  Google Scholar 

  4. Lopera-Vasquez R, Hamdi M, Fernandez-Fuertes B, et al. Extracellular vesicles from BOEC in in vitro embryo development and quality. PLoS One. 2016;11(2):e0148083. https://doi.org/10.1371/journal.pone.0148083.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Machado GM, Caixeta ES, Lucci CM, Rumpf R, Franco MM, Dode MA. Post-hatching development of bovine embryos in vitro: the effects of tunnel preparation and gender. Zygote. 2012;20(2):123–34. https://doi.org/10.1017/S0967199411000086.

    Article  CAS  PubMed  Google Scholar 

  6. Machado GM, Ferreira AR, Guardieiro MM, Bastos MR, Carvalho JO, Lucci CM, et al. Morphology, sex ratio and gene expression of day 14 in vivo and in vitro bovine embryos. Reprod Fertil Dev. 2013;25(4):600–8. https://doi.org/10.1071/RD11282.

    Article  CAS  PubMed  Google Scholar 

  7. Hoelker M, Rings F, Lund Q, Ghanem N, Phatsara C, Griese J, et al. Effect of the microenvironment and embryo density on developmental characteristics and gene expression profile of bovine preimplantative embryos cultured in vitro. Reproduction. 2009;137(3):415–25. https://doi.org/10.1530/REP-08-0370.

    Article  CAS  PubMed  Google Scholar 

  8. Gomez E, Caamano JN, Bermejo-Alvarez P, et al. Gene expression in early expanded parthenogenetic and in vitro fertilized bovine blastocysts. J Reprod Dev. 2009;55(6):607–14.

    Article  CAS  Google Scholar 

  9. El-Sayed A, Hoelker M, Rings F, et al. Large-scale transcriptional analysis of bovine embryo biopsies in relation to pregnancy success after transfer to recipients. Physiol Genomics. 2006;28(1):84–96. https://doi.org/10.1152/physiolgenomics.00111.2006.

    Article  CAS  PubMed  Google Scholar 

  10. Gomez E, Gutierrez-Adan A, Diez C, et al. Biological differences between in vitro produced bovine embryos and parthenotes. Reproduction. 2009;137(2):285–95. https://doi.org/10.1530/REP-08-0220.

    Article  CAS  PubMed  Google Scholar 

  11. Lazzari G, Colleoni S, Duchi R, Galli A, Houghton FD, Galli C. Embryonic genotype and inbreeding affect preimplantation development in cattle. Reproduction. 2011;141(5):625–32. https://doi.org/10.1530/REP-10-0282.

    Article  CAS  PubMed  Google Scholar 

  12. Bermejo-Alvarez P, Lonergan P, Rath D, Gutierrez-Adan A, Rizos D. Developmental kinetics and gene expression in male and female bovine embryos produced in vitro with sex-sorted spermatozoa. Reprod Fertil Dev. 2010;22(2):426–36. https://doi.org/10.1071/RD09142.

    Article  CAS  PubMed  Google Scholar 

  13. Sultana N, Islam S, Juhasz A, Yang R, She M, Alhabbar Z, et al. Transcriptomic study for identification of major nitrogen stress responsive genes in Australian bread wheat cultivars. Front Genet. 2020;11:583785. https://doi.org/10.3389/fgene.2020.583785.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lee H, Kim JI, Park JS, Roh JI, Lee J, Kang BC, et al. CRISPR/Cas9-mediated generation of a Plac8 knockout mouse model. Lab Anim Res. 2018;34(4):279–87. https://doi.org/10.5625/lar.2018.34.4.279.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Sasaki D, Kotoh J, Watadani R, Matsumoto K. New animal models reveal that coenzyme Q2 (Coq2) and placenta-specific 8 (Plac8) are candidate genes for the onset of type 2 diabetes associated with obesity in rats. Mamm Genome. 2015;26(11–12):619–29. https://doi.org/10.1007/s00335-015-9597-4.

    Article  CAS  PubMed  Google Scholar 

  16. Cebrian-Serrano A, Salvador I, García-Roselló E, Pericuesta E, Pérez-Cerezales S, Gutierrez-Adán A, et al. Effect of the bovine Oviductal fluid onIn VitroFertilization, development and gene expression ofIn vitro-produced bovine blastocysts. Reprod Domest Anim. 2013;48(2):331–8. https://doi.org/10.1111/j.1439-0531.2012.02157.x.

    Article  CAS  PubMed  Google Scholar 

  17. Chang WL, Liu YW, Dang YL, et al. PLAC8, a new marker for human interstitial extravillous trophoblast cells, promotes their invasion and migration. Development. 2018;145(2):dev148932. https://doi.org/10.1242/dev.148932.

  18. Suwik K, Sinderewicz E, Boruszewska D, et al. mRNA expression and role of PPARgamma and PPARdelta in bovine preimplantation embryos depending on the quality and developmental stage. Animals (Basel). 2020;10(12):2358. https://doi.org/10.3390/ani10122358.

  19. Korzekwa AJ, Kotlarczyk AM, Szczepanska AA, Grzyb M, Siergiej A, Woclawek-Potocka I. Antioxidative potential of red deer embryos depends on reproductive stage of hind as a oocyte donor. Animals (Basel). 2020;10(7):1190. https://doi.org/10.3390/ani10071190.

  20. Blue EK, Sheehan BM, Nuss ZV, et al. Epigenetic regulation of placenta-specific 8 contributes to altered function of endothelial Colony-forming cells exposed to intrauterine gestational diabetes mellitus. Diabetes. 2015;64(7):2664–75. https://doi.org/10.2337/db14-1709.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Machado GM, Ferreira AR, Pivato I, Fidelis A, Spricigo JF, Paulini F, et al. Post-hatching development of in vitro bovine embryos from day 7 to 14 in vivo versus in vitro. Mol Reprod Dev. 2013;80(11):936–47. https://doi.org/10.1002/mrd.22230.

    Article  CAS  PubMed  Google Scholar 

  22. Slade CD, Reagin KL, Lakshmanan HG, Klonowski KD, Watford WT. Placenta-specific 8 limits IFNgamma production by CD4 T cells in vitro and promotes establishment of influenza-specific CD8 T cells in vivo. PLoS One. 2020;15(7):e0235706. https://doi.org/10.1371/journal.pone.0235706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tanaka TS, Jaradat SA, Lim MK, Kargul GJ, Wang X, Grahovac MJ, et al. Genome-wide expression profiling of mid-gestation placenta and embryo using a 15,000 mouse developmental cDNA microarray. Proc Natl Acad Sci U S A. 2000;97(16):9127–32. https://doi.org/10.1073/pnas.97.16.9127.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Libault M, Stacey G. Evolution of FW2.2-like (FWL) and PLAC8 genes in eukaryotes. Plant Signal Behav. 2010;5(10):1226–8. https://doi.org/10.4161/psb.5.10.12808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Rogulski K, Li Y, Rothermund K, Pu L, Watkins S, Yi F, et al. Onzin, a c-Myc-repressed target, promotes survival and transformation by modulating the Akt-Mdm2-p53 pathway. Oncogene. 2005;24(51):7524–41. https://doi.org/10.1038/sj.onc.1208897.

    Article  CAS  PubMed  Google Scholar 

  26. Jimenez-Preitner M, Berney X, Uldry M, Vitali A, Cinti S, Ledford JG, et al. Plac8 is an inducer of C/EBPbeta required for brown fat differentiation, thermoregulation, and control of body weight. Cell Metab. 2011;14(5):658–70. https://doi.org/10.1016/j.cmet.2011.08.008.

    Article  CAS  PubMed  Google Scholar 

  27. Nakano M, Iida K, Nyunoya H, Iida H. Determination of structural regions important for ca (2+) uptake activity in Arabidopsis MCA1 and MCA2 expressed in yeast. Plant Cell Physiol. 2011;52(11):1915–30. https://doi.org/10.1093/pcp/pcr131.

    Article  CAS  PubMed  Google Scholar 

  28. Di Vietro L, Daghino S, Abbà S, Perotto S. Gene expression and role in cadmium tolerance of two PLAC8-containing proteins identified in the ericoid mycorrhizal fungus Oidiodendron maius. Fungal Biol. 2014;118(8):695–703. https://doi.org/10.1016/j.funbio.2014.04.011.

    Article  CAS  PubMed  Google Scholar 

  29. Song WY, Hortensteiner S, Tomioka R, Lee Y, Martinoia E. Common functions or only phylogenetically related? The large family of PLAC8 motif-containing/PCR genes. Mol Cells. 2011;31(1):1–7. https://doi.org/10.1007/s10059-011-0024-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cabreira-Cagliari C. Dias NdC, Bohn B, et al. revising the PLAC8 gene family: from a central role in differentiation, proliferation, and apoptosis in mammals to a multifunctional role in plants. Genome. 2018;61(12):857–65. https://doi.org/10.1139/gen-2018-0035.

    Article  CAS  PubMed  Google Scholar 

  31. Pang Q, Gao L, Bai Y, Deng H, Han Y, Hu W, et al. Identification and characterization of a novel multifunctional placenta specific protein 8 in Dugesia japonica. Gene. 2017;613:1–9. https://doi.org/10.1016/j.gene.2017.02.024.

    Article  CAS  PubMed  Google Scholar 

  32. Li M, Liu D, Wang L, Wang W, Wang A, Yao Y. Expression of placenta-specific 8 in human oocytes, embryos, and models of in vitro implantation. Fertil Steril. 2016;106(3):781–9 e782. https://doi.org/10.1016/j.fertnstert.2016.05.018.

    Article  CAS  PubMed  Google Scholar 

  33. Li C, Ma H, Wang Y, Cao Z, Graves-Deal R, Powell AE, et al. Excess PLAC8 promotes an unconventional ERK2-dependent EMT in colon cancer. J Clin Invest. 2014;124(5):2172–87. https://doi.org/10.1172/JCI71103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Mao M, Chen Y, Jia Y, Yang J, Wei Q, Li Z, et al. PLCA8 suppresses breast cancer apoptosis by activating the PI3k/AKT/NF-kappaB pathway. J Cell Mol Med. 2019;23(10):6930–41. https://doi.org/10.1111/jcmm.14578.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Huang M-L, Zou Y, Yang R, Jiang Y, Sheng JF, Han JB, et al. Placenta specific 8 gene induces epithelial-mesenchymal transition of nasopharyngeal carcinoma cells via the TGF-β/Smad pathway. Exp Cell Res. 2019;374(1):172–80. https://doi.org/10.1016/j.yexcr.2018.11.021.

    Article  CAS  PubMed  Google Scholar 

  36. Chen Y, Jia Y, Mao M, et al. PLAC8 promotes adriamycin resistance via blocking autophagy in breast cancer. J Cell Mol Med. 2021.

  37. Kaistha BP, Lorenz H, Schmidt H, Sipos B, Pawlak M, Gierke B, et al. PLAC8 localizes to the inner plasma membrane of pancreatic Cancer cells and regulates cell growth and disease progression through critical cell-cycle regulatory pathways. Cancer Res. 2016;76(1):96–107. https://doi.org/10.1158/0008-5472.CAN-15-0216.

    Article  CAS  PubMed  Google Scholar 

  38. Kinsey C, Balakrishnan V, O'Dell MR, et al. Plac8 links oncogenic mutations to regulation of autophagy and is critical to pancreatic cancer progression. Cell Rep. 2014;7(4):1143–55. https://doi.org/10.1016/j.celrep.2014.03.061.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ballabio A, Bonifacino JS. Lysosomes as dynamic regulators of cell and organismal homeostasis. Nat Rev Mol Cell Biol. 2020;21(2):101–18. https://doi.org/10.1038/s41580-019-0185-4.

    Article  CAS  PubMed  Google Scholar 

  40. Grate LR. Many accurate small-discriminatory feature subsets exist in microarray transcript data: biomarker discovery. BMC Bioinformatics. 2005;6(1):97. https://doi.org/10.1186/1471-2105-6-97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kolluru V, Pal D, Papu John AMS, Ankem MK, Freedman JH, Damodaran C. Induction of Plac8 promotes pro-survival function of autophagy in cadmium-induced prostate carcinogenesis. Cancer Lett. 2017;408:121–9. https://doi.org/10.1016/j.canlet.2017.08.023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zou L, Chai J, Gao Y, Guan J, Liu Q, Du JJ. Down-regulated PLAC8 promotes hepatocellular carcinoma cell proliferation by enhancing PI3K/Akt/GSK3beta/Wnt/beta-catenin signaling. Biomed Pharmacother. 2016;84:139–46. https://doi.org/10.1016/j.biopha.2016.09.015.

    Article  CAS  PubMed  Google Scholar 

  43. Daghino S, Di Vietro L, Petiti L, et al. Yeast expression of mammalian Onzin and fungal FCR1 suggests ancestral functions of PLAC8 proteins in mitochondrial metabolism and DNA repair. Sci Rep. 2019;9(1):6629. https://doi.org/10.1038/s41598-019-43136-3.

  44. Guo M, Rupe MA, Dieter JA, Zou J, Spielbauer D, Duncan KE, et al. Cell number Regulator1 affects plant and organ size in maize: implications for crop yield enhancement and heterosis. Plant Cell. 2010;22(4):1057–73. https://doi.org/10.1105/tpc.109.073676.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zeng X, Liu Q, Yang Y, Jia W, Li S, He D, et al. Placenta-specific protein 8 promotes the proliferation of lung adenocarcinoma PC-9 cells and their tolerance to an epidermal growth factor receptor tyrosine kinase inhibitor by activating the ERK signaling pathway. Oncol Lett. 2019;18(5):5621–7. https://doi.org/10.3892/ol.2019.10911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Mansouri-Attia N, Aubert J, Reinaud P, Giraud-Delville C, Taghouti G, Galio L, et al. Gene expression profiles of bovine caruncular and intercaruncular endometrium at implantation. Physiol Genomics. 2009;39(1):14–27. https://doi.org/10.1152/physiolgenomics.90404.2008.

    Article  CAS  PubMed  Google Scholar 

  47. Sweeney TE, Khatri P. Comprehensive validation of the FAIM3:PLAC8 ratio in time-matched public gene expression data. Am J Respir Crit Care Med. 2015;192(10):1260–1.

    Article  CAS  Google Scholar 

  48. Sweeney TE, Khatri P. Benchmarking Sepsis gene expression diagnostics using public data. Crit Care Med. 2017;45(1):1–10. https://doi.org/10.1097/CCM.0000000000002021.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Tang Y, Yang X, Shu H, Yu Y, Pan S, Xu J, et al. Bioinformatic analysis identifies potential biomarkers and therapeutic targets of septic-shock-associated acute kidney injury. Hereditas. 2021;158(1):13. https://doi.org/10.1186/s41065-021-00176-y.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Wu SF, Huang Y, Hou JK, Yuan TT, Zhou CX, Zhang J, et al. The downregulation of onzin expression by PKCepsilon-ERK2 signaling and its potential role in AML cell differentiation. Leukemia. 2010;24(3):544–51. https://doi.org/10.1038/leu.2009.280.

    Article  CAS  PubMed  Google Scholar 

  51. Jimenez-Preitner M, Berney X, Thorens B. Plac8 is required for white adipocyte differentiation in vitro and cell number control in vivo. PLoS One. 2012;7(11):e48767. https://doi.org/10.1371/journal.pone.0048767.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Reddy RB, Bhat AR, James BL, Govindan SV, Mathew R, DR R, et al. Meta-analyses of microarray datasets identifies ANO1 and FADD as prognostic markers of head and neck Cancer. PLoS One. 2016;11(1):e0147409. https://doi.org/10.1371/journal.pone.0147409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Li H, Wang X, Fang Y, Huo Z, Lu X, Zhan X, et al. Integrated expression profiles analysis reveals novel predictive biomarker in pancreatic ductal adenocarcinoma. Oncotarget. 2017;8(32):52571–83. https://doi.org/10.18632/oncotarget.16732.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Tatura M, Schmidt H, Haijat M, Stark M, Rinke A, Diels R, et al. Placenta-specific 8 is overexpressed and regulates cell proliferation in low-grade human pancreatic neuroendocrine tumors. Neuroendocrinology. 2020;110(1–2):23–34. https://doi.org/10.1159/000500541.

    Article  CAS  PubMed  Google Scholar 

  55. Hung CS, Wang YC, Guo JW, Yang RN, Lee CL, Shen MH, et al. Expression pattern of placenta specific 8 and keratin 20 in different types of gastrointestinal cancer. Mol Med Rep. 2020;21(2):659–66. https://doi.org/10.3892/mmr.2019.10871.

    Article  CAS  PubMed  Google Scholar 

  56. Dixon Scott J, Lemberg Kathryn M, Lamprecht Michael R, et al. Ferroptosis: an Iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72. https://doi.org/10.1016/j.cell.2012.03.042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, et al. Classification of cell death: recommendations of the nomenclature committee on cell death 2009. Cell Death Differ. 2009;16(1):3–11. https://doi.org/10.1038/cdd.2008.150.

    Article  CAS  PubMed  Google Scholar 

  58. D’Arcy MS. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 2019;43(6):582–92. https://doi.org/10.1002/cbin.11137.

    Article  PubMed  Google Scholar 

  59. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res. 2019;29(5):347–64. https://doi.org/10.1038/s41422-019-0164-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Van Opdenbosch N, Lamkanfi M. Caspases in cell death, inflammation, and disease. Immunity. 2019;50(6):1352–64. https://doi.org/10.1016/j.immuni.2019.05.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Newton K, Wickliffe KE, Maltzman A, Dugger DL, Reja R, Zhang Y, et al. Activity of caspase-8 determines plasticity between cell death pathways. Nature. 2019;575(7784):679–82. https://doi.org/10.1038/s41586-019-1752-8.

    Article  CAS  PubMed  Google Scholar 

  62. Huang CC, Shen MH, Chen SK, Yang SH, Liu CY, Guo JW, et al. Gut butyrate-producing organisms correlate to placenta specific 8 protein: importance to colorectal cancer progression. J Adv Res. 2020;22:7–20. https://doi.org/10.1016/j.jare.2019.11.005.

    Article  CAS  PubMed  Google Scholar 

  63. Lee CL, Huang CJ, Yang SH, Chang CC, Huang CC, Chien CC, et al. Discovery of genes from feces correlated with colorectal cancer progression. Oncol Lett. 2016;12(5):3378–84. https://doi.org/10.3892/ol.2016.5069.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Uehara H, Takahashi T, Izumi K. Induction of retinol-binding protein 4 and placenta-specific 8 expression in human prostate cancer cells remaining in bone following osteolytic tumor growth inhibition by osteoprotegerin. Int J Oncol. 2013;43(2):365–74. https://doi.org/10.3892/ijo.2013.1954.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Mourtada-Maarabouni M, Watson D, Munir M, Farzaneh F. T. Williams G. apoptosis suppression by candidate oncogene PLAC8 is reversed in other cell types. Curr Cancer Drug Targets. 2013;13(1):80–91. https://doi.org/10.2174/156800913804486584.

    Article  CAS  PubMed  Google Scholar 

  66. Chang CC, Huang CC, Yang SH, Chien CC, Lee CL, Huang CJ. Data on clinical significance of GAS2 in colorectal cancer cells. Data Brief. 2016;8:82–6. https://doi.org/10.1016/j.dib.2016.05.010.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Xu W, Gao L, Li T, Zheng J, Shao A, Zhang J. Mesencephalic astrocyte-derived neurotrophic factor (MANF) protects against neuronal apoptosis via activation of Akt/MDM2/p53 signaling pathway in a rat model of intracerebral hemorrhage. Front Mol Neurosci. 2018;11:176. https://doi.org/10.3389/fnmol.2018.00176.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17(9):528–42. https://doi.org/10.1038/nrc.2017.53.

    Article  CAS  PubMed  Google Scholar 

  69. Yang S, Wang X, Contino G, Liesa M, Sahin E, Ying H, et al. Pancreatic cancers require autophagy for tumor growth. Genes Dev. 2011;25(7):717–29. https://doi.org/10.1101/gad.2016111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ying H, Dey P, Yao W, Kimmelman AC, Draetta GF, Maitra A, et al. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2016;30(4):355–85. https://doi.org/10.1101/gad.275776.115.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Prior IA, Hood FE, Hartley JL. The frequency of Ras mutations in Cancer. Cancer Res. 2020;80(14):2969–74. https://doi.org/10.1158/0008-5472.CAN-19-3682.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Levine AJ. p53: 800 million years of evolution and 40 years of discovery. Nat Rev Cancer. 2020;20(8):471–80. https://doi.org/10.1038/s41568-020-0262-1.

    Article  CAS  PubMed  Google Scholar 

  73. Huang ML, Qi CL, Zou Y, et al. Plac8-mediated autophagy regulates nasopharyngeal carcinoma cell function via AKT/mTOR pathway. J Cell Mol Med. 2020;24(14):7778–88.

    Article  CAS  Google Scholar 

  74. Segawa S, Kondo Y, Nakai Y, Iizuka A, Kaneko S, Yokosawa M, et al. Placenta specific 8 suppresses IL-18 production through regulation of autophagy and is associated with adult still disease. J Immunol. 2018;201(12):3534–45. https://doi.org/10.4049/jimmunol.1800667.

    Article  CAS  PubMed  Google Scholar 

  75. Feng X, Wei Z, Tao X, et al. PLAC8 promotes the autophagic activity and improves the growth priority of human trophoblast cells. FASEB J. 2021;35(3):e21351. https://doi.org/10.1096/fj.202002075RR.

  76. Zhou Y, Shen Y, Chen C, Sui X, Yang J, Wang L, et al. The crosstalk between autophagy and ferroptosis: what can we learn to target drug resistance in cancer? Cancer Biol Med. 2019;16(4):630–46. https://doi.org/10.20892/j.issn.2095-3941.2019.0158.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wang M, Wang XF, Li YM, Chen N, Fan Y, Huang WK, et al. Cross-talk between autophagy and apoptosis regulates testicular injury/recovery induced by cadmium via PI3K with mTOR-independent pathway. Cell Death Dis. 2020;11(1):46. https://doi.org/10.1038/s41419-020-2246-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Yang R, Tao ZZ, Huang ML, Zheng YF, Dai MY, Zou Y, et al. Knockout of the placenta specific 8 gene radiosensitizes nasopharyngeal carcinoma cells by activating the PI3K/AKT/GSK3beta pathway. Am J Transl Res. 2018;10(2):455–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Pan G, Thomson JA. Nanog and transcriptional networks in embryonic stem cell pluripotency. Cell Res. 2007;17(1):42–9. https://doi.org/10.1038/sj.cr.7310125.

    Article  CAS  PubMed  Google Scholar 

  80. Schaefer T, Lengerke C. SOX2 protein biochemistry in stemness, reprogramming, and cancer: the PI3K/AKT/SOX2 axis and beyond. Oncogene. 2020;39(2):278–92. https://doi.org/10.1038/s41388-019-0997-x.

    Article  CAS  PubMed  Google Scholar 

  81. Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 2005;122(6):947–56. https://doi.org/10.1016/j.cell.2005.08.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Sugimura S, Kobayashi S, Hashiyada Y, Ohtake M, Kaneda M, Yamanouchi T, et al. Follicular growth-stimulated cows provide favorable oocytes for producing cloned embryos. Cell Reprogram. 2012;14(1):29–37. https://doi.org/10.1089/cell.2011.0060.

    Article  CAS  PubMed  Google Scholar 

  83. Fogarty NME, McCarthy A, Snijders KE, Powell BE, Kubikova N, Blakeley P, et al. Genome editing reveals a role for OCT4 in human embryogenesis. Nature. 2017;550(7674):67–73. https://doi.org/10.1038/nature24033.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Yin M, Zhou HJ, Lin C, et al. CD34+KLF4+ stromal stem cells contribute to endometrial regeneration and repair. Cell Rep. 2019;27(9):2709–2724.e2703.

    Article  CAS  Google Scholar 

  85. Sun Y, Lai X, Yu Y, Li J, Cao L, Lin W, et al. Inhibitor of DNA binding 1 (Id1) mediates stemness of colorectal cancer cells through the Id1-c-Myc-PLAC8 axis via the Wnt/β-catenin and Shh signaling pathways</p>. Cancer Manag Res. 2019;11:6855–69. https://doi.org/10.2147/CMAR.S207167.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Wang Q, Jiang J, Ying G, et al. Tamoxifen enhances stemness and promotes metastasis of ERalpha36(+) breast cancer by upregulating ALDH1A1 in cancer cells. Cell Res. 2018;28(3):336–58.

    Article  CAS  Google Scholar 

  87. Mori Y, Yamawaki K, Ishiguro T, Yoshihara K, Ueda H, Sato A, et al. ALDH-dependent glycolytic activation mediates Stemness and paclitaxel resistance in patient-derived spheroid models of uterine endometrial Cancer. Stem Cell Rep. 2019;13(4):730–46. https://doi.org/10.1016/j.stemcr.2019.08.015.

    Article  CAS  Google Scholar 

  88. Fei X, Wang G, Shen H, Gu X. Placenta-specific 8 is a potential novel target for osimertinib resistance in non-small cell lung cancer. Oncol Lett. 2019;18(1):955–61. https://doi.org/10.3892/ol.2019.10344.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Jin Z, Guan L, Xiang GM, Gao BA. Radiation resistance of the lung adenocarcinoma is related to the AKT-Onzin-POU5F1 axis. Biochem Biophys Res Commun. 2018;499(3):538–43. https://doi.org/10.1016/j.bbrc.2018.03.185.

    Article  CAS  PubMed  Google Scholar 

  90. Jia Y, Ying X, Zhou J, Chen Y, Luo X, Xie S, et al. The novel KLF4/PLAC8 signaling pathway regulates lung cancer growth. Cell Death Dis. 2018;9(6):603. https://doi.org/10.1038/s41419-018-0580-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Pastushenko I, Blanpain C. EMT transition states during tumor progression and metastasis. Trends Cell Biol. 2019;29(3):212–26. https://doi.org/10.1016/j.tcb.2018.12.001.

    Article  CAS  PubMed  Google Scholar 

  92. Zhang Y, Hu Q, Li G, Li L, Liang S, Zhang Y, et al. ONZIN upregulation by mutant p53 contributes to osteosarcoma metastasis through the CXCL5-MAPK sign9aling pathway. Cell Physiol Biochem. 2018;48(3):1099–111. https://doi.org/10.1159/000491976.

  93. Qi C, Hong L, Cheng Z, Yin Q. Identification of metastasis-associated genes in colorectal cancer using metaDE and survival analysis. Oncol Lett. 2016;11(1):568–74. https://doi.org/10.3892/ol.2015.3956.

    Article  CAS  PubMed  Google Scholar 

  94. Shi L, Xiao L, Heng B, Mo S, Chen W, Su Z. Overexpression of placenta specific 8 is associated with malignant progression and poor prognosis of clear cell renal cell carcinoma. Int Urol Nephrol. 2017;49(7):1165–76. https://doi.org/10.1007/s11255-017-1578-y.

    Article  CAS  PubMed  Google Scholar 

  95. Wu J, Wang X, Shang A, Vella G, Sun Z, Ji P, et al. PLAC8 inhibits oral squamous cell carcinogenesis and epithelial-mesenchymal transition via the Wnt/beta-catenin and PI3K/Akt/GSK3beta signaling pathways. Oncol Lett. 2020;20(5):128. https://doi.org/10.3892/ol.2020.11989.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Mao M, Hu D, Yang J, Chen Y, Zhang X, Shen J, et al. Regulation of tamoxifen sensitivity by the PLAC8/MAPK pathway axis is antagonized by curcumin-induced protein stability change. J Mol Med. 2021;99(6):845–58. https://doi.org/10.1007/s00109-021-02047-5.

    Article  CAS  PubMed  Google Scholar 

  97. Qin X-H, Wang H-X, Ma L, Shen J, Liu Q-H, Xue L. Knockout of the placenta specific 8 gene affects the proliferation and migration of human embryonic kidney 293T cell. Cell Biochem Biophys. 2019;78(1):55–64. https://doi.org/10.1007/s12013-019-00893-2.

    Article  CAS  PubMed  Google Scholar 

  98. Tibbitt CA, Stark JM, Martens L, Ma J, Mold JE, Deswarte K, et al. Single-cell RNA sequencing of the T helper cell response to house dust mites defines a distinct gene expression signature in airway Th2 cells. Immunity. 2019;51(1):169–184.e5. https://doi.org/10.1016/j.immuni.2019.05.014.

    Article  CAS  PubMed  Google Scholar 

  99. Gerner MC, Ziegler LS, Schmidt RLJ, Krenn M, Zimprich F, Uyanik-Ünal K, et al. The TGF-b/SOX4 axis and ROS-driven autophagy co-mediate CD39 expression in regulatory T-cells. FASEB J. 2020;34(6):8367–84. https://doi.org/10.1096/fj.201902664.

    Article  CAS  PubMed  Google Scholar 

  100. Johnson RM, Kerr MS, Slaven JE. Plac8-dependent and inducible NO synthase-dependent mechanisms clear Chlamydia muridarum infections from the genital tract. J Immunol. 2012;188(4):1896–904. https://doi.org/10.4049/jimmunol.1102764.

    Article  CAS  PubMed  Google Scholar 

  101. Johnson RM, Kerr MS, Slaven JE. Perforin is detrimental to controlling [corrected] C. muridarum replication in vitro, but not in vivo. PLoS One. 2013;8(5):e63340.

    Article  CAS  Google Scholar 

  102. Johnson RM, Kerr MS, Slaven JE. An atypical CD8 T-cell response to Chlamydia muridarum genital tract infections includes T cells that produce interleukin-13. Immunology. 2014;142(2):248–57. https://doi.org/10.1111/imm.12248.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. El-Sheikh Ali H, Dini P, Scoggin K, et al. Transcriptomic analysis of equine placenta reveals key regulators and pathways involved in ascending placentitis†. Biol Reprod. 2021;104(3):638–56. https://doi.org/10.1093/biolre/ioaa209.

    Article  PubMed  Google Scholar 

  104. Zhai J, Li S, Sen S, et al. m (6) A RNA methylation regulators contribute to eutopic endometrium and myometrium dysfunction in adenomyosis. Front Genet. 2020;11:716.

    Article  CAS  Google Scholar 

  105. McHugh L, Seldon TA, Brandon RA, Kirk JT, Rapisarda A, Sutherland AJ, et al. A molecular host response assay to discriminate between Sepsis and infection-negative systemic inflammation in critically ill patients: discovery and validation in independent cohorts. PLoS Med. 2015;12(12):e1001916. https://doi.org/10.1371/journal.pmed.1001916.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Verboom DM, Koster-Brouwer ME, Varkila MRJ, Bonten MJM, Cremer OL. Profile of the SeptiCyte™ LAB gene expression assay to diagnose infection in critically ill patients. Expert Rev Mol Diagn. 2019;19(2):95–108. https://doi.org/10.1080/14737159.2019.1567333.

    Article  CAS  PubMed  Google Scholar 

  107. Gyurina K, Karai B, Ujfalusi A, et al. Coagulation FXIII-A protein expression defines three novel sub-populations in pediatric B-cell progenitor acute lymphoblastic leukemia characterized by distinct gene expression signatures. Front Oncol. 2019;9:1063. https://doi.org/10.3389/fonc.2019.01063.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Gong K, Gong Z-J, Lu P-X, Ni XL, Shen S, Liu H, et al. PLAC8 overexpression correlates with PD-L1 upregulation and acquired resistance to chemotherapies in gallbladder carcinoma. Biochem Biophys Res Commun. 2019;516(3):983–90. https://doi.org/10.1016/j.bbrc.2019.06.121.

    Article  CAS  PubMed  Google Scholar 

  109. Li Q, Qin T, Bi Z, Hong H, Ding L, Chen J, et al. Rac1 activates non-oxidative pentose phosphate pathway to induce chemoresistance of breast cancer. Nat Commun. 2020;11(1):1456. https://doi.org/10.1038/s41467-020-15308-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Ziegler A, Marti E, Summerfield A, Baumann A. Identification and characterization of equine blood plasmacytoid dendritic cells. Dev Comp Immunol. 2016;65:352–7. https://doi.org/10.1016/j.dci.2016.08.005.

    Article  CAS  PubMed  Google Scholar 

  111. Scicluna BP, Klein Klouwenberg PM, van Vught LA, et al. A molecular biomarker to diagnose community-acquired pneumonia on intensive care unit admission. Am J Respir Crit Care Med. 2015;192(7):826–35. https://doi.org/10.1164/rccm.201502-0355OC.

    Article  CAS  PubMed  Google Scholar 

  112. Y-y M, Liu J, Zhu J, et al. The effect of botulinum toxin type a on expression profiling of long noncoding RNAs in human dermal fibroblasts. BioMed Res Int. 2017;2017:1–13.

    Google Scholar 

  113. Boualia SK, Gaitan Y, Tremblay M, Sharma R, Cardin J, Kania A, et al. A core transcriptional network composed of Pax2/8, Gata3 and Lim1 regulates key players of pro/mesonephros morphogenesis. Dev Biol. 2013;382(2):555–66. https://doi.org/10.1016/j.ydbio.2013.07.028.

    Article  CAS  PubMed  Google Scholar 

  114. Cheng Z, Chauhan L, Barry AT, Abudureyimu A, Oguejiofor CF, Chen X, et al. Acute bovine viral diarrhea virus infection inhibits expression of interferon tau-stimulated genes in bovine endometrium. Biol Reprod. 2017;96(6):1142–53. https://doi.org/10.1093/biolre/iox056.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The work was supported by the National Natural Science Foundation of China (No. 81972453 and No. 81972597), Zhejiang Provincial Natural Science Foundation of China (under Grant Nos. LY19H160055, LY19H160059, LY18H160005, and LY20H160026), and Zhejiang Provincial Medical and Health Science and Technology (Youth Talent Program) Project No. 2021RC016.

Author information

Authors and Affiliations

Authors

Contributions

MMS, CYF, JZC and WLB designed the review. MMS and CYF researched the literature and drafted the manuscript. YJJ, XL, CYX, JSW, LZQ, CC, and XZ edited the manuscript. All authors approved the final version of the manuscript.

Corresponding authors

Correspondence to Jichun Zhou or Linbo Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This 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. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mao, M., Cheng, Y., Yang, J. et al. Multifaced roles of PLAC8 in cancer. Biomark Res 9, 73 (2021). https://doi.org/10.1186/s40364-021-00329-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-021-00329-1

Keywords