Next Article in Journal
Head-to-Head Comparison of BRAF/MEK Inhibitor Combinations Proposes Superiority of Encorafenib Plus Trametinib in Melanoma
Next Article in Special Issue
Synthesis and Biological Activity of a VHL-Based PROTAC Specific for p38α
Previous Article in Journal
Mutant C. elegans p53 Together with Gain-of-Function GLP-1/Notch Decreases UVC-Damage-Induced Germline Cell Death but Increases PARP Inhibitor-Induced Germline Cell Death
Previous Article in Special Issue
Targeting Protein Degradation Pathways in Tumors: Focusing on their Role in Hematological Malignancies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Presumed Roles of APRO Family Proteins in Cancer Invasiveness

Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(19), 4931; https://doi.org/10.3390/cancers14194931
Submission received: 15 September 2022 / Revised: 4 October 2022 / Accepted: 6 October 2022 / Published: 8 October 2022
(This article belongs to the Special Issue Targeting Mechanisms of Protein Degradation for Cancer Therapy)

Abstract

:

Simple Summary

Here, we discuss the invasiveness of cancer cells in relation to APRO family proteins on the basis of understanding the function of matrix metalloproteinases (MMPs) and/or exosomes. Although APRO family proteins could regulate cancer invasiveness, alternative consequences might occur due to the distinctive effects of MMPs and/or exosomes containing certain microRNAs. Such knowledge could be of use to bring about novel strategies for cancer therapy.

Abstract

The APRO family members may be involved in the regulation of cell growth, migration, and/or invasion. Although an APRO protein could suppress the invasiveness of several cancer cells, it has been reported that overexpression of the same APRO protein could also promote the invasiveness and/or metastasis of the same cancer cells. In general, the invasiveness of cancer cells might be associated with the function of matrix metalloproteinases (MMPs) as well as with the function of certain exosomes. However, it has been shown that exosomes involving particular APRO proteins, MMPs, and/or microRNA could contribute to the regulation of invasiveness. Here, we discuss contradictory reports on invasiveness in relation to APRO family proteins on the basis of understanding the function of MMPs and/or various exosomes. A better understanding of those mechanisms could be of use to bring about innovative strategies for cancer treatment.

1. Introduction

The APRO family is composed of at least six distinct members in vertebrates, namely, Tob1, Tob2, BTG1, BTG2/TIS21/PC3, ANA/Tob5/BTG3, and PC3b [1]. The main characteristic of this family is the presence of a highly conserved, 110-amino-acid N-terminal region, designated as the APRO homology domain [1], which holds two highly homologous regions, designated Box-A and Box-B [1,2]. Box-A has been suggested to play an antiproliferative role [2]. All the APRO family members may be involved in the regulation of cell proliferation, and actually, have the potential to regulate tumor cell growth [1,3]. Tob1 was isolated as a protein associating with the ErbB2 receptor protein [3]. Subsequently, Tob2 was isolated on the basis of its similarity to Tob1 [4]. Other family members had been identified using several different strategies [5]. A significant association has been identified between the expression level of Tob1 and clinicopathological characteristics, including the depth of invasion and/or the lymph node metastasis stage [6]. The downregulated expression of Tob1 has been found in malignant gastric cancer, suggesting that the low expression of Tob1 may be an independent indicator of poor prognosis in gastric cancers [7]. Similarly, the downregulation of Tob1 may be associated with the shorter survival of gastric cancer patients [8]. Consistently, Tob1 overexpression could not only increase the expression of PTEN, but also regulate the downstream effectors in the PI3K/PTEN signaling pathway, leading to the suppression of cancer cell proliferation [9]. In addition, significant prognostic effects of the whole APRO family have been found in lung adenocarcinoma and ovarian, colorectal, and brain cancers, but not in squamous-cell lung carcinoma [10]. Thus, accumulating evidence has shown that APRO family proteins may function as a tumor suppressor.
However, it has been also reported that Tob1 expression may be upregulated during the progression of colon cancer, which is significantly correlated with tumor size and a prognostic indicator, such as survival rate in colon cancer [11]. In addition, Tob1 deficiency appears to lead to the reduced tumorigenesis in DSS-treated cancer, suggesting that Tob1 is an adverse prognostic factor [11]. Consistently, the downregulation of Tob1 is associated with the shorter survival of gastric cancer patients [8,12]. The suppression of Tob1 could enhance the metastasis in lung carcinoma cell line A549 cells [9,13]. To summarize the above, although Tob1 could suppress the proliferation and/or carcinogenesis in a cell, Tob1 overexpression could also promote the invasiveness and/or metastasis of several cancers. The invasiveness and metastasis of tumors are associated with matrix metalloproteinases (MMPs) [14,15]. In addition, invasiveness, metastasis, and concomitant poor prognosis may be also involved in the function of certain exosomes [16,17]. Here, we discuss contradictory reports on the invasiveness of cancer cells in relation to the actions of Tob1, a member of the APRO family, on the basis of understanding the function of MMPs and/or exosomes. We wish we could combine MMPs and exosomes in the context of APRO regulation for the invasiveness of cancer cells. A better understanding of these mechanisms might be used to design more efficient cancer therapies [18]. Such knowledge could also be of use to engineer novel strategies for cancer treatment.

2. Exosomes with APRO Proteins and/or Certain MicroRNAs May Contribute to Cancer Invasion

Exosomes are a class of extracellular membrane vesicles with a circular lipid bilayer ranging in diameter from about 30 to 150 nm [19], which are capable of mediating invasion and/or metastasis by transferring their contents, including proteins, lipids, and nucleic acids, to adjacent cells [20]. Exosomes are broadly distributed in blood cells, dendritic cells, tumor cells, and other cells [21], which can be used for diagnosis and/or prognosis within cancer patients [22]. Molecular machineries of prevailing biogenesis, cargo loading, and/or delivery of exosomes may be intricate and are still under investigation. Exosomes secreted from gastric cancer cells overexpressing Tob1 could induce autophagy of LC3-II accumulation in the gastric cancer cells [23], which may influence the proliferation, migration, and invasion of cancer cells [24]. It has been shown that exosomes containing the BTG1 protein are present in the pleural fluid obtained from patients suffering from mesothelioma, lung cancer, breast cancer, and ovarian cancer [25]. The BTG1 protein has been also identified in plasma exosomes. In addition, the plasma-exosome-derived BTG-1 levels have been related to tumor diameter, stage, tumor metastasis, the degree of tumor differentiation, and abnormal CEA levels, in accordance with previous findings of BTG-1 expression in other cancers [25]. Furthermore, a low number of plasma exosomes with low levels of BTG-1 have been observed in the poor differentiation group, suggesting that plasma-exosome-derived BTG-1 might be a potential biomarker for a prognosis in patients with non-small-cell lung cancer [25].
Noncoding RNAs in exosomes from a variety of cells have been shown to influence metastasis via various mechanisms [26]. In particular, the microRNAs (miRNAs) commonly detected in exosomes are single-strand, non-encoding RNAs with the length of about 20 nucleotides, which are usually found in eukaryotic organisms [27]. The miRNAs can affect the stability and/or translational efficacy of their target mRNAs, consequently resulting in decreased protein translation [28]. More than 1000 miRNAs have been shown to regulate a lot of biological processes including proliferation, migration, and/or differentiation in cells [29]. Interestingly, miR590 in the exosome exerts its effects through targeting Tob1 [30]. miR32 might regulate the invasion of cancer stem cells, as it is upregulated in colorectal cancer tissues compared to the adjacent normal tissues [31,32]. An exosome containing miR-135a-5p could activate MMP-7 to promote liver metastasis in colorectal cancer [33] (Figure 1). Exosomal miR-4443 may also promote the metastasis of breast cancer cells through downregulating tissue inhibitors of metalloproteinase 2 (TIMP2) and upregulating several MMPs [34]. For enhanced migration, exosomal miR-21 may influence MMP-9 and TIMP-2 through the PI3K/AKT signal pathway, but not MMP-2 and TIMP-1 [35]. TIMP3 might be targeted by macrophage-derived exosomal miR-21-5p [36]. MMP-2 expression might negatively correlate with miR-29c expression in urinary exosomes [37]. These findings are indicative of the potential of miRNAs and/or exosomes as therapeutic markers in various cancers.

3. MMPs and TIMPs Could Be Also Involved in Cancer Invasion and Metastasis

MMPs are key players in matrix remodeling. Their function has been principally investigated in cancer biology; they are involved in different steps of cancer development, from local expansion by the proliferation of cancer cells to tissue invasion and/or metastasis through extracellular matrix degradation [38]. MMPs could promote cell migration and tumor invasion through the proteolytic degradation of the extracellular basement. MMPs are synthesized as pre-proMMPs, from which the signal peptide is removed during translation to produce mature proMMPs. MMP expression can be also affected by several hormones, growth factors, and/or cytokines [39]. A higher expression of MMPs has been revealed as a potential marker of higher invasiveness and/or worse prognosis in patients with various cancers. For example, ovarian hormones could affect the expression of several MMPs, which might participate in endometrial tissue remodeling during menstrual cycles [40]. Additionally, increases in estrogen and/or progesterone, as well as vascular endothelial growth factor (VEGF), during pregnancy could promote the expression of several MMPs, which might also facilitate the tissue invasion of cytotrophoblasts [41]. Exosomes from vascular smooth muscle cells (VSMCs) may be burdened with the MMP-2 protein and specific miRNAs for controlling cell adhesion and/or migration [42]. Furthermore, macrophage-derived exosomes could trigger the expression of MMP-2 in the VSMCs via JNK and p38 pathways [43]. Interestingly, the Box-A domain in the Tob1 protein may have protease activity, which activates the MMP-7 [44]. It has been discovered that MMPs exist in exosomes from various cell types and/or some body fluids [45].
The activity of MMPs could be regulated by endogenous tissue inhibitors of matrix metalloproteinases (TIMPs). In general, MMPs are regulated at multiple levels, including in mRNA expression, the activation of the proenzyme to the active form, and the counteracting actions of these TIMPs, which are specific for each MMP type. There are four homologous members in the TIMP family with a similar structure [46], which could also regulate remodeling and turnover of the extracellular matrix (ECM) during normal and/or pathological conditions [47]. The N-terminal domain of each TIMP protein holds the inhibitory activity for the wasting potential of the MMPs [48]. The role of TIMPs in the ECM turnover could be defined as the potential inhibition of MMPs with various efficacies. Increased MMP activity and/or decreased TIMP expression could lead to MMP/TIMP imbalance, which might result in various pathological conditions including cancer invasion and/or metastasis. For example, TIMP-1 has been shown to interact with several MMPs and the matrix-degrading properties of the MMPs, which could play a fundamental role in the spread of cancer [49]. In addition to the MMP-inhibitory function, TIMP-1 could also stimulate cell growth [50], and it exhibits antiapoptotic activity [51]. It has been reported that exosome-bound TIMP1 may be a circulating biomarker for a noninvasive risk stratification in patients with colorectal liver metastases [52]. In addition, it may be extremely possible to diagnose cancers by precisely analyzing the expression of TIMP-1 in exosomes [53].

4. Activated MMPs and/or Certain MicroRNAs in Exosomes Could Contribute to the Enhanced Migration, Invasion, and/or Metastasis of Cancer Cells

In general, various MMPs are upregulated in various cancers and inflamed regions. Cancer progression could be a complex process, during which numerous cells, including malignant cells, inflammatory cells, and/or surrounding stromal cells, might communicate with each other in the microenvironment. MMPs may be involved in the remodeling of the extracellular matrix in the microenvironment to allow dissemination and/or metastasis of cancer cells [54]. Among them, MMP-2 and MMP-9 are the most distinctive MMPs characterized by a strong proteolytic activity in the extracellular matrix [55], which could be overexpressed in tumor cells and may be linked to risky metastasis and/or poor prognosis [56] (Figure 1). Exosomes could also regulate the migration of lung cancer cells into the rich vasculature by promoting MMP-2 expression [57]. In addition, exosomes could activate MMP-2 to enhance the invasiveness required for the first step in the metastasis of cancer cells [58]. Furthermore, it has been shown that exosomes derived from renal cancer cells may contribute to renal cancer development, progression, and/or invasion via the increased expression of MMP-9 [59]. Additionally, exosomes with MMP-13 could enhance migration and/or invasiveness to promote the aggressiveness of nasopharyngeal carcinoma cells [60]. High levels of MMP-1 in exosomes could potentiate the metastasis in triple-negative breast cancer [61]. Exosomes from cancer stem cells could enhance the proliferative, migratory, and/or invasive abilities of fibroblasts, accompanied by the upregulated expression of MMP-2 and MMP-9 [62].
Exosomes could promote the proliferation, migration, invasion, and angiogenesis of HUVECs, in which the increased mRNA and protein levels of VEGF and/or MMP-9 are detected [63]. Melanoma-derived exosomes may also provide an invasive capability with the higher expressions of MMP-2 and/or MMP-9, in which miR-21 is at least partially responsible for the effect [64]. Exosomes with miR-205-5p could promote angiogenesis and metastasis by enhancing the expression of MMP-2 and/or MMP-9 [65]. Exosomal miR-106b could enhance the invasive ability of lung cancer cells and increase MMP-2 and MMP-9 expression [66]. Exosomal miR-4435 could affect the migration and/or invasion of colorectal cancer cells [67]. Interestingly, exosomes with CRN2, which is an actin filament-binding protein involved in the regulation of cell migration and invasion, could promote perivascular invasion of glioblastoma cells by increasing the catalytic activity of MMP-14 [68]. Similarly, exosomal CXCR4 could promote hepatocarcinoma cell migration, invasion, and/or lymphangiogenesis by enhancing the secretions of MMP-2 and MMP-9 [69].
MMPs might be crucial for ECM remodeling under the pathological conditions of cancers. MMPs are of crucial importance for the invasiveness of cancer cells. For a good invasion performance, cancer cells must adjust the activation rate of MMPs corresponding to the solidity of the surrounding ECM. Accordingly, their expression may correlate with metastatic potential and to a significant prognostic marker.

5. Activated MMPs Could Also Regulate the Responses of Immune Cells against Cancers

Cancers must evade antitumor immune responses to continue to grow. In fact, cancer cells can often escape from immune surveillance, which has been shown to be associated with various types of immune cells including Tregs and Th17 cells [70] (Figure 1). Therefore, immune responses against cancer have been revealed as a crucial issue in the treatment of cancer. Most tumor cells express antigens that can mediate recognition by host CD8+ T cells. Interestingly, high levels of MMP-9 detected in laryngeal cancer could play a critical role in the development of Treg cells, which have an ability to suppress the tumor-specific CD8+ T cells [71]. In addition, the increased production of MMP-7 might trigger an increase in the suppressive function of Treg cells [72]. Additionally, the expression of MMP-9 might be correlated with the markers of Th1 cells and/or T-cell exhaustion [73]. Furthermore, upregulated expressions of MMP-2 and MMP-9 may promote the migration and/or invasiveness of esophageal adenocarcinoma via the action of IL-17A, which is a proinflammatory cytokine secreted from Th17 cells [74]. Likewise, the MMP inhibitor may regulate the expression of TGF-β, thus reducing the number of Tregs [75]. Amazingly, the expression of MMP-7 caused by H. pylori infection could contribute to poor responses of the adaptive immune system characterized by insufficient Th1 and/or Th17 cells and the inappropriate activation of Treg cells [76,77]. Human chorionic gonadotropin (hCG), a hormone essential for pregnancy, is also ectopically expressed by a variety of cancers and is associated with a poor prognosis, which could induce the synthesis of MMP-2 and/or MMP-9, thereby increasing invasiveness in an MMP-dependent manner. The hCG could also upmodulate the secretion of TGFβ and IL-10, thereby inhibiting T-cell proliferation [78].
Consistently, the inhibition of MMP-2/MMP-9 may improve the efficacy of PD-1 or CTLA4 blockade therapy in the treatment of aggressive metastatic cancers [79]. The PD-1 or CTLA4 checkpoint blockade are dramatic therapies for several cancers that enhance antitumor immune activity. Immune checkpoints are diligently related to tumor immune escape, which may be related to the poor prognosis of some tumors in the survival analysis [80]. The PD-1 ligand is regulated through proteolytic cleavage by endogenous MMPs from stromal fibrocytes (Figure 1). For example, increased MMP-10 expression in CD90+ fibroblasts may contribute to mucosal tolerance via the suppression of Th1 cells through the cell surface membrane-bound PD-L1, which could suppress Th1 and/or Th17 responses from activated CD4+ T cells [81]. In this case, supplementation of the MMP inhibitors could restore the suppression of Th1/Th17 cells. PD-L1 could be also cleaved by MMP-13, whereas PD-L2 is sensitive to broader MMP activities. Accordingly, MMPs might play a significant role in the immune checkpoint responses in cancer therapy. In fact, the MMP-dependent cleavage of PD-1 ligands on fibroblasts may limit their immunosuppressive capacity [82]. Interestingly, a combined treatment with the MMP inhibitor and anti-CTLA-4 antibody could delay tumor growth and reduce the metastases compared with anti-CTLA-4 treatment alone in lung and liver cancers [83]. Similarly, MMP-9 inhibition with an anti-MMP-9 monoclonal antibody could promote antitumor immunity through the disruption of biochemical barriers to the T-cell trafficking of tumors [84].

6. Discussion and Perspectives

There are contradictory reports on the invasiveness of cancer cells in the exploitation of the BTG1 protein, the other member of the APRO family, as well as those of Tob1 as shown in the introduction. A body of evidence indicates that BTG1 expression is negatively correlated with tumor invasion, lymph node metastasis, the clinical stage, and/or a low survival rate in patients with various cancers. For example, the low expression of BTG1 might be involved in the progression of pancreatic ductal adenocarcinoma, suggesting that BTG1 might be a poor prognostic marker of the survival rate in cancer [85]. BTG1 expression has been shown at lower levels in colorectal cancer than in the control, due to the hypermethylation of the BTG1 promoter [86]. In addition, the low expression of BTG1 has been reported to be associated with aggressive features and/or a worse prognosis of thyroid cancer [87], esophageal cancer [88], and squamous cell skin carcinoma [89]. The cumulative survival rate of BTG1-positive patients with colon cancer is significantly higher than that of BTG1-negative patients [90]. However, it has also been reported that BTG1 expression is positively linked to aggressive features of colorectal cancer, including depth of invasion and/or lymph node metastasis [91]. In addition, colorectal metastatic cancer cells in the lymph nodes has shown more BTG1 expression than that in the primary cancer site, suggesting that the overexpression of BTG1 might promote the invasion and/or metastasis of the colorectal cancer [91,92]. Similarly, the BTG1-overexpressing endothelial cells have exhibited increased cell migration [93]. Taken together, BTG1’s high expression might be involved in the poor progression of several cancers, and might be considered as a marker indicating that BTG1 promotes migration, invasion, and/or metastasis. Consequently, we should be cautious to employ APRO proteins for the target of cancer therapy.
How is this situation explained? Our answer is as follows: It has been shown that the ubiquitin–proteasome system could mediate the degradation of many proliferative and antiproliferative gene products. Therefore, this system might play an important role in the degradation of APRO proteins, as well as unusual key proteins [94]. In fact, the degradation of BTG2 is inhibited by lactacystin, a proteasome-specific inhibitor [95]. This ubiquitin–proteasome system could be affected by the alteration of cellular homeostasis that is cell-type dependent [96], which may be one of the reasons (reason 1). A balance between the expression of MMPs and that of TIMPs could control cancer cell migration, invasion, and/or metastasis. Both the synthesis and degradation of these proteins are important for determining how they work. As shown here, the balance of these proteins might be cell-type or cancer-type dependent, which might be the other reason (reason 2). In opposition to the invasion-facilitating exosomes, as shown in Section 2 and 3, some exosomes could slow down angiogenesis, migration, invasion, and/or metastasis [97,98,99], which might be also cell-type or cancer-type dependent (reason 3). APRO family proteins might be a key modulator of microRNAs [100]. Even though APRO members could inhibit cell proliferation, migration, and/or invasion, the other conditions such as the expression levels of MMPs or TIMPs, presence of exosomes, and/or functions of proteins/microRNAs in exosomes could further modify the effect of APRO proteins, probably for those reasons (Figure 2). However, the pathophysiological significance of APRO family proteins in cancer remains unknown. Evolving evidence suggests that PIWI-interacting RNAs (piRNAs) may be important epigenetic regulators of gene expression in human cancers [101], which may also significantly contribute to cancer pathogenesis [102]. BTG1 has been shown as a direct target of piR-1245, suggesting an inverse correlation between BTG1 expression and piR-1245 in colorectal cancer [101]. BTG1 has also been shown as a direct target of miR-330-3p, which could increase the expression of MMP-9 [103]. The rational connection to APRO proteins and MMP proteins could be elucidated through the intensive upcoming research in terms of cancer therapeutics.

7. Conclusions

Although APRO family proteins could regulate the invasiveness of cancer cells, alternative consequences might occur due to the special effects of MMPs and/or exosomes containing certain microRNAs.

Author Contributions

Conceptualization, Y.I. and S.M.; original draft preparation and editing, K.T., H.S., S.Y., A.T. and S.M.; visualization, Y.I. and S.M.; supervision, S.M. Each author (Y.I., K.T., H.S., S.Y., A.T. and S.M.) has participated sufficiently in this work of drafting the article and/or revising the article. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in this article.

Conflicts of Interest

The authors declare that they have no competing financial interests.

Abbreviations

APROsantiproliferative proteins
ECMextracellular matrix
hCGhuman chorionic gonadotropin
miRNAsmicroRNAs
MMPsmatrix metalloproteinases
PTENphosphatase and tensin homolog
TIMPtissue inhibitors of metalloproteinase
VEGFvascular endothelial growth factor
VSMCsvascular smooth muscle cells

References

  1. Matsuda, S.; Rouault, J.; Magaud, J.; Berthet, C. In search of a function for the TIS21/PC3/BTG1/TOB family. FEBS Lett. 2001, 497, 67–72. [Google Scholar] [CrossRef]
  2. Guéhenneux, F.; Duret, L.; Callanan, M.B.; Bouhas, R.; Hayette, S.; Berthet, C.; Samarut, C.; Rimokh, R.; Birot, A.M.; Wang, Q.; et al. Cloning of the mouse BTG3 gene and definition of a new gene family (the BTG family) involved in the negative control of the cell cycle. Leukemia 1997, 11, 370–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Matsuda, S.; Kawamura-Tsuzuku, J.; Ohsugi, M.; Yoshida, M.; Emi, M.; Nakamura, Y.; Onda, M.; Yoshida, Y.; Nishiyama, A.; Yamamoto, T. Tob, a novel protein that interacts with p185erbB2, is associated with anti-proliferative activity. Oncogene 1996, 12, 705–713. [Google Scholar] [PubMed]
  4. Ikematsu, N.; Yoshida, Y.; Kawamura-Tsuzuku, J.; Ohsugi, M.; Onda, M.; Hirai, M.; Fujimoto, J.; Yamamoto, T. Tob2, a novel anti-proliferative Tob/BTG1 family member, associates with a component of the CCR4 transcriptional regulatory complex capable of binding cyclin-dependent kinases. Oncogene 1999, 18, 7432–7441. [Google Scholar] [CrossRef] [Green Version]
  5. Buanne, P.; Corrente, G.; Micheli, L.; Palena, A.; Lavia, P.; Spadafora, C.; Lakshmana, M.K.; Rinaldi, A.; Banfi, S.; Quarto, M.; et al. Cloning of PC3B, a novel member of the PC3/BTG/TOB family of growth inhibitory genes, highly expressed in the olfactory epithelium. Genomics 2000, 68, 253–263. [Google Scholar] [CrossRef] [Green Version]
  6. Zhang, S.Q.; Sun, K.K.; Wu, X.Y.; Zhong, N.; Zhao, H.; Li, D.C. Clinicopathological significance of cytoplasmic transducer of ErbB2. 1 expression in gastric cancer. Mol. Med. Rep. 2015, 12, 1177–1182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Yang, Y.; Shen, X.; Sun, K. Prognostic value of transducer of ErbB2.1 (TOB1) expression in patients with gastric cancer: Tissue microarray analysis. Int. J. Clin. Exp. Pathol. 2018, 11, 4060–4066. [Google Scholar] [PubMed]
  8. Guo, H.; Zhang, R.; Afrifa, J.; Wang, Y.; Yu, J. Decreased expression levels of DAL-1 and TOB1 are associated with clinicopathological features and poor prognosis in gastric cancer. Pathol. Res. Pract. 2019, 215, 152403. [Google Scholar] [CrossRef]
  9. Jiao, Y.; Sun, K.K.; Zhao, L.; Xu, J.Y.; Wang, L.L.; Fan, S.J. Suppression of human lung cancer cell proliferation and metastasis in vitro by the transducer of ErbB-2.1 (TOB1). Acta Pharm. Sin. 2012, 33, 250–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Bai, Y.; Qiao, L.; Xie, N.; Shi, Y.; Liu, N.; Wang, J. Expression and prognosis analyses of the Tob/BTG antiproliferative (APRO) protein family in human cancers. PLoS ONE 2017, 12, e0184902. [Google Scholar] [CrossRef] [PubMed]
  11. Li, D.; Xiao, L.; Ge, Y.; Fu, Y.; Zhang, W.; Cao, H.; Chen, B.; Wang, H.; Zhan, Y.Y.; Hu, T. High expression of Tob1 indicates poor survival outcome and promotes tumour progression via a Wnt positive feedback loop in colon cancer. Mol. Cancer 2018, 17, 159. [Google Scholar] [CrossRef]
  12. Guan, R.; Peng, L.; Wang, D.; He, H.; Wang, D.; Zhang, R.; Wang, H.; Hao, H.; Zhang, J.; Song, H.; et al. Decreased TOB1 expression and increased phosphorylation of nuclear TOB1 promotes gastric cancer. Oncotarget 2017, 8, 75243–75253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Sun, K.K.; Yang, Y.; Zhao, L.; Wang, L.L.; Jiao, Y. Transducer of erbB2.1 is a potential cellular target of gefitinib in lung cancer therapy. Oncol. Lett. 2013, 5, 373–377. [Google Scholar] [CrossRef] [Green Version]
  14. Zhou, J.; Liu, T.; Wang, W. Prognostic significance of matrix metalloproteinase 9 expression in osteosarcoma: A meta-analysis of 16 studies. Medicine 2018, 97, e13051. [Google Scholar] [CrossRef] [PubMed]
  15. Dofara, S.G.; Chang, S.L.; Diorio, C. Gene Polymorphisms and Circulating Levels of MMP-2 and MMP-9: A Review of Their Role in Breast Cancer Risk. Anticancer Res. 2020, 40, 3619–3631. [Google Scholar] [CrossRef]
  16. Chu, X.; Yang, Y.; Tian, X. Crosstalk between Pancreatic Cancer Cells and Cancer-Associated Fibroblasts in the Tumor Microenvironment Mediated by Exosomal MicroRNAs. Int. J. Mol. Sci. 2022, 23, 9512. [Google Scholar] [CrossRef] [PubMed]
  17. Sunami, Y.; Häußler, J.; Zourelidis, A.; Kleeff, J. Cancer-Associated Fibroblasts and Tumor Cells in Pancreatic Cancer Microenvironment and Metastasis: Paracrine Regulators, Reciprocation and Exosomes. Cancers 2022, 14, 744. [Google Scholar] [CrossRef] [PubMed]
  18. Thuault, S.; Ghossoub, R.; David, G.; Zimmermann, P. A Journey on Extracellular Vesicles for Matrix Metalloproteinases: A Mechanistic Perspective. Front Cell Dev. Biol. 2022, 10, 886381. [Google Scholar] [CrossRef] [PubMed]
  19. Conde-Vancells, J.; Rodriguez-Suarez, E.; Embade, N.; Gil, D.; Matthiesen, R.; Valle, M.; Elortza, F.; Lu, S.C.; Mato, J.M.; Falcon-Perez, J.M. Characterization and comprehensive proteome profiling of exosomes secreted by hepatocytes. J. Proteome Res. 2008, 7, 5157–5166. [Google Scholar] [CrossRef] [Green Version]
  20. Moradi-Chaleshtori, M.; Hashemi, S.M.; Soudi, S.; Bandehpour, M.; Mohammadi-Yeganeh, S. Tumor-derived exosomal microRNAs and proteins as modulators of macrophage function. J. Cell Physiol. 2019, 234, 7970–7982. [Google Scholar] [CrossRef] [PubMed]
  21. Yu, Y.; Zhang, W.S.; Guo, Y.; Peng, H.; Zhu, M.; Miao, D.; Su, G. Engineering of exosome-triggered enzyme-powered DNA motors for highly sensitive fluorescence detection of tumor-derived exosomes. Biosens. Bioelectron. 2020, 167, 112482. [Google Scholar] [CrossRef] [PubMed]
  22. Li, M.; Xia, B.; Wang, Y.; You, M.J.; Zhang, Y. Potential Therapeutic Roles of Exosomes in Multiple Myeloma: A Systematic Review. J. Cancer 2019, 10, 6154–6160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wang, Y.; Song, Y.; Zhou, L.; Wang, M.; Wang, D.; Bai, J.; Fu, S.; Yu, J. The Overexpression of TOB1 Induces Autophagy in Gastric Cancer Cells by Secreting Exosomes. Dis. Markers 2022, 2022, 7925097. [Google Scholar] [CrossRef]
  24. Zhu, H.; Yang, Y.; Wang, L.; Xu, X.; Wang, T.; Qian, H. Leptomycin B inhibits the proliferation, migration, and invasion of cultured gastric carcinoma cells. Biosci. Biotechnol. Biochem. 2020, 84, 290–296. [Google Scholar] [CrossRef] [PubMed]
  25. Wan, L.; Chen, X.; Deng, J.; Zhang, S.; Tu, F.; Pei, H.; Hu, R.; Liu, J.; Yu, H. Plasma exosome-derived B-cell translation gene 1: A predictive marker for the prognosis in patients with non-small cell lung cancer. J. Cancer. 2021, 12, 1538–1547. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, D.; Zhang, W.; Zhang, C.; Wang, L.; Chen, H.; Xu, J. Exosomal non-coding RNAs have a significant effect on tumor metastasis. Mol. Nucleic Acids 2022, 29, 16–35. [Google Scholar] [CrossRef]
  27. Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [Green Version]
  28. Mohr, A.M.; Mott, J.L. Overview of microRNA biology. Semin. Liver Dis. 2015, 35, 3–11. [Google Scholar] [CrossRef] [Green Version]
  29. Dexheimer, P.J.; Cochella, L. MicroRNAs: From Mechanism to Organism. Front Cell Dev. Biol. 2020, 8, 409. [Google Scholar] [CrossRef]
  30. Liu, Q.; Gao, Q.; Zhang, Y.; Li, Z.; Mei, X. MicroRNA-590 promotes pathogenic Th17 cell differentiation through targeting Tob1 and is associated with multiple sclerosis. Biochem. Biophys. Res. Commun. 2017, 493, 901–908. [Google Scholar] [CrossRef]
  31. Tang, G.; Wang, J.; Dong, W.; Dai, K.; Du, J. Exosomal miRNA Expression Profiling and the Roles of Exosomal miR-4741, miR-32, miR-3149, and miR-6727 on Gastric Cancer Progression. Biomed. Res. Int. 2022, 2022, 1263812. [Google Scholar] [CrossRef] [PubMed]
  32. Wu, W.; Chen, Y.; Ye, S.; Yang, H.; Yang, J.; Quan, J. Transcription factor forkhead box K1 regulates miR-32 expression and enhances cell proliferation in colorectal cancer. Oncol. Lett. 2021, 21, 407. [Google Scholar] [CrossRef]
  33. Sun, H.; Meng, Q.; Shi, C.; Yang, H.; Li, X.; Wu, S.; Familiari, G.; Relucenti, M.; Aschner, M.; Wang, X.; et al. Hypoxia-Inducible Exosomes Facilitate Liver-Tropic Premetastatic Niche in Colorectal Cancer. Hepatology 2021, 74, 2633–2651. [Google Scholar] [CrossRef]
  34. Wang, J.; Zhang, Q.; Wang, D.; Yang, S.; Zhou, S.; Xu, H.; Zhang, H.; Zhong, S.; Feng, J. Microenvironment-induced TIMP2 loss by cancer-secreted exosomal miR-4443 promotes liver metastasis of breast cancer. J. Cell Physiol. 2020, 235, 5722–5735. [Google Scholar] [CrossRef] [PubMed]
  35. Yang, C.; Luo, L.; Bai, X.; Shen, K.; Liu, K.; Wang, J.; Hu, D. Highly-expressed micoRNA-21 in adipose derived stem cell exosomes can enhance the migration and proliferation of the HaCaT cells by increasing the MMP-9 expression through the PI3K/AKT pathway. Arch. Biochem. Biophys. 2020, 681, 108259. [Google Scholar] [CrossRef] [PubMed]
  36. Dong, J.; Zhu, W.; Wan, D. Downregulation of microRNA-21–5p from macrophages-derived exosomes represses ventricular remodeling after myocardial infarction via inhibiting tissue inhibitors of metalloproteinase 3. Int. Immunopharmacol. 2021, 96, 107611. [Google Scholar] [CrossRef] [PubMed]
  37. Solé, C.; Cortés-Hernández, J.; Felip, M.L.; Vidal, M.; Ordi-Ros, J. miR-29c in urinary exosomes as predictor of early renal fibrosis in lupus nephritis. Nephrol. Dial. Transplant. 2015, 30, 1488–1496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. De Almeida, L.G.N.; Thode, H.; Eslambolchi, Y.; Chopra, S.; Young, D.; Gill, S.; Devel, L.; Dufour, A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol. Rev. 2022, 74, 712–768. [Google Scholar] [CrossRef] [PubMed]
  39. Verma, R.P.; Hansch, C. Matrix metalloproteinases (MMPs): Chemical-biological functions and (Q)SARs. Bioorg. Med. Chem. 2007, 15, 2223–2268. [Google Scholar] [CrossRef]
  40. Grzechocińska, B.; Dąbrowski, F.; Cyganek, A.; Panek, G.; Wielgoś, M. The role of metalloproteinases in endometrial remodelling during menstrual cycle. Ginekol. Pol. 2017, 88, 337–342. [Google Scholar] [CrossRef] [PubMed]
  41. Duffy, D.M.; Stouffer, R.L. Luteinizing hormone acts directly at granulosa cells to stimulate periovulatory processes: Modulation of luteinizing hormone effects by prostaglandins. Endocrine 2003, 22, 249–256. [Google Scholar] [CrossRef]
  42. Kapustin, A.N.; Shanahan, C.M. Emerging roles for vascular smooth muscle cell exosomes in calcification and coagulation. J. Physiol. 2016, 594, 2905–2914. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, Y.; Jia, L.; Xie, Y.; Cai, Z.; Liu, Z.; Shen, J.; Lu, Y.; Wang, Y.; Su, S.; Ma, Y.; et al. Involvement of macrophage-derived exosomes in abdominal aortic aneurysms development. Atherosclerosis 2019, 289, 64–72. [Google Scholar] [CrossRef] [PubMed]
  44. Nakamura, R.; Konishi, M.; Taniguchi, M.; Hatakawa, Y.; Akizawa, T. The discovery of shorter synthetic proteolytic peptides derived from Tob1 protein. Peptides 2019, 116, 71–77. [Google Scholar] [CrossRef]
  45. Shimoda, M. Extracellular vesicle-associated MMPs: A modulator of the tissue microenvironment. Adv. Clin. Chem. 2019, 88, 35–66. [Google Scholar] [PubMed]
  46. Kaur, N.; Sunil Kumar, B.V.; Mahajan, K.; Singh, S. Expression and characterization of tissue inhibitor of metalloproteinase 4 from complex canine mammary carcinomas. J. Immunoass. Immunochem. 2016, 37, 515–526. [Google Scholar] [CrossRef]
  47. Gomis-Rüth, F.X.; Maskos, K.; Betz, M.; Bergner, A.; Huber, R.; Suzuki, K.; Yoshida, N.; Nagase, H.; Brew, K.; Bourenkov, G.P.; et al. Mechanism of inhibition of the human matrix metalloproteinase stromelysin-1 by TIMP-1. Nature 1997, 389, 77–81. [Google Scholar] [CrossRef] [PubMed]
  48. Raeeszadeh-Sarmazdeh, M.; Greene, K.A.; Sankaran, B.; Downey, G.P.; Radisky, D.C.; Radisky, E.S. Directed evolution of the metalloproteinase inhibitor TIMP-1 reveals that its N- and C-terminal domains cooperate in matrix metalloproteinase recognition. J. Biol. Chem. 2019, 294, 9476–9488. [Google Scholar] [CrossRef] [PubMed]
  49. Thaysen-Andersen, M.; Thøgersen, I.B.; Lademann, U.; Offenberg, H.; Giessing, A.M.; Enghild, J.J.; Nielsen, H.J.; Brünner, N.; Højrup, P. Investigating the biomarker potential of glycoproteins using comparative glycoprofiling-application to tissue inhibitor of metalloproteinases-1. Biochim. Biophys. Acta. 2008, 1784, 455–463. [Google Scholar] [CrossRef] [PubMed]
  50. Luparello, C.; Avanzato, G.; Carella, C.; Pucci-Minafra, I. Tissue inhibitor of metalloprotease (TIMP)-1 and proliferative behaviour of clonal breast cancer cells. Breast Cancer Res. Treat. 1999, 54, 235–244. [Google Scholar] [CrossRef]
  51. Guedez, L.; Stetler-Stevenson, W.G.; Wolff, L.; Wang, J.; Fukushima, P.; Mansoor, A.; Stetler-Stevenson, M. In vitro suppression of programmed cell death of B cells by tissue inhibitor of metalloproteinases-1. J. Clin. Investig. 1998, 102, 2002–2010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Rao, V.S.; Gu, Q.; Tzschentke, S.; Lin, K.; Ganig, N.; Thepkaysone, M.L.; Wong, F.C.; Polster, H.; Seifert, L.; Seifert, A.M.; et al. Extravesicular TIMP-1 is a non-invasive independent prognostic marker and potential therapeutic target in colorectal liver metastases. Oncogene 2022, 41, 1809–1820. [Google Scholar] [CrossRef]
  53. Lin, C.; Liang, S.; Li, Y.; Peng, Y.; Huang, Z.; Li, Z.; Yang, Y.; Luo, X. Localized plasmonic sensor for direct identifying lung and colon cancer from the blood. Biosens. Bioelectron. 2022, 211, 114372. [Google Scholar] [CrossRef]
  54. Liao, W.T.; Ye, Y.P.; Deng, Y.J.; Bian, X.W.; Ding, Y.Q. Metastatic cancer stem cells: From the concept to therapeutics. Am. J. Stem. Cells 2014, 3, 46–62. [Google Scholar] [PubMed]
  55. Huang, Q.; Lan, F.; Wang, X.; Yu, Y.; Ouyang, X.; Zheng, F.; Han, J.; Lin, Y.; Xie, Y.; Xie, F.; et al. IL-1β-induced activation of p38 promotes metastasis in gastric adenocarcinoma via upregulation of AP-1/c-fos, MMP2 and MMP9. Mol. Cancer 2014, 13, 18. [Google Scholar] [CrossRef] [PubMed]
  56. Roy, R.; Yang, J.; Moses, M.A. Matrix metalloproteinases as novel biomarkers and potential therapeutic targets in human cancer. J. Clin. Oncol. 2009, 27, 5287–5297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Wu, D.M.; Deng, S.H.; Liu, T.; Han, R.; Zhang, T.; Xu, Y. TGF-β-mediated exosomal lnc-MMP2–2 regulates migration and invasion of lung cancer cells to the vasculature by promoting MMP2 expression. Cancer Med. 2018, 7, 5118–5129. [Google Scholar] [CrossRef]
  58. McCready, J.; Sims, J.D.; Chan, D.; Jay, D.G. Secretion of extracellular hsp90alpha via exosomes increases cancer cell motility: A role for plasminogen activation. BMC Cancer 2010, 10, 294. [Google Scholar] [CrossRef] [Green Version]
  59. Chen, G.; Zhang, Y.; Wu, X. 786–0 Renal cancer cell line-derived exosomes promote 786–0 cell migration and invasion in vitro. Oncol. Lett. 2014, 7, 1576–1580. [Google Scholar] [CrossRef] [Green Version]
  60. Shan, Y.; You, B.; Shi, S.; Shi, W.; Zhang, Z.; Zhang, Q.; Gu, M.; Chen, J.; Bao, L.; Liu, D.; et al. Hypoxia-Induced Matrix Metalloproteinase-13 Expression in Exosomes from Nasopharyngeal Carcinoma Enhances Metastases. Cell Death Dis. 2018, 9, 382. [Google Scholar] [CrossRef]
  61. Zhu, Y.; Tao, Z.; Chen, Y.; Lin, S.; Zhu, M.; Ji, W.; Liu, X.; Li, T.; Hu, X. Exosomal MMP-1 transfers metastasis potential in triple-negative breast cancer through PAR1-mediated EMT. Breast Cancer Res. Treat. 2022, 193, 65–81. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, D.; Li, D.; Shen, L.; Hu, D.; Tang, B.; Guo, W.; Wang, Z.; Zhang, Z.; Wei, G.; He, D. Exosomes derived from Piwil2-induced cancer stem cells transform fibroblasts into cancer-associated fibroblasts. Oncol. Rep. 2020, 43, 1125–1132. [Google Scholar] [CrossRef] [PubMed]
  63. Xue, X.; Huang, J.; Yu, K.; Chen, X.; He, Y.; Qi, D.; Wu, Y. YB-1 transferred by gastric cancer exosomes promotes angiogenesis via enhancing the expression of angiogenic factors in vascular endothelial cells. BMC Cancer 2020, 20, 996. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, C.; Wang, Y.; Chang, X.; Ba, X.; Hu, N.; Liu, Q.; Fang, L.; Wang, Z. Melanoma-Derived Exosomes Endow Fibroblasts with an Invasive Potential via miR-21 Target Signaling Pathway. Cancer Manag. Res. 2020, 12, 12965–12974. [Google Scholar] [CrossRef] [PubMed]
  65. Yang, W.; Tan, S.; Yang, L.; Chen, X.; Yang, R.; Oyang, L.; Lin, J.; Xia, L.; Wu, N.; Han, Y.; et al. Exosomal miR-205–5p enhances angiogenesis and nasopharyngeal carcinoma metastasis by targeting desmocollin-2. Mol. Ther. Oncolytics. 2022, 24, 612–623. [Google Scholar] [CrossRef]
  66. Sun, S.; Chen, H.; Xu, C.; Zhang, Y.; Zhang, Q.; Chen, L.; Ding, Q.; Deng, Z. Exosomal miR-106b serves as a novel marker for lung cancer and promotes cancer metastasis via targeting PTEN. Life Sci. 2020, 244, 117297. [Google Scholar] [CrossRef]
  67. Hong, J.W.; Kim, J.M.; Kim, J.E.; Cho, H.; Kim, D.; Kim, W.; Oh, J.W.; Kwon, H.J. MiR-4435 is an UQCRB-related circulating miRNA in human colorectal cancer. Sci. Rep. 2020, 10, 2833. [Google Scholar] [CrossRef] [Green Version]
  68. Solga, R.; Behrens, J.; Ziemann, A.; Riou, A.; Berwanger, C.; Becker, L.; Garrett, L.; de Angelis, M.H.; Fischer, L.; Coras, R.; et al. CRN2 binds to TIMP4 and MMP14 and promotes perivascular invasion of glioblastoma cells. Eur. J. Cell Biol. 2019, 98, 151046. [Google Scholar] [CrossRef]
  69. Li, M.; Lu, Y.; Xu, Y.; Wang, J.; Zhang, C.; Du, Y.; Wang, L.; Li, L.; Wang, B.; Shen, J.; et al. Horizontal transfer of exosomal CXCR4 promotes murine hepatocarcinoma cell migration, invasion and lymphangiogenesis. Gene 2018, 676, 101–109. [Google Scholar] [CrossRef]
  70. Srivastava, M.K.; Zhu, L.; Harris-White, M.; Huang, M.; St John, M.; Lee, J.M.; Salgia, R.; Cameron, R.B.; Strieter, R.; Dubinett, S.; et al. Targeting myeloid-derived suppressor cells augments antitumor activity against lung cancer. Immunotargets Ther. 2012, 2012, 7–12. [Google Scholar]
  71. Wang, B.Q.; Zhang, C.M.; Gao, W.; Wang, X.F.; Zhang, H.L.; Yang, P.C. Cancer-derived matrix metalloproteinase-9 contributes to tumor tolerance. J. Cancer Res. Clin. Oncol. 2011, 137, 1525–1533. [Google Scholar] [CrossRef]
  72. Lee, S.; Yamamoto, S.; Srinivas, B.; Shimizu, Y.; Sada, N.; Yoshitome, K.; Ito, T.; Kumagai-Takei, N.; Nishimura, Y.; Otsuki, T. Increased production of matrix metalloproteinase-7 (MMP-7) by asbestos exposure enhances tissue migration of human regulatory T-like cells. Toxicology 2021, 452, 152717. [Google Scholar] [CrossRef]
  73. Zeng, Y.; Gao, M.; Lin, D.; Du, G.; Cai, Y. Prognostic and Immunological Roles of MMP-9 in Pan-Cancer. Biomed Res. Int. 2022, 2022, 2592962. [Google Scholar] [CrossRef]
  74. Liu, D.; Zhang, R.; Wu, J.; Pu, Y.; Yin, X.; Cheng, Y.; Wu, J.; Feng, C.; Luo, Y.; Zhang, J. Interleukin-17A promotes esophageal adenocarcinoma cell invasiveness through ROS-dependent, NF-κB-mediated MMP-2/9 activation. Oncol. Rep. 2017, 37, 1779–1785. [Google Scholar] [CrossRef] [Green Version]
  75. Krstic, J.; Santibanez, J.F. Transforming growth factor-beta and matrix metalloproteinases: Functional interactions in tumor stroma-infiltrating myeloid cells. Sci. World J. 2014, 2014, 521754. [Google Scholar] [CrossRef] [Green Version]
  76. Wilson, K.T.; Crabtree, J.E. Immunology of Helicobacter pylori: Insights into the failure of the immune response and perspectives on vaccine studies. Gastroenterology 2007, 133, 288–308. [Google Scholar] [CrossRef]
  77. Ogden, S.R.; Noto, J.M.; Allen, S.S.; Patel, D.A.; Romero-Gallo, J.; Washington, M.K.; Fingleton, B.; Israel, D.A.; Lewis, N.D.; Wilson, K.T.; et al. Matrix metalloproteinase-7 and premalignant host responses in Helicobacter pylori-infected mice. Cancer Res. 2010, 70, 30–35. [Google Scholar] [CrossRef] [Green Version]
  78. Khare, P.; Bose, A.; Singh, P.; Singh, S.; Javed, S.; Jain, S.K.; Singh, O.; Pal, R. Gonadotropin and tumorigenesis: Direct and indirect effects on inflammatory and immunosuppressive mediators and invasion. Mol. Carcinog. 2017, 56, 359–370. [Google Scholar] [CrossRef]
  79. Ye, Y.; Kuang, X.; Xie, Z.; Liang, L.; Zhang, Z.; Zhang, Y.; Ma, F.; Gao, Q.; Chang, R.; Lee, H.-H.; et al. Small-molecule MMP2/MMP9 inhibitor SB-3CT modulates tumor immune surveillance by regulating PD-L1. Genome Med. 2020, 12, 83. [Google Scholar] [CrossRef]
  80. Chen, Z.; Zhuo, S.; He, G.; Tang, J.; Hao, W.; Gao, W.Q.; Yang, K.; Xu, H. Prognosis and Immunotherapy Significances of a Cancer-Associated Fibroblasts-Related Gene Signature in Gliomas. Front Cell Dev. Biol. 2021, 9, 721897. [Google Scholar] [CrossRef]
  81. Aguirre, J.E.; Beswick, E.J.; Grim, C.; Uribe, G.; Tafoya, M.; Chacon Palma, G.; Samedi, V.; McKee, R.; Villeger, R.; Fofanov, Y.; et al. Matrix metalloproteinases cleave membrane-bound PD-L1 on CD90+ (myo-)fibroblasts in Crohn’s disease and regulate Th1/Th17 cell responses. Int. Immunol. 2020, 32, 57–68. [Google Scholar] [CrossRef]
  82. Dezutter-Dambuyant, C.; Durand, I.; Alberti, L.; Bendriss-Vermare, N.; Valladeau-Guilemond, J.; Duc, A.; Magron, A.; Morel, A.P.; Sisirak, V.; Rodriguez, C.; et al. A novel regulation of PD-1 ligands on mesenchymal stromal cells through MMP-mediated proteolytic cleavage. Oncoimmunology 2015, 5, e1091146. [Google Scholar] [CrossRef] [Green Version]
  83. Li, M.; Xing, S.; Zhang, H.; Shang, S.; Li, X.; Ren, B.; Li, G.; Chang, X.; Li, Y.; Li, W. A matrix metalloproteinase inhibitor enhances anti-cytotoxic T lymphocyte antigen-4 antibody immunotherapy in breast cancer by reprogramming the tumor microenvironment. Oncol. Rep. 2016, 35, 1329–1339. [Google Scholar] [CrossRef] [Green Version]
  84. Juric, V.; O’Sullivan, C.; Stefanutti, E.; Kovalenko, M.; Greenstein, A.; Barry-Hamilton, V.; Mikaelian, I.; Degenhardt, J.; Yue, P.; Smith, V.; et al. MMP-9 inhibition promotes anti-tumor immunity through disruption of biochemical and physical barriers to T-cell trafficking to tumors. PLoS ONE. 2018, 13, e0207255. [Google Scholar] [CrossRef] [Green Version]
  85. Huang, Y.; Zheng, J.; Tan, T.; Song, L.; Huang, S.; Zhang, Y.; Lin, L.; Liu, J.; Zheng, P.; Chen, X.; et al. BTG1 low expression in pancreatic ductal adenocarcinoma is associated with a poorer prognosis. Int. J. Biol. Markers. 2018, 33, 189–194. [Google Scholar] [CrossRef] [Green Version]
  86. Jung, Y.Y.; Sung, J.Y.; Kim, J.Y.; Kim, H.S. Down-regulation of B-Cell Translocation Gene 1 by Promoter Methylation in Colorectal Carcinoma. Anticancer Res. 2018, 38, 691–697. [Google Scholar]
  87. Lu, Y.F.; Sun, G.G.; Liu, Q.; Yang, C.R.; Cheng, Y.J. BTG1 expression in thyroid carcinoma: Diagnostic indicator and prognostic marker. Int. J. Oncol. 2014, 45, 1574–1582. [Google Scholar] [CrossRef] [Green Version]
  88. Sun, G.G.; Wang, Y.D.; Cheng, Y.J.; Hu, W.N. BTG1 underexpression is an independent prognostic marker in esophageal squamous cell carcinoma. Tumour. Biol. 2014, 35, 9707–9716. [Google Scholar] [CrossRef]
  89. Li, G.; Yue, Y.; Li, W.; Chen, Y.; Fan, H.; Yuan, J.; Chen, Z.; Liu, J. Prognostic significance of low expression of B-cell translocation gene 1 (BTG 1) in skin squamous cell carcinoma. J. Biosci. 2020, 45, 70. [Google Scholar] [CrossRef]
  90. Zhang, J.; Dong, W. Expression of B Cell Translocation Gene 1 Protein in Colon Carcinoma and its Clinical Significance. Recent Pat. Anticancer Drug Discov. 2020, 15, 78–85. [Google Scholar] [CrossRef] [PubMed]
  91. Zheng, H.C.; Li, J.; Shen, D.F.; Yang, X.F.; Zhao, S.; Wu, Y.Z.; Takano, Y.; Sun, H.Z.; Su, R.J.; Luo, J.S.; et al. BTG1 expression correlates with pathogenesis, aggressive behaviors and prognosis of gastric cancer: A potential target for gene therapy. Oncotarget 2015, 6, 19685–19705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Zhao, S.; Xue, H.; Hao, C.L.; Jiang, H.M.; Zheng, H.C. BTG1 Overexpression Might Promote Invasion and Metastasis of Colorectal Cancer via Decreasing Adhesion and Inducing Epithelial-Mesenchymal Transition. Front. Oncol. 2020, 10, 598192. [Google Scholar] [CrossRef] [PubMed]
  93. Iwai, K.; Hirata, K.; Ishida, T.; Takeuchi, S.; Hirase, T.; Rikitake, Y.; Kojima, Y.; Inoue, N.; Kawashima, S.; Yokoyama, M. An anti-proliferative gene BTG1 regulates angiogenesis in vitro. Biochem. Biophys. Res. Commun. 2004, 316, 628–635. [Google Scholar] [CrossRef] [PubMed]
  94. Pickart, C.M. Mechanisms underlying ubiquitination. Annu. Rev. Biochem. 2001, 70, 503–533. [Google Scholar] [CrossRef]
  95. Ficazzola, M.A.; Fraiman, M.; Gitlin, J.; Woo, K.; Melamed, J.; Rubin, M.A.; Walden, P.D. Antiproliferative B cell translocation gene 2 protein is down-regulated post-transcriptionally as an early event in prostate carcinogenesis. Carcinogenesis 2001, 22, 1271–1279. [Google Scholar] [CrossRef] [Green Version]
  96. Yadav, D.; Lee, J.Y.; Puranik, N.; Chauhan, P.S.; Chavda, V.; Jin, J.O.; Lee, P.C.W. Modulating the Ubiquitin-Proteasome System: A Therapeutic Strategy for Autoimmune Diseases. Cells 2022, 11, 1093. [Google Scholar] [CrossRef]
  97. Yu, S.; Zhou, Y.; Niu, L.; Qiao, Y.; Yan, Y. Mesenchymal stem cell-derived exosome mir-342–3p inhibits metastasis and chemo-resistance of breast cancer through regulating ID4. Genes Genom. 2022, 44, 539–550. [Google Scholar] [CrossRef] [PubMed]
  98. Chen, Z.; Xie, Y.; Chen, W.; Li, T.; Chen, X.; Liu, B. microRNA-6785–5p-loaded human umbilical cord mesenchymal stem cells-derived exosomes suppress angiogenesis and metastasis in gastric cancer via INHBA. Life Sci. 2021, 284, 119222. [Google Scholar] [CrossRef]
  99. Liu, X.; Zhang, G.; Yu, T.; He, J.; Liu, J.; Chai, X.; Zhao, G.; Yin, D.; Zhang, C. Exosomes deliver lncRNA DARS-AS1 siRNA to inhibit chronic unpredictable mild stress-induced TNBC metastasis. Cancer Lett. 2022, 543, 215781. [Google Scholar] [CrossRef]
  100. Ikeda, Y.; Taniguchi, K.; Nagase, N.; Tsuji, A.; Kitagishi, Y.; Matsuda, S. Reactive oxygen species may influence on the crossroads of stemness, senescence, and carcinogenesis in a cell via the roles of APRO family proteins. Explor. Med. 2021, 2, 443–454. [Google Scholar] [CrossRef]
  101. Weng, W.; Liu, N.; Toiyama, Y.; Kusunoki, M.; Nagasaka, T.; Fujiwara, T.; Wei, Q.; Qin, H.; Lin, H.; Ma, Y.; et al. Novel evidence for a PIWI-interacting RNA (piRNA) as an oncogenic mediator of disease progression, and a potential prognostic biomarker in colorectal cancer. Mol. Cancer. 2018, 17, 16. [Google Scholar] [CrossRef]
  102. Moyano, M.; Stefani, G. piRNA involvement in genome stability and human cancer. J. Hematol. Oncol. 2015, 8, 38. [Google Scholar] [CrossRef] [PubMed]
  103. Zhao, X.; Chen, G.Q.; Cao, G.M. Abnormal expression and mechanism of miR-330–3p/BTG1 axis in hepatocellular carcinoma. Eur. Rev. Med. Pharm. Sci. 2019, 23, 6888–6898. [Google Scholar]
Figure 1. Hypothetical schematic image of the relationship among the APRO proteins (APROs), exosomes, immune cells, immune check point PD-L1 on stromal fibroblasts, and the cancer cells’ invasion/metastasis. Indicated molecules are examples. Arrowhead means stimulation, whereas hammerhead represents inhibition. Note that some critical pathways such as inflammation activation and/or cancer cell growth pathway have been omitted for clarity. Abbreviations: APROs—APRO family proteins; miRNAs—microRNAs; MMPs—matrix metalloproteinases; PD-L1—programmed cell-death ligand 1.
Figure 1. Hypothetical schematic image of the relationship among the APRO proteins (APROs), exosomes, immune cells, immune check point PD-L1 on stromal fibroblasts, and the cancer cells’ invasion/metastasis. Indicated molecules are examples. Arrowhead means stimulation, whereas hammerhead represents inhibition. Note that some critical pathways such as inflammation activation and/or cancer cell growth pathway have been omitted for clarity. Abbreviations: APROs—APRO family proteins; miRNAs—microRNAs; MMPs—matrix metalloproteinases; PD-L1—programmed cell-death ligand 1.
Cancers 14 04931 g001
Figure 2. Although APRO family proteins (APROs) could individually inhibit the progression, invasion, and/or metastasis of cancer cells, the other conditions such as the expression levels of MMPs or TIMPs, the presence of some exosomes, and/or the function of microRNAs (miRNAs) or specific proteins in the exosomes could further alter the effect of APRO proteins, either of promotion or inhibition, on the invasiveness of cancer cells. Indicated molecules are examples. Arrowhead means stimulation, whereas hammerhead represents inhibition. Note that some critical pathways have been omitted for clarity. Abbreviations: APROs—APRO family proteins; miRNAs—microRNAs; MMPs—matrix metalloproteinases; TIMPs—tissue inhibitors of metalloproteinases.
Figure 2. Although APRO family proteins (APROs) could individually inhibit the progression, invasion, and/or metastasis of cancer cells, the other conditions such as the expression levels of MMPs or TIMPs, the presence of some exosomes, and/or the function of microRNAs (miRNAs) or specific proteins in the exosomes could further alter the effect of APRO proteins, either of promotion or inhibition, on the invasiveness of cancer cells. Indicated molecules are examples. Arrowhead means stimulation, whereas hammerhead represents inhibition. Note that some critical pathways have been omitted for clarity. Abbreviations: APROs—APRO family proteins; miRNAs—microRNAs; MMPs—matrix metalloproteinases; TIMPs—tissue inhibitors of metalloproteinases.
Cancers 14 04931 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ikeda, Y.; Taniguchi, K.; Sawamura, H.; Yoshikawa, S.; Tsuji, A.; Matsuda, S. Presumed Roles of APRO Family Proteins in Cancer Invasiveness. Cancers 2022, 14, 4931. https://doi.org/10.3390/cancers14194931

AMA Style

Ikeda Y, Taniguchi K, Sawamura H, Yoshikawa S, Tsuji A, Matsuda S. Presumed Roles of APRO Family Proteins in Cancer Invasiveness. Cancers. 2022; 14(19):4931. https://doi.org/10.3390/cancers14194931

Chicago/Turabian Style

Ikeda, Yuka, Kurumi Taniguchi, Haruka Sawamura, Sayuri Yoshikawa, Ai Tsuji, and Satoru Matsuda. 2022. "Presumed Roles of APRO Family Proteins in Cancer Invasiveness" Cancers 14, no. 19: 4931. https://doi.org/10.3390/cancers14194931

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop