Review
Wnt5a: A promising therapeutic target in ovarian cancer

https://doi.org/10.1016/j.prp.2021.153348Get rights and content

Abstract

Despite the rapid development of novel adjuvant and neoadjuvant chemotherapeutic drugs in recent years, advanced ovarian cancer still lacks effective treatment strategies and remains one of the main causes of death among women. Wnt protein family is widely expressed in a variety of human tissues, and Wnt5a is an important member of the noncanonical Wnt pathway. Wnt5a has been found to induce tumor suppression as well as to function as an oncogene depending upon the specific cancer type. In ovarian cancer, Wnt5a protein expression has been observed to be significantly upregulated and associated with poor prognosis. Researchers found that downregulating Wnt5a expression levels effectively suppresses ovarian cancer cell migration and invasion abilities. We believe that the downregulation of Wnt5a will be an attractive and promising antimetastatic therapeutic approach for the future treatment of patients with advanced ovarian cancer. The present review focuses on the mechanisms and therapeutic potential of Wnt5a as a promising novel therapeutic target for ovarian cancer.

Introduction

The Wnt protein family was originally identified in mouse breast cancer and is widely expressed in a variety of human tissues [1]. It is a large family of secreted glycoproteins and regulates cell proliferation, differentiation, migration, polarity and death during embryonic development. There are 19 Wnt family members found in mammals, including Wnt1, Wnt2, Wnt3, Wnt4, and Wnt5a, among others [2].

There are 3 different Wnt pathways [3]: (1) One pathway is the canonical Wnt pathway, also known as the Wnt/β-catenin pathway. In this pathway, the binding of Wnt protein to the Frizzled receptor inactivates the multiprotein destruction complex, causing the aggregation of β-catenin in the cytoplasm. Then, β-catenin enters the nucleus and combines with T cell factor/lymphoid enhancing factor (TCF/LEF), which results in activation of the transcription of downstream target genes (MMP7, c-myc, cyclin D1, etc.) [4]. Abnormal activation of this pathway is closely related to the development of many tumors, such as colon cancer, lung cancer, melanoma, breast cancer, and ovarian cancer. [[5], [6], [7], [8], [9], [10]]. (2) The second pathway is the Wnt/Ca2+ pathway. It is activated by Wnt4 and Wnt5a, among others. After binding to the Frizzled-2 receptor, Wnt5a increases the intracellular Ca2+ concentration, thereby activating calmodulin-dependent kinase II (CaMKII) and protein kinase C (PKC). Then, PKC and CaMKII inactivate the transcription factor nuclear factor of activated T cells (NFAT) to regulate cell adhesion, migration, and differentiation. Mice with Frizzled-2 deletion or knockout of Wnt5a show symptoms of facial deformity, dwarfism, a short tail, and respiratory problems [11]. Obviously, the Wnt5a/PKCa pathway plays a critical role in certain normal physical processes. (3) The third pathways is the Wnt/PCP pathway. Wnt5a binds to the Frizzled receptor and then activates the intracellular signaling protein dishevelled (Dvl). Dvl activates dishevelled-associated activator of morphogenesis 1 (DAAM1), leading to a small increase in the GTPase RhoA, which activates Rho-associated kinase (ROCK) and results in cytoskeletal reorganization and changes to cell polarity and migration. The second and third pathways are called noncanonical pathways [12].

There are multiple signal transduction methods and pathways in cells and interactions between the various pathways, eventually forming a complex Wnt protein interaction network [[13], [14], [15]].

Wnt5a is an important member of the noncanonical Wnt pathway. It is located at chromosome 3p14-p21 and has a molecular weight of 38 kDa [16]. It can both activate the canonical Wnt/β-catenin pathway and the noncanonical Wnt/Ca2+ pathway. The interaction between these two pathways may cause Wnt5a to play different roles in different tumors. In adult T cell leukemia, Wnt5a contributes to the increase in osteolytic bone lesions and hypercalcemia [17]. Scientists have not yet reached a consensus on the role of Wnt5a in bladder cancer [18,19] and breast cancer [20,21]. In cervical cancer, Wnt5a plays a role in promoting cancer and can strengthen the proliferation and invasion of tumor cells [22]. In chronic lymphocytic leukemia, researchers have found that Wnt5a can increase tumor cell proliferation and migration [23]. In classic Hodgkin lymphoma, Wnt5a enhances tumor metastasis [24]. In colon cancer [25] and esophageal cancer [26], Wnt5a is a tumor suppressor gene, yet in gastric cancer [27], it promotes invasion and metastasis. Wnt5a is also an oncogene in several other cancers, including glioblastoma [28], kidney renal clear cell carcinoma [29], liver cancer [30], melanoma [31], nasopharyngeal carcinoma [32], non-small-cell lung cancer [33], pancreatic cancer [34], and osteosarcoma [35]. However, Wnt5a inhibits proliferation, migration and invasion in thyroid cancer [36]. Moreover, researchers have made more interesting discoveries about Wnt5a in prostate cancer. Mikels and Nusse have found out that signaling initiated by Wnt5 is regulated by receptor availability instead of Wnt5a itself, which may produce totally different biological roles even in the same type of cancer [37]. For example, with Frizzled-2 as receptor, overexpression of Wnt5a can enhance invasion activity of prostate cancer cells [38]. On the contrary, Ren et al. have reported that Wnt5a acts as a tumor suppressor, which induces and maintains dormancy of prostate cancer cells in the bone and inhibits bone metastasis of prostate cancer by binding to the receptor-tyrosine-kinase-like orphan receptors 2 (ROR2) receptor [39]. We summarized the role of Wnt5a in different tumors in Table 1.

Researchers have found that Wnt5a protein expression in ovarian cancer is significantly higher than that in benign ovarian tumors and normal ovarian tissues [40]. The differences in prognoses and survival times in patients with different levels of Wnt5a expression suggest that Wnt5a is involved in the progression of ovarian cancer. In this review, we determined the expression level of Wnt5a in ovarian cancer and the effect of Wnt5a on the migration and invasion of ovarian cancer cells. The role of Wnt5a in the metabolism and aging of ovarian cancer cells and its potential therapeutic value are also discussed.

Section snippets

Wnt5a expression in ovarian cancer

Ovarian cancer is the most common type of reproductive tract. Over 85 % percent of ovarian cancers originate from epithelial tissue. Ovarian cancer is still one of the most lethal gynecologic malignancies in the United States [41]. Ovarian cancer can be classified into four different types: clear cell, endometrioid, mucinous and serous [42]. A study of a large cohort study of 623 patients found that Wnt5a was highly expressed in ovarian cancer and that Wnt5a knockdown significantly decreased

Wnt5a signaling regulates ovarian cancer cell migration and invasion

The metastasis and spread of ovarian cancer cells is a continuous series of events, including increased tumor cell migration, invasion of the basement membrane, entry into the circulatory system and eventually arrival at distant organs that the tumor cells can invade. Studies have shown that Wnt5a signaling pathways are involved in the regulation of ovarian cancer cell invasion, migration and metastasis [48]. In this section, we will discuss how the Wnt5a signaling pathways affect the response

Wnt5a and cellular senescence

Senescence is the arrest of cell growth and can be caused by DNA damage, cancer drugs, activation of some oncogenes such as HRas, and severe shortening of telomeres [60,61]. Strategies inducing senescence represent a new potential avenue for cancer therapy. Some studies have proven that it is possible to induce cancer cell senescence by reactivating p53 and other oncogenes [62]. Wnt5a has been proven to play an important role in inducing ovarian cancer senescence [63]. In this study, the

Wnt5a and chemotherapy resistance in ovarian cancer

Wnt5a is more than only a potential tumor treatment target; there is also evidence that Wnt5a plays an important role in ovarian cancer chemotherapy resistance, which is worthy of further study. A study showed that Wnt5a-overexpressing ovarian cancer cell lines had higher levels of resistance to paclitaxel, 5-fluorouracil, epirubicin and etoposide than control cell lines [45]. Wnt5a was also found to be overexpressed in an oxaliplatin-resistant human ovarian carcinoma cell line (A2780/C10B) [67

Therapeutic targeting of Wnt5a signaling

Since Wnt5a plays an important role in human ovarian cancer migration, invasion, metabolism, and senescence, Wnt5a is considered a potential target for novel targeted therapy. A group of researchers found that Wnt5a is overexpressed in ovarian cancer patient ascites and potentiates ovarian cancer cell prometastatic behavior. Mice with whole-body silencing of Wnt5a showed a 3-11-fold reduction in overall peritoneal tumor burden relative to controls. Furthermore, the researchers found that the

Wnt5a signaling regulates cancer cell metabolism: a promising direction in ovarian cancer

Cancer cells can escape apoptosis through an abnormal sugar metabolism mechanism called the Warburg effect [74]. Under this effect, cancer cells can survive with highly efficient aerobic glycolysis and create a hypoxic microenvironment to inhibit the monitoring and cytotoxic effects of T lymphocytes. Therefore, the mechanism enhances the proliferation and migration ability of cancer cells [74]. A recent study showed that the noncanonical Wnt pathways could control the metabolic reprogramming of

Conclusion and perspectives

Ovarian cancer is one of the most common cancers in women, and over 70 % of patients are diagnosed with metastasis [41]. Therefore, we need new biomarkers for early diagnosis and potential therapeutic targets for new treatments. The Wnt (Wingless) gene was first discovered in mouse breast cancer cells [2]. It regulates the proliferation, differentiation, migration, polarity and death of cells during embryonic development and is related to the occurrence and development of multiple human

Funding

This research was supported by the Six talent peaks project in Jiangsu Province (WSN186).

CRediT authorship contribution statement

Weijian Zhou: Conceptualization, Writing - original draft, Visualization. Jie Mei: Resources, Writing - review & editing, Visualization. Dingyi Gu: Writing - review & editing. Junying Xu: Writing - review & editing. Runjie Wang: Writing - review & editing. Huiyu Wang: Funding acquisition. Chaoying Liu: Project administration.

Declaration of Competing Interest

The authors report no declarations of interest.

References (78)

  • C.E. Ford et al.

    The non-canonical Wnt ligand, Wnt5a, is upregulated and associated with epithelial to mesenchymal transition in epithelial ovarian cancer

    Gynecol. Oncol.

    (2014)
  • J. Mei et al.

    Overexpressed DAAM1 correlates with metastasis and predicts poor prognosis in breast cancer

    Pathol. Res. Pract.

    (2020)
  • J. Mei et al.

    DAAM1-mediated migration and invasion of ovarian cancer cells are suppressed by miR-208a-5p

    Pathol. Res. Pract.

    (2019)
  • A.W. Tai et al.

    p53 restoration leads to tumor senescence and regression: implications for cancer therapy

    Gastroenterology

    (2007)
  • S. Sengupta et al.

    Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress

    Mol. Cell

    (2010)
  • W. Zuidervaart et al.

    Expression of Wnt5a and its downstream effector β-catenin in uveal melanoma

    Melanoma Res.

    (2007)
  • G. Akiri et al.

    Wnt pathway aberrations including autocrine Wnt activation occur at high frequency in human non-small-cell lung carcinoma

    Oncogene

    (2009)
  • V. Sherwood

    WNT signaling: an emerging mediator of cancer cell metabolism?

    Mol. Cell. Biol.

    (2015)
  • L. Qi et al.

    Dickkopf-1 inhibits epithelial-mesenchymal transition of colon cancer cells and contributes to colon cancer suppression

    Cancer Sci.

    (2012)
  • J.R. Prosperi et al.

    A Wnt-ow of opportunity: targeting the Wnt/beta-catenin pathway in breast cancer

    Curr. Drug Targets

    (2010)
  • Y.C. Hseu et al.

    The antitumor activity of Antrodia camphorata in melanoma cells: modulation of wnt/β-Catenin signaling pathways

    Evid. Complement. Alternat. Med.

    (2012)
  • S. Monga

    Role of Wnt/β-catenin signaling in liver metabolism and cancer

    Int. J. Biochem. Cell Biol.

    (2009)
  • A.V. Bazhin et al.

    cGMP-phosphodiesterase 6, transducin and Wnt5a/Frizzled-2-signaling control cGMP and Ca(2+) homeostasis in melanoma cells

    Cell. Mol. Life Sci.

    (2010)
  • M.V. Semenov et al.

    SnapShot: noncanonical wnt signaling pathways

    Cell

    (2007)
  • A. Sato et al.

    Wnt5a regulates distinct signalling pathways by binding to Frizzled2

    EMBO J.

    (2010)
  • M. Saling et al.

    Wnt5a / planar cell polarity signaling pathway in urothelial carcinoma, a potential prognostic biomarker

    Oncotarget

    (2017)
  • X. Chen et al.

    MicroRNA-374a inhibits aggressive tumor biological behavior in bladder carcinoma by suppressing wnt/β-Catenin signaling

    Cell. Physiol. Biochem.

    (2018)
  • K. Leandersson et al.

    Wnt-5a mRNA translation is suppressed by the Elav-like protein HuR in human breast epithelial cells

    Nucleic Acids Res.

    (2006)
  • C.P. Prasad et al.

    WNT5A as a therapeutic target in breast cancer

    Cancer Metastasis Rev.

    (2018)
  • K. Shojima et al.

    Wnt5a promotes cancer cell invasion and proliferation by receptor-mediated endocytosis-dependent and -independent mechanisms, respectively

    Sci. Rep.

    (2015)
  • J. Yu et al.

    Wnt5a induces ROR1/ROR2 heterooligomerization to enhance leukemia chemotaxis and proliferation

    J. Clin. Invest.

    (2016)
  • F. Linke et al.

    WNT5A: a motility-promoting factor in Hodgkin lymphoma

    Oncogene

    (2017)
  • R. Cheng et al.

    Wnt5a suppresses colon cancer by inhibiting cell proliferation and epithelial-mesenchymal transition

    J. Cell. Physiol.

    (2014)
  • O. Lyros et al.

    Dysregulation of WNT5A/ROR2 signaling characterizes the progression of barrett-associated esophageal adenocarcinoma

    Mol. Cancer Res.

    (2016)
  • H. Yamamoto et al.

    Laminin gamma2 mediates Wnt5a-induced invasion of gastric cancer cells

    Gastroenterology

    (2009)
  • E. Binda et al.

    Wnt5a drives an invasive phenotype in human glioblastoma stem-like cells

    Cancer Res.

    (2017)
  • Z. Hu et al.

    lncRNA MSC-AS1 activates Wnt/β-catenin signaling pathway to modulate cell proliferation and migration in kidney renal clear cell carcinoma via miR-3924/WNT5A

    J. Cell. Biochem.

    (2020)
  • Z. Chen et al.

    LncFZD6 initiates Wnt/β-catenin and liver TIC self-renewal through BRG1-mediated FZD6 transcriptional activation

    Oncogene

    (2018)
  • P.D. Da Forno et al.

    WNT5A expression increases during melanoma progression and correlates with outcome

    Clin. Cancer Res.

    (2008)
  • Cited by (5)

    • Recent advancements in therapeutic targeting of the Warburg effect in refractory ovarian cancer: A promise towards disease remission

      2021, Biochimica et Biophysica Acta - Reviews on Cancer
      Citation Excerpt :

      In line with these findings, majority of solid tumors including EOC display overexpression of gluts, aberrant regulation and function of glycolytic enzymes, elevated rate of aerobic glycolysis and diminished oxidative phosphorylation (OXPHOS)-all of which not only favour metabolic plasticity but also confer chemoresistance to the cancer cells [40,99]. Despite emergence of several novel drug targets including PARP, Nrf2, Wnt5a and dendritic cells [50,100–102], rEOC remains far from a manageable pathology and this “glucose addiction” and high propensity to consume and metabolize glucose into lactate by EOC cells have been exploited to develop novel therapeutic interventions against this lethal pathology [69]. As discussed earlier, several oncogenes and signalling cascades such as RAS, MYC, PI3K, STAT3/SIRT3/HIF1-α and FAK/ERK1/2/MMP9 favour aerobic glycolysis, confer drug resistance (Fig. 3) and small molecule inhibition of altered glycolysis by candidates including BAY876, glupin, PFK-158 and lonidamine showed promise in reversal of chemoresistance and disease management in rEOC (Fig. 4).

    1

    Equal contribution.

    View full text