Open Access

Effect of melanoma stem cells on melanoma metastasis (Review)

  • Authors:
    • Qiliang Yin
    • Xiumin Shi
    • Shijie Lan
    • Haofan Jin
    • Di Wu
  • View Affiliations

  • Published online on: May 29, 2021     https://doi.org/10.3892/ol.2021.12827
  • Article Number: 566
  • Copyright: © Yin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer stem cells (CSCs) are involved in the metastatic process, the resistance of many types of cancer to therapeutic treatments and consequently the onset of recurrences. The CSC concept therefore significantly extends our understanding of melanoma biology. More recently, melanoma stem cells (MSCs) have been described in melanoma as expressing specific biomarkers. These primitive melanoma cells are not only capable of self‑renewal and differentiation plasticity, but may also confer virulence via immune evasion and multidrug resistance, and potentially, via vasculogenic mimicry and transition to migratory and metastasizing derivatives. This review will present the specific biomarkers of MSCs, including CD133, ATP binding cassette subfamily B member 5, CD271, CD20 and aldehyde dehydrogenase, which can regulate the transduction of tumor‑related signals. These signal molecules can reversely act on tumor cells and regulate tumor angiogenesis, leading to the occurrence of melanoma metastasis. Targeting these specific biomarkers could inhibit the progression of melanoma and may help the development of novel therapeutic strategies for melanoma.

Introduction

Melanoma is a malignant tumor of melanocytes that typically arises in the skin. It is highly malignant, prone to metastasis and recurrence, accounting for ~75% of skin cancer-associated mortality (1). The treatment of advanced or metastatic melanoma is particularly challenging, and there is a high tendency for patients to relapse and become resistant to current therapeutic agents (2). Although molecular targeted therapy and immunotherapy have been reported to prolong the survival time of patients, most patients will develop drug resistance within one year (35), resulting in melanoma metastasis. The existence of melanoma stem cells (MSCs) is one of the potential causes of melanoma invasion and metastasis.

Cancer stem cells (CSCs) have been shown to be an integral part of solid tumors (6). Furthermore, CSCs exhibit distinctive and remarkable capacities of self-renewal, differentiation and proliferation, which are believed to have a key role in all aspects of carcinogenesis, including tumor recurrence and metastasis (7,8). Previous studies have demonstrated that the essence of tumor metastasis is the transfer and homing of CSCs (6,9). In the last decade, with the rise of CSCs, several lines of evidence suggested that CSCs may be at the origin of tumor metastasis (1012). Interestingly, the CSC subpopulation is responsible for many aspects of tumorigenesis and has been reported to serve a crucial role in melanoma development, progression, drug resistance and metastasis (13,14). The fact that the CSCs are resistant to chemotherapy also explains that traditional anticancer drugs can only inhibit or narrow the tumor, but not completely eradicate it, leading to tumor metastasis and recurrence (15,16). In addition, CSCs have been reported to express a variety of biomarkers, such as CD34, aldehyde dehydrogenase 1 (ALDH1), CD271, CD44 and lysine demethylase 5B (JARID1B); however, none of these markers have been shown to be CSCs-specific (1719). Several potential biomarkers of CSCs have been demonstrated to be expressed by certain human solid tumors such as melanoma (16), including CD133, ATP binding cassette subfamily B member 5 (ABCB5), CD271, CD20 and ALDH (Table I). Although the mechanism of MSCs promoting tumor metastasis and recurrence has not been fully elucidated (13,20,21), the activation of the signaling pathways, including Notch, Hedgehog and Wnt, is modulated by these biomarkers to maintain the characteristic of MSCs, thus promoting angiogenesis and epithelial-mesenchymal transition (EMT) of melanoma and accelerating tumor metastasis (7,20,22) (Fig. 1). It is therefore important to determine the role of MSCs in the invasion and metastasis of melanoma.

Table I.

The role of specific biomarkers in melanoma.

Table I.

The role of specific biomarkers in melanoma.

CD133CD133+ melanoma stem-like cells confer resistance to taxol (33).
CD133 and ABCG2 positive melanoma cells have the potential ability to promote tumorigenesis (27)
CD133+ population within cutaneous tissues promotes the continuous progress of melanoma (32)
CD133 is a melanoma immunogenic target of which expression is often associated with expression of cancer/testis antigens (34,35)
ABCB5Expression of ABCB5 in melanoma cells promotes resistance to chemical agent (40)
ABCB5+ tumor cells detected in human melanoma patients show a primitive molecular phenotype and correlate with clinical melanoma progression (37)
ABCB5 interacts with CD166, which promotes clinical malignant melanoma progression (39)
CD271CD271 is associated with neurotrophins and their receptors, which plays an important role in promoting melanoma cell invasion in vitro and migration (52,55)
A relatively high frequency of CD271/Sox10-positive cells correlates with higher metastatic potential and worse prognosis in human melanoma (65)
CD271+ cells show higher tumorigenicity and metastatic ability in melanoma (49)
The low affinity neurotrophin receptor CD271 plays a dual role as a mediator of phenotype switching, suppressing melanoma cell proliferation while concomitantly promoting metastasis formation in vivo (58).
CD20Melanomas contain distinct cell subpopulations including one expressing CD20 with stem cell-like and tumor-initiating characteristics (36,72).
The CD20-expressing melanoma subpopulation is characterized by self-renewal, differentiation into several cell lineages and high tumorigenicity (73).
ALDHALDH1 is a promising new marker for cancer stem cells and catalyzes the oxidation of intracellular aldehydes, thus conferring multidrug resistance (83).
ALDH1 is important for cell proliferation, survival and resistance to chemotherapeutic agents (85,86).
ALDH+ melanoma cells are more tumorigenic than ALDH- cells in both NOD/SCID mice and NOD/ SCID/IL2rγnull mice (88).
Sox10Loss of Sox10 impairs neural crest stem cell maintenance and reduces the number of CD271-positive cells and counteracts tumorigenesis in melanoma (92,93).
Following Sox10 knockdown, human melanoma cells are no longer able to initiate tumors in a xenotransplantation model (67).

[i] ABCB5, ATP binding cassette subfamily B member 5; ALDH, aldehyde dehydrogenase; Sox10, SRY-Box transcription factor 10.

Melanoma metastasis is modulated by molecular markers of MSCs

CD133

As a surface protein with unknown function, CD133 (prominin-1) can be expressed on human melanoma, but hardly be detected in normal skin (23). It has been reported that CD133, a stem cell-related surface antigen, is closely related to tumor proliferation and progression in various types of tumor, including melanoma (15,24,25). Furthermore, CD133+ melanoma cells exhibit 411 upregulated genes, which are associated with angiogenesis, adhesion and migration (26). Melanoma CD133+ CSCs have the potential to initiate tumor progression. In addition, CD133 can activate MAPK signal pathway through notch receptor 1 (Notch1), regulate the expression of vascular endothelial growth factor (VEGF) and matrix metalloproteinases and promote the interaction of tumor endothelial cells, leading to the increase of tumor angiogenesis and lung metastasis (26). Furthermore, the expression of CD133, p-p38 and p-MEK3/6 in metastatic melanoma is significantly higher than that in paracancerous tissues (26,27), suggesting that Notch1 and its MAPK signaling pathway network might be considered as potential targets for MSCs-mediated melanoma targeted therapy (28,29). Furthermore, it is generally known that tumor angiogenesis is an essential factor for tumor growth and metastasis. In particular, CD133+ and ABCB5+ MSCs are involved in the formation of perivascular niches (30). When RNAi is used to block the expression of CD133, the tumorigenicity of stem cells in vivo is significantly decreased and the expression of CD144 and ABCB5 that are closely related to the vascular microenvironment is downregulated (30). However, ABCB5 cells lose the ability to form CD144+ angiogenic mimicry. It has been reported that CD133+/ABCB5+ MSCs exist in CD144+ angiogenic mimicry, suggesting that CD133+ MSCs could promote tumor metastasis by increasing the formation of angiogenic mimicry and specific vascular microenvironment (30). Zimmerer et al (31) demonstrated by fluorescence microscopy that CD133+ melanoma D10 cells xenograft into nude mice can trigger an important angiogenesis process. A clonal dominance of a CD133+ population exists within the hierarchy of cells in cutaneous tissues from patients that have undergone successive progressive stages of melanoma, from primary to metastatic lesions (32). In addition, in the CD133 melanoma cells subpopulation, exposure to taxol induces the activation of apoptosis signal-regulating kinase1/c-jun-N-terminal kinase, p38 and ERK pathways and Bax expression; however, in CD133+ cells, taxol only enhances the activity of the ERK pathway (33). Furthermore, it was demonstrated that the expression of CD133+ in patients with recurrent and metastatic melanoma is twice higher than in patients with primary melanoma (32). Mechanistically, CD133 downregulation in human metastatic melanoma cells can decrease the capacity of sphere-forming and the metastasis potential of melanocytes (28). In addition, CD133, is closely related to the expression of certain tumor associated antigens and could thus serve as a potential target for immunotherapy (34,35). CD133 may therefore be considered as a predictive marker of melanoma and as a potential therapeutic target of high-risk melanoma.

ABCB5

ABCB5 is a member of the ATP binding cassettes (ABC) transporter family and a regulator of cell membrane potential. ABCB5 can regulate the fusion of normal skin progenitor cells and is considered as one marker of MSCs (36,37). It is now well accepted that ABC transporters mediate multidrug resistance through drug efflux in cancer cells, which is usually related to cancer stem cells (3840). In vivo genetic lineage tracking demonstrated a specific capacity of ABCB5+ sub-populations for self-renewal and differentiation, as ABCB5+ cancer cells generate both ABCB5+ and ABCB5 progeny whereas ABCB5 tumor populations give rise, at lower rates, exclusively to ABCB5 cells (37). Subsequently, ABCB5, a marker of MSCs, has high tumorigenicity potential and is co-expressed with other MSCs markers, such as CD133 (39,41,42). ABCB5+ tumor cells detected in human melanoma patients show a primitive molecular phenotype and correlate with clinical melanoma progression (43). Analysis of some clinical data demonstrated that overexpression of ABCB5 can promote tumor progression, and that ABCB5 expression is usually low in pigmented nevus subpopulation but high in primary and metastatic melanoma cell subpopulation (37,44). In serial human-to-mouse xenotransplantation experiments, ABCB5+ melanoma cells possess greater tumorigenic capacity than ABCB5 bulk populations (44,45). Ma and Frank (45) reported that ABCB5+ melanoma cells exist in the peripheral blood of patients with melanoma. Subsequently, transplanting these cells to Nod/SCID/IL2 mice can induce distant metastasis, and the degree of metastasis is directly proportional to the number of ABCB5+ melanoma cells transplanted.

Frank et al (46) demonstrated that ABCB5+ human melanoma cells promote the formation of angiogenic mimicry by expressing endothelium-specific markers and other angiogenic proteins. Because angiogenic mimicry plays an essential role in angiogenesis of melanoma (21), the ABCB5+ subpopulation of human melanoma expresses preferentially the proteins tyrosine kinase with immunoglobulin like and EGF like domains 1 and CD144 (VE-cadherin) and other angiogenic differentiation markers. Furthermore, ABCB5+ and CD133+ MSCs express preferentially VEGFR1 and VEGF, which are important for the angiogenic mimicry of human melanoma cells and further promote melanoma metastasis (26,46). It was reported that in vivo targeting of VEGFR-1 blocks the development of ABCB5+ vascular mimicry and inhibits tumor growth in melanoma (46). Furthermore, one potential mechanism of metastasis promotion and melanoma recurrence by ABCB5 is that ABCB5 might inhibit the activation of T cells and contribute to immune escape of tumor cells (47). Furthermore, the levels of MHC class I receptor, melanoma associated antigen, costimulatory molecule B7.2 and PD-1 are lower in ABCB5+ cells than in ABCB5 cells (48). These findings indicate that targeting ABCB5 MSCs markers in the treatment of melanoma might be beneficial, ongoing clinical trials have proved this view (48).

CD271

CD271, also known as low affinity nerve growth factor receptor or p75NTR, is a characteristic marker of MSCs (36,49). High Expression of CD271 has been reported in numerous human neural-crest-derived tissues and in some human cancers, including melanomas (5052). Previous studies demonstrated that CD271+ melanoma cells have a higher tumorigenicity potential than CD271 cells and are involved in the metastasis of melanoma in vivo, especially in peripheral nerves (49,5355). CD271 is the most reliable cell surface marker for the identification of melanoma heterogeneous subsets (49). In addition, not only CD271 marks dedifferentiated melanoma cells emerging, for instance, through TGFβ-mediated EMT, BRAF inhibitor-induced reprogramming (56) or in response to immunotherapies (57), but it is also functionally involved in promoting low rates of proliferation and high metastatic capacity (58). In fully immunocompromised mouse models, including NOD/SCID/IL2rγnull mice, melanoma cells expressing the neurotrophin receptor CD271 have a higher tumor-initiation capacity than CD271 cells, although the negative fraction is also able to generate tumors in this mouse model (49). Furthermore, CD271+ melanoma cells are prone to form liver and lung metastasis in mice, whereas CD271 melanoma cells rarely form metastasis (59). In human samples, the proportion of CD271/ SRY-Box transcription factor 10 (Sox10) positive cells is significantly increased in metastatic melanoma compared with primary melanoma cells (49,60). Interestingly, CD271 inactivation not only results in decreased melanoma cell survival, but also in increased sensitivity to BRAF inhibitor treatment, suggesting that CD271 might confer therapy resistance (61). Schnegg et al (62) demonstrated that CD133+ and CD271+ MSCs accumulate in the perivascular niche that melanoma cells with formation of angiogenic mimicry positively express CD271. Similarly, in the human uveal melanoma cell line c918 cultured in 2D and 3D cultures, evaluation of CD271 expression through immunofluorescence showed that CD271 is expressed on the tumor cells that form the vasculogenic mimicry (63). It was confirmed that melanoma cells that form the vasculogenic mimicry acquire the tumor stem cell-like phenotype and participate in angiogenesis (63).

A previous study reported that the number of MSCs positive for ABCB5, CD271 and receptor activator of nuclear factor κ B in circulating tumor cells (CTCs) of patients with advanced melanoma is significantly increased (64). In addition, CTCs are highly enriched in MSCs, which is a vital parameter inducing the formation of distant secondary tumors (64). Furthermore, melanoma-associated antigens, such as melanoma antigen recognized by T-cells 1, are less exposed to CD271 and ABCB5 positive melanoma cells, which supports the hypothesis that melanoma cells expressing the MSCs makers can escape the attack of host immune system (65). A previous study demonstrated that CD271 overexpression in melanoma cells inhibits the production of melanoma-specific cytotoxic T lymphocytes (CTLs), and that interferon-γ from CTLs subsequently triggers the expression of CD271 in melanoma cells, downregulating therefore the production of melanoma antigens (66). Similarly, it was reported that melanoma cells highly expressing CD271 are associated with high tumor metastasis potential and poor prognosis of patients (67).

CD20

In melanoma, numerous subpopulations with the capacity of self-renewal, differentiation, tumorigenicity and/or drug resistance have been described (49,6870), including one subpopulation expressing the B cell marker CD20 (36,7173). Further characterization with respect to melanoma-associated antigen indicated that a more primitive melanoma phenotype revealed overexpression of CD20 (16). Importantly, CD20 was initially identified on a small percentage of human melanoma cells when cultured in embryonic stem cell medium and found on nonadherent spheres. These CD20+ melanoma cells with the ability of self-renewal and differentiation followed the definition of tumor stem cells. Consistent with the view that cancer stem cells occupy a small part of tumors, CD20+ cells only account for ~2% of the total number of melanoma cells (74). However, CD20+ melanoma cells were demonstrated to be highly tumorigenic in vivo following xenotransplantation, suggesting that these cells exhibit tumor-initiating capacity (16). A previous study reported that the melanoma cells WM115 in the non-adherent form (melanoma spheroid cells) express a higher level of CD20 compared with adherent WM115 cells and that it is more likely to develop tumor when melanoma spheroid cells are transplanted into mice (16). Similarly to all stem cells, melanoma spheroid cells are also capable of proliferation, differentiation and self-renewal (16). In the late stage of metastatic melanoma, the effect of targeted therapy with anti-CD20 monoclonal antibody is more significant than that of non-targeted therapy and can even achieve a clinical complete response (7577). In melanoma patients resistant to chemotherapeutic drugs, intratumoral injections of rituximab, which is the specific antibody against CD20, induces a regression of tumor growth, accompanied by a significant decrease in serum levels of inflammatory markers (77). Although there is no clear evidence that CD20+ MSCs are directly involved in melanoma metastasis, the stem cell-like characteristics of CD20+ melanoma cells have been confirmed, and their high tumorigenicity and migration ability are the main reasons for tumor progression (16,76). CD20 may therefore be considered as a new target for melanoma treatment in the future.

ALDH

ALDH represents a group of isoenzymes that can oxidize acetaldehyde to acetic acid. The enzymatic activity of ALDH has been used to identify stem or progenitor cells from various malignancies including breast, colon and lung cancers (7880). According to previous studies, ALDH, which is a marker in many CSCs (78), is associated with multidrug resistance and immune tolerance of different types of solid tumor (78,8183), can inhibit oxidative stress and enhance resistance to chemotherapeutic drugs, such as oxazolidine, taxanes and platinum drugs (8486). It has been reported that melanoma cells with high expression of ALDH exhibit MSCs characteristics (87,88). Furthermore, ALDH-positive melanoma cells are more resistant to chemotherapeutic agents, and silencing ALDH1A using small interfering RNA can sensitize melanoma cells to drug-induced cell death (88). ALDHhigh cells (melanoma cells with high expression of ALDH) can produce more melanoma clone spheres than ALDHlow cells (melanoma cells with low expression of ALDH) (87); however, the ability of melanoma formation in vivo is significantly inhibited following ALDH silencing (88). For example, following downregulation of aryl hydrocarbon receptor (AHR) and/or ALDH1 in mouse melanoma B16F10 cells through retroviral transduction, Contador et al (89) demonstrated that ALDH1 downregulation could inhibit the metastasis of melanoma cells without AHR expression, reduce the number of CD133+/CD29+/CD44+ cells and the size of melanospheres, confirming that overactivation of ALDH1 could promote the progression of melanoma in the context of AHR deficiency. Similarly, ALDH1 silencing in melanoma cells using short hairpin (sh)RNA significantly delays the appearance and growth of xenograft melanoma and dramatically decreases the number and load of metastases in mice (90). These studies confirm that targeting ALDH1 may be considered as an effective strategy for the treatment of advanced melanoma.

Other molecular markers of MSCs

Sox10 is a key nuclear transcription factor involved in the malignant transformation of melanocytes that has the potential to be a marker of MSCs (91). The positive rate of Sox10 in sentinel lymph node micrometastasis of melanoma is close to 100%, which is significantly higher than other melanoma markers responsible for melanoma metastasis, such as S100, HMB45 and Melania (91). Furthermore, it has also been reported that the stem cell function of CD271+ melanoma cells may be correlated to the CD271/Sox10 interaction network, although the underlying mechanism remains unclear (59,92). High levels of organic cation transporter (OCT)3/4, Nanog homeobox (Nanog) and Sox10 are found in CD133+ transgenic mice and human melanoma cells, and were demonstrated to promote tumor neovascularization, indicating that Sox10 and other MSCs markers can regulate each other and accelerate tumor progression (26). However, Sox10 silencing in human melanoma cells suppresses neural crest stem cell properties, inhibits cell proliferation and survival and completely abolishes in vivo tumor formation (93). Sox10 may therefore represent a promising target for the treatment of congenital naevi and melanoma in human patients.

C-X-C motif chemokine receptor 6 (CXCR6) is also an important marker of MSCs. Compared with ATP binding cassette subfamily G member 2 (ABCG2)+ melanoma cells, CXCR6+ melanoma cells can produce larger tumors in a shorter time, and ABCG2+ and CXCR6+ double-positive cells have a higher tumorigenic potential than ABCG2+ or CXCR6+ single-positive melanoma cells in promoting tumor metastasis (94).

CD44 has been used as a specific marker of MSCs in preclinical previous studies (95,96). It has been reported that blocking insulin-like growth factor-1 can prevent the metastatic and EMT processes of melanoma cells by downregulating the stem cell markers Sox2, OCT3/4, CD44 CD133 and deleting stem cell functional characteristics (97). CD44 may therefore serve an important role in melanoma metastasis.

Melanoma stem cells and angiogenesis

The process of angiogenesis is an important hallmark of the growth and progression of tumors, including melanoma (98). Evidence has shown that tumor vessels are derived from capillaries and veins in the host tissue, following activation by pro-angiogenic factors of pre-existing endothelial cells migrating into the tumor and developing into new vessel networks (99). The endothelial progenitor cells found in the peripheral circulation also contribute to tumor angiogenesis by differentiating and proliferating in a local tumor (100). Similarly, mesenchymal stem cells promote the growth and angiogenesis of tumors thanks to their self-renewal capacity, long-term viability and differentiation potential toward diverse cell types (101104). For example, CD133+ glioma cells secrete higher levels of VEGF than CD133 cells, which indicates that tumor stem cells promote tumor angiogenesis (105). Furthermore, MSCs, which are involved in tumor angiogenesis, can accelerate melanoma metastasis. The renal and melanoma derived-CSCs are able to differentiate into endothelial like cells when cultured in endothelial cell growth specific medium (26,106). In addition, melanoma cells with stem cell-like characteristics, particularly those locating at the margin of the tumor, such as melanoma initiating cells, express and deliver in the microenvironment several factors (including VEGF, bFGF and PDGF) associated with angiogenesis (20). Since MSCs have high degree of differentiation plasticity, they can contribute to the de novo formation of tumor angiogenesis via a process named vasculogenic mimicry (VM) (107). Interestingly, tumor cells with abundant VM have a high plasticity, and their ability to mimic vascular endothelial cells may be related to the stemness of tumor cells (107,108). It has been reported that increasing the expression of stem cell-like genes in melanoma can improve the plasticity of tumor cells (108,109). Furthermore, ABCB5+ MSCs express specific endothelial and proangiogenic factors as well as VE-cadherin, Tie2, VEGF and its receptors, which are specific markers of VM (46). Consistently with these observations, melanoma cells expressing the CD133 and ABCG2 stem cells markers overexpress proangiogenic proteins, such as VEGF and its receptor VEGFR-2, Tie2 and angiopoietin (27). Furthermore, CD271+ MSCs were demonstrated to be associated with VM, through activation of the VEGR receptor/PKC signaling pathway (64,110). Thus, MSCs in angiogenesis contribute to melanoma growth and metastasis.

Targeting molecular markers of melanoma stem cells

The molecular markers of MSCs are of relative specificity and can serve as targets of molecular targeted therapy. They not only contribute to the removal of MSCs but also prevent normal cells from being damaged, in order to achieve the highest benefit of tumor therapy. For example, tubacin, which is an inhibitor of histone deacetylase 6, can promote the release of CD133+ exosomes in the human metastatic melanoma cells FEMX–I, decrease the content of CD133 in cells and inhibit the proliferation and clonogenesis of FEMX–I cells (111). In addition, monoclonal antibodies against different epitopes of CD133 have exhibited a dose-dependent cytotoxicity in FEMX–I cells (28,112). Furthermore, CD133 monoclonal antibody can inhibit the proliferation of FEMX–I melanoma cells and prevent the growth of melanoma through its cytotoxic effect. It was also demonstrated that CD133 downregulation by shRNA can decrease the appearance of lung and spinal cord metastases from melanoma (28). In addition, andrographolide can block the expression of notch1-dependent CD133 in melanoma cells and decrease the activation of MAPK signaling pathway, leading thus to inhibition of tumor growth, angiogenesis and metastasis (26). Similarly, the elimination of chemoresistant ABCB5-positive cells may significantly inhibit the overall growth of xenotransplanted melanomas by a selective antibody (37). ABCB5 is involved in the in vitro and in vivo survival of melanoma cells following exposure to dacarbazine and the BRAF inhibitor vemurafenib (113), and systemic administration of anti-ABCB5 antibody inhibits melanoma tumorigenesis in nude mice (37). Furthermore, cuprous oxide nanoparticles can significantly decrease the expression of Sox10 and CD271, which accelerates the apoptosis and inhibits the tumorigenicity of CD271 overexpressing A375 and WM266-4 cells (114). Lunasin can decrease the expression level of ALDH, a marker of MSCs, and the expression level of Nanog, a stem cell-related factor. Lunasin can also promote the upregulation of microphthalmia associated transcription factor that inhibits the colony-forming ability of tumor cells and the growth of xenograft tumors, and can increase the transformation from ALDHhigh to ALDHlow melanoma cells (115). Furthermore, magnolol can decrease the expression of CD271, CD166, JARID1B and ABCB5 through reducing the expression level of notch 2, a downstream target protein of HES-1, and of the cell cycle-related protein cyclin D1, inhibiting therefore the proliferation of melanoma cells and inducing their autophagy (116). In a clinical trial, rituximab, an anti-CD20 monoclonal antibody, was used to treat CD20+ metastatic melanoma. The anti-CD20 monoclonal antibody can eliminate CD20+ melanoma cells and increase the level of peripheral B cells in patients with melanoma (77). In an in vivo and in vitro study of melanoma cells, MSCs-induced angiogenesis mimicry was shown be a potential biological target for some anticancer compounds, such as the natural phytochemicals lupeol, which can prevent tumor metastasis by inhibiting angiogenesis (117). However, current research mainly uses cell and animal models, and large-scale clinical trials are required to support these conclusions.

Conclusion

The present review on MSCs provided some insights into the metastasis, recurrence, drug resistance and treatment of melanoma. MSCs serve a crucial role in the occurrence and progression of melanoma, especially in tumor metastasis. MSCs can promote tumor progression via MSCs-specific markers and the subsequent regulation of related signaling pathways. It is therefore crucial to fully understand the underlying mechanisms of MSCs markers in the process of tumor metastasis, which would allow the discovery of effective targets for the targeted therapy of melanoma. By exploring MSCs markers and the related signal transduction pathways, we believe that effective treatment strategies to inhibit tumor metastasis or eradicate melanoma might be discovered in the near future.

Acknowledgements

Not applicable.

Funding

This research was supported by the Project Agreement for Science & Technology Development, Jilin Province (grant no. 20200404135YY), the Youth Fund of National Natural Science Foundation of China (grant no. 81801562), the Clinical Research Foundation of First Hospital of Jilin University and the Eighth Youth Developmental Foundation of First Hospital of Jilin University (grant no. JDYY82017014).

Availability of data and materials

Not applicable.

Authors' contributions

QY drafted the initial manuscript, and edited and critically revised the manuscript. HJ and DW gave guidance on the conception and design of the review. XS and SL were involved in conceiving and designing the review. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Lo JA and Fisher DE: The melanoma revolution: From UV carcinogenesis to a new era in therapeutics. Science. 46:945–949. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Holderfield M, Deuker MM, McCormick F and McMahon M: Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond. Nat Rev Cancer. 14:455–467. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, et al: Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 364:2507–2516. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Flaherty KT, Robert C, Hersey P, Nathan P, Garbe C, Milhem M, Demidov LV, Hassel JC, Rutkowski P, Mohr P, et al: Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med. 367:107–114. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Christiansen SA, Khan S and Gibney GT: Targeted therapies in combination with immune therapies for the treatment of metastatic melanoma. Cancer J. 23:59–62. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Nandy SB and Lakshmanaswamy R: Cancer stem cells and metastasis. Prog Mol Biol Transl Sci. 151:137–176. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Parmiani G: Melanoma cancer stem cells: Markers and functions. Cancers (Basel). 8:342016. View Article : Google Scholar : PubMed/NCBI

8 

Fink J, Andersson-Rolf A and Koo BK: Adult stem cell lineage tracing and deep tissue imaging. BMB Rep. 48:655–667. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Ricci E, Mattei E, Dumontet C, Eaton CL, Hamdy F, van der Pluije G, Cecchini M, Thalmann G, Clezardin P and Colombel M: Increased expression of putative cancer stem cell markers in the bone marrow of prostate cancer patients is associated with bone metastasis progression. Prostate. 73:1738–1746. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Croker AK and Allan AL: Cancer stem cells: Implications for the progression and treatment of metastatic disease. J Cell Mol Med. 12:374–390. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Li F, Tiede B, Massague J and Kang Y: Beyond tumorigenesis: Cancer stem cells in metastasis. Cell Res. 17:3–14. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Reya T, Morrison SJ, Clarke MF and Weissman IL: Stem cells, cancer, and cancer stem cells. Nature. 414:105–111. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Wickremesekera AC, Brasch HD, Lee VM, Davis PF, Woon K, Johnson R, Tan ST and Itinteang T: Expression of cancer stem cell markers in metastatic melanoma to the brain. J Clin Neurosci. 60:112–116. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Nguyen N, Couts KL, Luo Y and Fujita M: Understanding melanoma stem cells. Melanoma Manag. 2:179–188. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Klein WM, Wu BP, Zhao S, Wu H, Klein-Szanto AJ and Tahan SR: Increased expression of stem cell markers in malignant melanoma. Mod Pathol. 20:102–107. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, Hotz S, Van Belle PA, Xu X, Elder DE and Herlyn M: A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res. 65:9328–9337. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Tirino V, Desiderio V, Paino F, De Rosa A, Papaccio F, Fazioli F, Pirozzi G and Papaccio G: Human primary bone sarcomas contain CD133+ cancer stem cells displaying high tumorigenicity in vivo. FASEB J. 25:2022–2030. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Desiderio V, Papagerakis P, Tirino V, Zheng L, Matossian M, Prince ME, Paino F, Mele L, Papaccio F, Montella R, et al: Increased fucosylation has a pivotal role in invasive and metastatic properties of head and neck cancer stem cells. Oncotarget. 6:71–84. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Collins AT, Berry PA, Hyde C, Stower MJ and Maitland NJ: Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 65:10946–10951. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Marzagalli M, Raimondi M, Fontana F, Montagnani Marelli M, Moretti RM and Limonta P: Cellular and molecular biology of cancer stem cells in melanoma: Possible therapeutic implications. Semin Cancer Biol. 59:221–235. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Kumar D, Gorain M, Kundu G and Kundu GC: Therapeutic implications of cellular and molecular biology of cancer stem cells in melanoma. Mol Cancer. 16:72017. View Article : Google Scholar : PubMed/NCBI

22 

Lee N, Barthel SR and Schatton T: Melanoma stem cells and metastasis: Mimicking hematopoietic cell trafficking? Lab Invest. 94:13–30. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Shakhova O and Sommer L: Testing the cancer stem cell hypothesis in melanoma: The clinics will tell. Cancer Lett. 338:74–81. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C and De Maria R: Identification and expansion of human colon-cancer-initiating cells. Nature. 445:111–115. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Zhang D, Tang DG and Rycaj K: Cancer stem cells: Regulation programs, immunological properties and immunotherapy. Semin Cancer Biol. 52:94–106. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Kumar D, Kumar S, Gorain M, Tomar D, Patil HS, Radharani NNV, Kumar TVS, Patil TV, Thulasiram HV and Kundu GC: Notch1-MAPK signaling axis regulates CD133(+) cancer stem cell-mediated melanoma growth and angiogenesis. J Invest Dermatol. 136:2462–2474. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Monzani E, Facchetti F, Galmozzi E, Corsini E, Benetti A, Cavazzin C, Gritti A, Piccinini A, Porro D, Santinami M, et al: Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. Eur J Cancer. 43:935–946. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Rappa G, Fodstad O and Lorico A: The stem cell-associated antigen CD133 (Prominin-1) is a molecular therapeutic target for metastatic melanoma. Stem Cells. 26:3008–3017. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Houben R, Wischhusen J, Menaa F, Synwoldt P, Schrama D, Brocker EB and Becker JC: Melanoma stem cells: Targets for successful therapy? J Dtsch Dermatol Ges. 6:541–546. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Lai CY, Schwartz BE and Hsu MY: CD133+ melanoma subpopulations contribute to perivascular niche morphogenesis and tumorigenicity through vasculogenic mimicry. Cancer Res. 72:5111–5118. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Zimmerer RM, Matthiesen P, Kreher F, Kampmann A, Spalthoff S, Jehn P, Bittermann G, Gellrich NC and Tavassol F: Putative CD133+ melanoma cancer stem cells induce initial angiogenesis in vivo. Microvasc Res. 104:46–54. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Sharma BK, Manglik V, O'Connell M, Weeraratna A, McCarron EC, Broussard JN, Divito KA, Simbulan-Rosenthal CM, Rosenthal DS and Zapas JL: Clonal dominance of CD133+ subset population as risk factor in tumor progression and disease recurrence of human cutaneous melanoma. Int J Oncol. 41:1570–1576. 2012. View Article : Google Scholar : PubMed/NCBI

33 

El-Khattouti A, Selimovic D, Haikel Y, Megahed M, Gomez CR and Hassan M: Identification and analysis of CD133(+) melanoma stem-like cells conferring resistance to taxol: An insight into the mechanisms of their resistance and response. Cancer Lett. 343:123–133. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Koshio J, Kagamu H, Nozaki K, Saida Y, Tanaka T, Shoji S, Igarashi N, Miura S, Okajima M, Watanabe S, et al: DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 3, X-linked is an immunogenic target of cancer stem cells. Cancer Immunol Immunother. 62:1619–1628. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Gedye C, Quirk J, Browning J, Svobodova S, John T, Sluka P, Dunbar PR, Corbeil D, Cebon J and Davis ID: Cancer/testis antigens can be immunological targets in clonogenic CD133+ melanoma cells. Cancer Immunol Immunother. 58:1635–1646. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Lang D, Mascarenhas JB and Shea CR: Melanocytes, melanocyte stem cells, and melanoma stem cells. Clin Dermatol. 31:166–178. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, et al: Identification of cells initiating human melanomas. Nature. 451:345–349. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Begicevic RR and Falasca M: ABC transporters in cancer stem cells: Beyond chemoresistance. Int J Mol Sci. 18:23622017. View Article : Google Scholar : PubMed/NCBI

39 

Frank NY, Margaryan A, Huang Y, Schatton T, Waaga-Gasser AM, Gasser M, Sayegh MH, Sadee W and Frank MH: ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. Cancer Res. 65:4320–4333. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, Brafford PA, Vultur A, Basu D, Gimotty P, Vogt T and Herlyn M: A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell. 141:583–594. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Wang S, Tang L, Lin J, Shen Z, Yao Y, Wang W, Tao S, Gu C, Ma J, Xie Y and Liu Y: ABCB5 promotes melanoma metastasis through enhancing NF-κB p65 protein stability. Biochem Biophys Res Commun. 492:18–26. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Xiao J, Egger ME, McMasters KM and Hao H: Differential expression of ABCB5 in BRAF inhibitor-resistant melanoma cell lines. BMC Cancer. 18:6752018. View Article : Google Scholar : PubMed/NCBI

43 

de Waard NE, Kolovou PE, McGuire SP, Cao J, Frank NY, Frank MH, Jager MJ and Ksander BR: Expression of multidrug resistance transporter ABCB5 in a murine model of human conjunctival melanoma. Ocul Oncol Pathol. 1:182–189. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Vasquez-Moctezuma I, Meraz-Rios MA, Villanueva-Lopez CG, Magana M, Martinez-Macias R, Sanchez-Gonzalez DJ, García-Sierra F and Herrera-González NE: ATP-binding cassette transporter ABCB5 gene is expressed with variability in malignant melanoma. Actas Dermosifiliogr. 101:341–348. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Ma J and Frank MH: Isolation of circulating melanoma cells. Methods Mol Biol. Sep 29–2015.(Epub ahead of print). doi: https://doi.org/10.1007/7651_2015_300. View Article : Google Scholar

46 

Frank NY, Schatton T, Kim S, Zhan Q, Wilson BJ, Ma J, Saab KR, Osherov V, Widlund HR, Gasser M, et al: VEGFR-1 expressed by malignant melanoma-initiating cells is required for tumor growth. Cancer Res. 71:1474–1485. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Schatton T, Schutte U, Frank NY, Zhan Q, Hoerning A, Robles SC, Zhou J, Hodi FS, Spagnoli GC, Murphy GF and Frank MH: Modulation of T-cell activation by malignant melanoma initiating cells. Cancer Res. 70:697–708. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Eggermont AM and Robert C: New drugs in melanoma: It's a whole new world. Eur J Cancer. 47:2150–2157. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Boiko AD, Razorenova OV, van de Rijn M, Swetter SM, Johnson DL, Ly DP, Butler PD, Yang GP, Joshua B, Kaplan MJ, et al: Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271. Nature. 466:133–137. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Chesa PG, Rettig WJ, Thomson TM, Old LJ and Melamed MR: Immunohistochemical analysis of nerve growth factor receptor expression in normal and malignant human tissues. J Histochem Cytochem. 36:383–389. 1988. View Article : Google Scholar : PubMed/NCBI

51 

Pietra G, Manzini C, Vitale M, Balsamo M, Ognio E, Boitano M, Queirolo P, Moretta L and Mingari MC: Natural killer cells kill human melanoma cells with characteristics of cancer stem cells. Int Immunol. 21:793–801. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Truzzi F, Marconi A, Lotti R, Dallaglio K, French LE, Hempstead BL and Pincelli C: Neurotrophins and their receptors stimulate melanoma cell proliferation and migration. J Invest Dermatol. 128:2031–2040. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Nielsen PS, Riber-Hansen R and Steiniche T: Immunohis tochemical CD271 expression correlates with melanoma progress in a case-control study. Pathology. 50:402–410. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Guo R, Fierro-Fine A, Goddard L, Russell M, Chen J, Liu CZ, Fung KM and Hassell LA: Increased expression of melanoma stem cell marker CD271 in metastatic melanoma to the brain. Int J Clin Exp Pathol. 7:8947–8951. 2014.PubMed/NCBI

55 

Denkins Y, Reiland J, Roy M, Sinnappah-Kang ND, Galjour J, Murry BP, Blust J, Aucoin R and Marchetti D: Brain metastases in melanoma: Roles of neurotrophins. Neuro Oncol. 6:154–165. 2004. View Article : Google Scholar : PubMed/NCBI

56 

Shaffer SM, Dunagin MC, Torborg SR, Torre EA, Emert B, Krepler C, Beqiri M, Sproesser K, Brafford PA, Xiao M, et al: Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature. 546:431–435. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Holzel M and Tuting T: Inflammation-induced plasticity in melanoma therapy and metastasis. Trends Immunol. 37:364–374. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Restivo G, Diener J, Cheng PF, Kiowski G, Bonalli M, Biedermann T, Reichmann E, Levesque MP, Dummer R and Sommer L: low neurotrophin receptor CD271 regulates phenotype switching in melanoma. Nat Commun. 8:19882017. View Article : Google Scholar : PubMed/NCBI

59 

Redmer T, Welte Y, Behrens D, Fichtner I, Przybilla D, Wruck W, Yaspo ML, Lehrach H, Schäfer R and Regenbrecht CR: The nerve growth factor receptor CD271 is crucial to maintain tumorigenicity and stem-like properties of melanoma cells. PLoS One. 9:e925962014. View Article : Google Scholar : PubMed/NCBI

60 

Prasmickaite L, Skrbo N, Hoifodt HK, Suo Z, Engebraten O, Gullestad HP, Aamdal S, Fodstad Ø and Maelandsmo GM: Human malignant melanoma harbours a large fraction of highly clonogenic cells that do not express markers associated with cancer stem cells. Pigment Cell Melanoma Res. 23:449–451. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Lehraiki A, Cerezo M, Rouaud F, Abbe P, Allegra M, Kluza J, Marchetti P, Imbert V, Cheli Y, Bertolotto C, et al: Increased CD271 expression by the NF-κB pathway promotes melanoma cell survival and drives acquired resistance to BRAF inhibitor vemurafenib. Cell Discov. 1:150302015. View Article : Google Scholar : PubMed/NCBI

62 

Schnegg CI, Yang MH, Ghosh SK and Hsu MY: Induction of vasculogenic mimicry overrides VEGF-A silencing and enriches stem-like cancer cells in melanoma. Cancer Res. 75:1682–1690. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Valyi-Nagy K, Kormos B, Ali M, Shukla D and Valyi-Nagy T: Stem cell marker CD271 is expressed by vasculogenic mimicry-forming uveal melanoma cells in three-dimensional cultures. Mol Vis. 18:588–592. 2012.PubMed/NCBI

64 

Gray ES, Reid AL, Bowyer S, Calapre L, Siew K, Pearce R, Cowell L, Frank MH, Millward M and Ziman M: Circulating melanoma cell subpopulations: Their heterogeneity and differential responses to treatment. J Invest Dermatol. 135:2040–2048. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Civenni G, Walter A, Kobert N, Mihic-Probst D, Zipser M, Belloni B, Seifert B, Moch H, Dummer R, van den Broek M and Sommer L: Human CD271-positive melanoma stem cells associated with metastasis establish tumor heterogeneity and long-term growth. Cancer Res. 71:3098–3109. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Furuta J, Inozume T, Harada K and Shimada S: CD271 on melanoma cell is an IFN-γ-inducible immunosuppressive factor that mediates downregulation of melanoma antigens. J Invest Dermatol. 134:1369–1377. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Roesch A: Melanoma stem cells. J Dtsch Dermatol Ges. 13:118–124. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Clarke MF and Fuller M: Stem cells and cancer: Two faces of eve. Cell. 124:1111–1115. 2006. View Article : Google Scholar : PubMed/NCBI

69 

Held MA, Curley DP, Dankort D, McMahon M, Muthusamy V and Bosenberg MW: Characterization of melanoma cells capable of propagating tumors from a single cell. Cancer Res. 70:388–397. 2010. View Article : Google Scholar : PubMed/NCBI

70 

Frank NY, Schatton T and Frank MH: The therapeutic promise of the cancer stem cell concept. J Clin Invest. 120:41–50. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Boonyaratanakornkit JB, Yue L, Strachan LR, Scalapino KJ, LeBoit PE, Lu Y, Leong SP, Smith JE and Ghadially R: Selection of tumorigenic melanoma cells using ALDH. J Invest Dermatol. 130:2799–2808. 2010. View Article : Google Scholar : PubMed/NCBI

72 

Pinc A, Somasundaram R, Wagner C, Hormann M, Karanikas G, Jalili A, Bauer W, Brunner P, Grabmeier-Pfistershammer K, Gschaider M, et al: Targeting CD20 in melanoma patients at high risk of disease recurrence. Mol Ther. 20:1056–1062. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Akbulut H, Babahan C, Abgarmi SA, Ocal M and Besler M: Recent advances in cancer stem cell targeted therapy. Crit Rev Oncog. 24:1–20. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Yaiza JM, Gloria RA, Maria Belen GO, Elena LR, Gema J, Juan Antonio M, María Ángel GC and Houria B: Melanoma cancer stem-like cells: Optimization method for culture, enrichment and maintenance. Tissue Cell. 60:48–59. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Schmidt P, Kopecky C, Hombach A, Zigrino P, Mauch C and Abken H: Eradication of melanomas by targeted elimination of a minor subset of tumor cells. Proc Natl Acad Sci USA. 108:2474–2479. 2011. View Article : Google Scholar : PubMed/NCBI

76 

Murphy GF, Wilson BJ, Girouard SD, Frank NY and Frank MH: Stem cells and targeted approaches to melanoma cure. Mol Aspects Med. 39:33–49. 2014. View Article : Google Scholar : PubMed/NCBI

77 

Schlaak M, Schmidt P, Bangard C, Kurschat P, Mauch C and Abken H: Regression of metastatic melanoma in a patient by antibody targeting of cancer stem cells. Oncotarget. 3:22–30. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, Jacquemier J, Viens P, Kleer CG, Liu S, et al: ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 1:555–567. 2007. View Article : Google Scholar : PubMed/NCBI

79 

Huang EH, Hynes MJ, Zhang T, Ginestier C, Dontu G, Appelman H, Fields JZ, Wicha MS and Boman BM: Aldehyde dehydrogenase 1 is a marker for normal and malignant human colonic stem cells (SC) and tracks SC overpopulation during colon tumorigenesis. Cancer Res. 69:3382–3389. 2009. View Article : Google Scholar : PubMed/NCBI

80 

Jiang F, Qiu Q, Khanna A, Todd NW, Deepak J, Xing L, Wang H, Liu Z, Su Y, Stass SA and Katz RL: Aldehyde dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer. Mol Cancer Res. 7:330–338. 2009. View Article : Google Scholar : PubMed/NCBI

81 

Carpentino JE, Hynes MJ, Appelman HD, Zheng T, Steindler DA, Scott EW and Huang EH: Aldehyde dehydrogenase-expressing colon stem cells contribute to tumorigenesis in the transition from colitis to cancer. Cancer Res. 69:8208–8215. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Roudi R, Korourian A, Shariftabrizi A and Madjd Z: Differential expression of cancer stem cell markers ALDH1 and CD133 in various lung cancer subtypes. Cancer Invest. 33:294–302. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Yoshida A, Hsu LC and Dave V: Retinal oxidation activity and biological role of human cytosolic aldehyde dehydrogenase. Enzyme. 46:239–244. 1992. View Article : Google Scholar : PubMed/NCBI

84 

Singh S, Brocker C, Koppaka V, Chen Y, Jackson BC, Matsumoto A, Thompson DC and Vasiliou V: Aldehyde dehydrogenases in cellular responses to oxidative/electrophilic stress. Free Radic Biol Med. 56:89–101. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Le Moguen K, Lincet H, Deslandes E, Hubert-Roux M, Lange C, Poulain L, Gauduchon P and Baudin B: Comparative proteomic analysis of cisplatin sensitive IGROV1 ovarian carcinoma cell line and its resistant counterpart IGROV1-R10. Proteomics. 6:5183–5192. 2006. View Article : Google Scholar : PubMed/NCBI

86 

Moreb JS, Gabr A, Vartikar GR, Gowda S, Zucali JR and Mohuczy D: Retinoic acid down-regulates aldehyde dehydrogenase and increases cytotoxicity of 4-hydroperoxycyclophosphamide and acetaldehyde. J Pharmacol Exp Ther. 312:339–345. 2005. View Article : Google Scholar : PubMed/NCBI

87 

Santini R, Vinci MC, Pandolfi S, Penachioni JY, Montagnani V, Olivito B, Gattai R, Pimpinelli N, Gerlini G, Borgognoni L and Stecca B: Hedgehog-GLI signaling drives self-renewal and tumorigenicity of human melanoma-initiating cells. Stem Cells. 30:1808–1818. 2012. View Article : Google Scholar : PubMed/NCBI

88 

Luo Y, Dallaglio K, Chen Y, Robinson WA, Robinson SE, McCarter MD, Wang J, Gonzalez R, Thompson DC, Norris DA, et al: ALDH1A isozymes are markers of human melanoma stem cells and potential therapeutic targets. Stem Cells. 30:2100–2113. 2012. View Article : Google Scholar : PubMed/NCBI

89 

Contador-Troca M, Alvarez-Barrientos A, Merino JM, Morales-Hernandez A, Rodriguez MI, Rey-Barroso J, Barrasa E, Cerezo-Guisado MI, Catalina-Fernández I, Sáenz-Santamaría J, et al: Dioxin receptor regulates aldehyde dehydrogenase to block melanoma tumorigenesis and metastasis. Mol Cancer. 14:1482015. View Article : Google Scholar : PubMed/NCBI

90 

Yue L, Huang ZM, Fong S, Leong S, Jakowatz JG, Charruyer-Reinwald A, Wei M and Ghadially R: Targeting ALDH1 to decrease tumorigenicity, growth and metastasis of human melanoma. Melanoma Res. 25:138–148. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Willis BC, Johnson G, Wang J and Cohen C: SOX10: A useful marker for identifying metastatic melanoma in sentinel lymph nodes. Appl Immunohistochem Mol Morphol. 23:109–112. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Paratore C, Goerich DE, Suter U, Wegner M and Sommer L: Survival and glial fate acquisition of neural crest cells are regulated by an interplay between the transcription factor Sox10 and extrinsic combinatorial signaling. Development. 128:3949–3961. 2001. View Article : Google Scholar : PubMed/NCBI

93 

Shakhova O, Zingg D, Schaefer SM, Hari L, Civenni G, Blunschi J, Claudinot S, Okoniewski M, Beermann F, Mihic-Probst D, et al: Sox10 promotes the formation and maintenance of giant congenital naevi and melanoma. Nat Cell Biol. 14:882–890. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Taghizadeh R, Noh M, Huh YH, Ciusani E, Sigalotti L, Maio M, Arosio B, Nicotra MR, Natali P, Sherley JL and La Porta CA: CXCR6, a newly defined biomarker of tissue-specific stem cell asymmetric self-renewal, identifies more aggressive human melanoma cancer stem cells. PLoS One. 5:e151832010. View Article : Google Scholar : PubMed/NCBI

95 

Zhao F, Zhang R, Wang J, Wu D, Pan M, Li M, Guo M and Dou J: Effective tumor immunity to melanoma mediated by B16F10 cancer stem cell vaccine. Int Immunopharmacol. 52:238–244. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Dou J, He X, Liu Y, Wang Y, Zhao F, Wang X, Chen D, Shi F and Wang J: Effect of downregulation of ZEB1 on vimentin expression, tumour migration and tumourigenicity of melanoma B16F10 cells and CSCs. Cell Biol Int. 38:452–461. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Le Coz V, Zhu C, Devocelle A, Vazquez A, Boucheix C, Azzi S, Gallerne C, Eid P, Lecourt S and Giron-Michel J: IGF-1 contributes to the expansion of melanoma-initiating cells through an epithelial-mesenchymal transition process. Oncotarget. 7:82511–82527. 2016. View Article : Google Scholar : PubMed/NCBI

98 

RS K: Tumor angiogenesis. N Engl J Med. 358:2039–2049. 2008. View Article : Google Scholar : PubMed/NCBI

99 

Carmeliet P and Jain RK: Angiogenesis in cancer and other diseases. Nature. 407:249–257. 2000. View Article : Google Scholar : PubMed/NCBI

100 

Rafii S, Lyden D, Benezra R, Hattori K and Heissig B: Vascular and haematopoietic stem cells: novel targets for anti-angiogenesis therapy? Nat Rev Cancer. 2:826–835. 2002. View Article : Google Scholar : PubMed/NCBI

101 

Huang WH, Chang MC, Tsai KS, Hung MC, Chen HL and Hung SC: Mesenchymal stem cells promote growth and angiogenesis of tumors in mice. Oncogene. 32:4343–4354. 2013. View Article : Google Scholar : PubMed/NCBI

102 

Jeon ES, Lee IH, Heo SC, Shin SH, Choi YJ, Park JH, Park DY and Kim JH: Mesenchymal stem cells stimulate angiogenesis in a murine xenograft model of A549 human adenocarcinoma through an LPA1 receptor-dependent mechanism. Biochim Biophys Acta. 1801:1205–1213. 2010. View Article : Google Scholar : PubMed/NCBI

103 

Otsu K, Das S, Houser SD, Quadri SK, Bhattacharya S and Bhattacharya J: Concentration-dependent inhibition of angiogenesis by mesenchymal stem cells. Blood. 113:4197–4205. 2009. View Article : Google Scholar : PubMed/NCBI

104 

Sun B, Zhang S, Ni C, Zhang D, Liu Y, Zhang W, Zhao X, Zhao C and Shi M: Correlation between melanoma angiogenesis and the mesenchymal stem cells and endothelial progenitor cells derived from bone marrow. Stem Cells Dev. 14:292–298. 2005. View Article : Google Scholar : PubMed/NCBI

105 

Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, Shi Q, McLendon RE, Bigner DD and Rich JN: Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 66:7843–7848. 2006. View Article : Google Scholar : PubMed/NCBI

106 

Bussolati B, Bruno S, Grange C, Ferrando U and Camussi G: Identification of a tumor-initiating stem cell population in human renal carcinomas. FASEB J. 22:3696–3705. 2008. View Article : Google Scholar : PubMed/NCBI

107 

Hendrix MJ, Seftor EA, Hess AR and Seftor RE: Vasculogenic mimicry and tumour-cell plasticity: Lessons from melanoma. Nat Rev Cancer. 3:411–421. 2003. View Article : Google Scholar : PubMed/NCBI

108 

Seftor RE, Hess AR, Seftor EA, Kirschmann DA, Hardy KM, Margaryan NV and Hendrix MJ: Tumor cell vasculogenic mimicry: From controversy to therapeutic promise. Am J Pathol. 181:1115–1125. 2012. View Article : Google Scholar : PubMed/NCBI

109 

Girouard SD and Murphy GF: Melanoma stem cells: Not rare, but well done. Lab Invest. 91:647–664. 2011. View Article : Google Scholar : PubMed/NCBI

110 

Vartanian A, Stepanova E, Grigorieva I, Solomko E, Baryshnikov A and Lichinitser M: VEGFR1 and PKCα signaling control melanoma vasculogenic mimicry in a VEGFR2 kinase-independent manner. Melanoma Res. 21:91–98. 2011. View Article : Google Scholar : PubMed/NCBI

111 

Chao OS, Chang TC, Di Bella MA, Alessandro R, Anzanello F, Rappa G, Goodman OB and Lorico A: The HDAC6 inhibitor tubacin induces release of CD133(+) extracellular vesicles from cancer cells. J Cell Biochem. 118:4414–4424. 2017. View Article : Google Scholar : PubMed/NCBI

112 

Alamodi AA, Eshaq AM, Hassan SY, Al Hmada Y, El Jamal SM, Fothan AM, Arain OM, Hassan SL, Haikel Y, Megahed M and Hassan M: Cancer stem cell as therapeutic target for melanoma treatment. Histol Histopathol. 31:1291–1301. 2016.PubMed/NCBI

113 

Luo Y, Ellis LZ, Dallaglio K, Takeda M, Robinson WA, Robinson SE, Liu W, Lewis KD, McCarter MD, Gonzalez R, et al: Side population cells from human melanoma tumors reveal diverse mechanisms for chemoresistance. J Invest Dermatol. 132:2440–2450. 2012. View Article : Google Scholar : PubMed/NCBI

114 

Yu B, Wang Y, Yu X, Zhang H, Zhu J, Wang C, Chen F, Liu C, Wang J and Zhu H: Cuprous oxide nanoparticle-inhibited melanoma progress by targeting melanoma stem cells. Int J Nanomedicine. 12:2553–2567. 2017. View Article : Google Scholar : PubMed/NCBI

115 

Shidal C, Al-Rayyan N, Yaddanapudi K and Davis KR: Lunasin is a novel therapeutic agent for targeting melanoma cancer stem cells. Oncotarget. 7:84128–84141. 2016. View Article : Google Scholar : PubMed/NCBI

116 

Kaushik G, Venugopal A, Ramamoorthy P, Standing D, Subramaniam D, Umar S, Jensen RA, Anant S and Mammen JM: Honokiol inhibits melanoma stem cells by targeting notch signaling. Mol Carcinog. 54:1710–1721. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Bhattacharyya S, Mitra D, Ray S, Biswas N, Banerjee S, Majumder B, Mustafi SM and Murmu N: Reversing effect of Lupeol on vasculogenic mimicry in murine melanoma progression. Microvasc Res. 121:52–62. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2021
Volume 22 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yin Q, Shi X, Lan S, Jin H and Wu D: Effect of melanoma stem cells on melanoma metastasis (Review). Oncol Lett 22: 566, 2021
APA
Yin, Q., Shi, X., Lan, S., Jin, H., & Wu, D. (2021). Effect of melanoma stem cells on melanoma metastasis (Review). Oncology Letters, 22, 566. https://doi.org/10.3892/ol.2021.12827
MLA
Yin, Q., Shi, X., Lan, S., Jin, H., Wu, D."Effect of melanoma stem cells on melanoma metastasis (Review)". Oncology Letters 22.1 (2021): 566.
Chicago
Yin, Q., Shi, X., Lan, S., Jin, H., Wu, D."Effect of melanoma stem cells on melanoma metastasis (Review)". Oncology Letters 22, no. 1 (2021): 566. https://doi.org/10.3892/ol.2021.12827