Abstract
The concept of cancer stem cells (CSC) has been established over the past decade or so, and their role in carcinogenic processes has been confirmed. In this review, we focus on cervical CSCs, including (1) their purported origin, (2) markers used for cervical CSC identification, (3) alterations to signalling pathways in cervical cancer and (4) the cancer stem cell niche. Although cervical CSCs have not yet been definitively identified and characterized, future studies pursuing them as therapeutic targets may provide novel insights for treatment of cervical cancer.
Introduction
Cervical carcinomas (Fig 1) are among the most common malignant tumours of women worldwide and their mortality and morbidity levels are second only to breast cancer. Gynaecological oncologists study cancer stem cells (CSC) to explore new avenues for diagnosis and therapy of cervical carcinoma.
Figure 1.
Image of cervical cancer lesion.
Theory of cancer stem cells
CSC theory postulates that not all tumour cells are equal with regard to self‐renewal, tumour initiation and maintenance potential 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12. It proposes that a minority of tumour cells, with indefinite proliferation potential, unlimited capacity for self‐renewal, asymmetric division, and ability to differentiate into several cell lineages 13, are stem cells that play decisive roles in oncogenesis, while the majority of tumour cells die after transient differentiation. Based on this theory, heterogeneity between multiple tumour cells provides the capacity for indefinite proliferation, continuous renewal and pluripotency. In addition, cell heterogeneity and hierarchy within tumours originates from CSCs, which give rise to daughter cells that proliferate and differentiate into the cell mass that makes up a significant portion of the bulk of a tumour 14. Furthermore, CSCs are thought to be responsible for therapy resistance, residual disease and relapse after initial successful therapy. Resistance of cancer stem cells to conventional chemotherapy and radiotherapy has been attributed to cellular mechanisms such as multidrug resistance, quiescence, enhanced DNA repair ability and anti‐apoptotic mechanisms 14, 15.
Direct evidence has been derived from numerous studies. Al‐Hajj 1 and Clarke et al. 16 first succeeded in isolating cancer stem cells from human breast cancers. Results of investigations on brain tumours, including astrocytoma and medulloblastoma, on liver cancer, retinoblastoma and prostate cancer, further provided compelling evidence for cancer stem cells 17, 18, 19, 20. CSCs have also been identified in multiple myeloma 7, amongst cancer cell lines 21, 22 and in solid tumours of the breast, ovary, endometrium, prostate, brain, and lung 1, 2, 3, 4, 10, 11, using experimental strategy that combines sorting tumour cell subpopulations, identified on the basis of the different expressions of surface markers, with functional transplantation into appropriate animal models.
Source of cervical cancer stem cells
There are two theories regarding the source of cancer stem cells, i.e. mutation of stem cells in normal tissue and de‐differentiation of mature cells. Recent studies have indicated that stem cells exist in the centre of tumours, a region where the oncogenic effects of carcinogens are enhanced. In addition, an increasing number of researchers support the theory that stem cells, because of the similarities between cancer stem cells and stem cells, are normal tissue cells that have mutated. This theory has directed attention to cervical reserve cells, which are widely considered to be progenitors of cervical epithelial cells.
Cervical reserve cells
The cervix is covered by a layer of epithelial cells. These may be either squamous or columnar (also called glandular cells). Squamous cells are flat and scaly, while columnar cells appear, as indicated by their name, column‐like.
Cervical carcinogenesis associated with metaplastic squamous epithelium occurs in a specific anatomical location, the transformation zone (TZ); topography and natural history of this region is complex and dynamic. In adult women, the transformation zone usually is on the vaginal surface of the cervix, an irregular line of demarcation dividing one type of epithelium from another. Microscopically, columnar epithelium lines both the endocervical canal and associated endocervical glands, whereas squamous epithelium covers the outer cervix. During cervical squamous metaplasia, foci of squamous cells are detectable among the endocervical glandular columnar epithelium. Reserve cells are pivotal in the transformation of columnar epithelium into squamous epithelium and are most likely also targeted when endocervical epithelium transforms into cervical intraepithelial neoplasia (CIN). The junction between the two migrates proximally as the endocervical columnar epithelium is replaced by metaplastic squamous epithelium as a result of increased oestrogen production and growth of vaginal bacterial flora 23. Concentration of subglandular reserve cells is highest close to the squamo‐columnar junction, in the upper third of the cervix. This indicates that a subpopulation of distal reserve cells provides progenitors for squamous and columnar epithelium and that proximal reserve cells can serve as progenitor cells for columnar epithelium 24, 25.
Recently, cervical cancer has been considered to originate from stem cells of the TZ by interplay between HR‐HPV (high risk‐human papilloma virus) viral oncogenes combined with cellular alterations 26, 27, 28. Stem cells from the TZ in the cervical epithelium are thought to be targets for malignant transformation because of their self‐renewal and proliferativity. The TZ of the cervical epithelium is a niche for cells with a unique expression profile and embryonic characteristics 9, 29 (Fig. 2).
Figure 2.
Transformation zone as the niche of cervical cancer stem cells.
Cervical reserve cells and human papilloma virus (HPV)
In 1977, HPV particles were first detected in a cervical cancer biopsy by electron microscopy. Subsequently, a large number of studies indicated that HPV was the major pathogen in cervical cancer. HPV infections in different countries arise in accord with different circumstances 30. Current infections can be measured with the highest sensitivity by HPV DNA testing, which can also be combined with Pap (Papanicolaou) smears for optimizing detection of high‐grade cervical intraepithelial neoplasia 31, 32. Pap screening has reduced the occurrence of HPV infection and cervical cancer 33, but barriers remain in some places 34, especially in some developing countries 35, 36, and Raychaudhuri et al. attempted to determine whether there were significant differences in context of knowledge, attitude and practice (KAP) and screening, between high income and low income countries 37.
Worldwide, the most common high‐risk HR‐HPVs are subtypes 16 and 18, approximately 70% cervical cancers (CC) being due to infection by these genotypes. However, persistent infection by HR‐HPV is a necessary but insufficient cause of the neoplasia 38. Most infections are transient, and the virus can spontaneously disappear ‐ continuous infection is required to produce CIN or cervical cancer. Progression of CIN‐I → CIN‐II → CIN‐III (CIS) → invasive cancer reflects consecutive processes of cervical cancer development (Fig. 3), however, the question remains regarding how HPV infects cervical epithelium and promotes development of the malignancy. It has been suggested 32 that only after HPV has infected undifferentiated cervical stem cells can the cancer start. Initial infection of cervical epithelial cells with HPV does not lead diretly to malignant pathology. After a second infection, carcinogenic E6/E7 gene can be integrated into cervical epithelial cell DNA, disturbing modulation of proliferation and differentiation, and finally resulting in transformation. HPV oncoprotein E6 binds and degrades wild‐type p53 protein product. Transfection with HPV E6 and E7 oncogenes in YD8(HPV‐negative, p53‐mutated oropharyngeal cell lines) reduces abundance of proteins encoded by tumour suppressor genes, such as p53 and pRb (Fig 4).
Figure 3.
Illustration of the two shocks of cervical cancer stem cells.
Figure 4.
HPV E6 and E7 oncoproteins and p53, p21 and Rb tumour suppressor proteins.
Thus, HPV may infect cervical stem cells, cervical reserve cells, as only reserve cells are undifferentiated and exist long enough to be infected a second time. Martens et al. measured expression of CK17 and p63 in CIN and cervical cancer and concluded that cervical reserve cells were, indeed, the target cells of HPV 39. Furthermore, one study by Regauer et al. indicated that HR‐HPV infection of cervical reserve cells resulted in occurrence of CIN‐II and increased p63 expression. There is considerable evidence that HPV‐infected target cells are cervical reserve cells, thus the marker profile of HPV infected cells is crucially important for treatment and prevention 40.
Markers of cervical cancer stem cells
Currently, isolation of cervical cancer stem cells (CCSCs) from tumours requires knowledge of their specific surface markers, but current knowledge regarding the markers is incomplete.
Embryonic stem cell transcription factor
Nanog is a unique homeobox transcription factor that is expressed mainly in the inner cell mass (ICM) of blastocysts 41. It is highly expressed in early embryonic stem (ES) cells, and its expression reduces with their differentiation 42. Nanog plays a critical role in regulating pluripotent ICM cells in embryonic development, preventing ES cells from differentiation and maintaining their undifferentiated state 43. Recent studies have shown that Nanog is highly related to tumourigenesis. For example, it has been detected in certain tumours in which existence of cancer stem cells has already been acknowledged, such as gastric carcinoma 44, breast cancer, glioblastoma 45, lung cancer 46 and various human sarcomas 47. In one study, it was observed that Nanog is strongly expressed in CIN‐II–III and cervical carcinoma tissues, in contrast to its low expression in CIN‐I cells and normal cervical epithelia 48. Furthermore, cells of cervical epithelial tissues that overexpress Nanog are more capable of forming tumours 49.
Cytokeratin (CK)
Cytokeratins (CK) are cytoskeletal intermediate filament proteins; there are 20 subtypes expressed in various types of human epithelial cells. CK isotype depends on cell type and its localization. It has been reported CaM kinase‐like‐1 (DCAMKL‐1), Lgr5, CD133, α‐fetoprotein, cytokeratin‐19 (CK19), Lin28 and c‐Myc are up‐regulated in hepatocellular carcinoma CSCs, DCAMKL‐1 possibly being a specific marker of CSCs 50.
Differences between CKs expressed in normal cervical epithelial cells and reserve cells in the uterine cervix have been analysed. CK8 and CK17 are important subtypes expressed in CIN and cervical cancer tissues, and have been investigated as valuable markers of cervical cancer stem cells. CK8 is expressed in reserve cells, cervical gland epithelial cells and immature squamous metaplastic cells in the cervix, but not in squamous cells in a part of or mature squamous metaplastic cells. CK17 is expressed in reserve cells and immature metaplastic cells but not in cervical gland epithelial cells, squamous cells in the portio or mature squamous metaplastic cells 51. In normal cervical epithelial cells, CK17 is only expressed at borders, while in cervical cancer cells, CK17 is overexpressed. In addition, cervical cancer cells with higher expression of CK17 have greater tendency to metastasise and are thus, more highly malignant 24, 52, 53; CK8 and CK17 have great significance in diagnosis of CIN and uterine cervical cancer. However, recent studies have attached importance to CK19 in distinguishing cervical cancer stem cells. A large part of CK19 mRNA is detectable by RT‐PCR in breast, prostate and gastric cancers and some studies have shown that it is highly expressed in sentinel lymph nodes in cervical cancer but has low expression in benign lesion groups 54. Moreover, expression of CK19 is related to lymph node metastasis 55; however, understanding the exact function of CK19 in cervical cancer stem cells requires further research.
Cell adhesion molecules (CAMs)
CD44 is a transmembrane glycoprotein that participates in many cellular processes, including growth, survival, differentiation and motility, and exists as a standard isoform (CD44s) and a range of variant isoforms (CD44v), generated by alternative splicing 56, 57. CD44 has been reported in many types of cancer and has been shown to play a role in cancer cell migration and matrix adhesion in response to cell microenvironment, thus enhancing aggregation and tumour cell growth 58. Recent studies have demonstrated that CD44 can be used to isolate cancer stem cells, in neoplasms of breast, prostate, pancreas and colorectal CSCs 1, 39, 59, 60, 61.
Cells isolated from uterine cervical cancer tissues have fibroblast‐like morphology and grow in colonies. Immunophenotyping by flow cytometry has revealed that isolated cells are positive for CD13, CD29, CD44, CD105 and HLA‐I, while not expressing CD10, CD14, CD31, CD34, CD38 and HLA‐DR 62. Sphere‐forming cells (SFCs) from cervical cancer cell lines have been used to successfully isolate or enrich CSCs from cancers 63, 64, 65. HeLa‐SFCs are resistant to multiple chemotherapeutic drugs and are more tumourigenic, as evidenced by growth of tumours in immunodeficient mice following injection with 1 × 104 cells, compared to 1 × 106 parental HeLa cells, required to grow tumours of similar size in the same time frame. These cells have been found to exhibit an expression pattern of CD44 (high)/CD24 (low) that resembles CSC surface biomarker pattern of breast cancer. It has also been demonstrated that HeLa‐SFCs express higher levels (6.9‐fold) of human papillomavirus oncogene E6 compared to parental HeLa cells 66.
Octamer‐binding transcription factor 4 (Oct4)
Oct4, the POU domain transcription factor, is important for maintenance of self‐renewal, regulation of pluripotency and differentiation into specific lineages in the early mammalian embryo 67. Normally, low amounts of Oct4 protein are expressed in somatic, but not germ line, cells; however, expression of Oct4 is up‐regulated in several tumours and tumour cell lines 68, 69, which contributes to malignant transformation of normal stem cells and proliferation and therapeutic resistance of cancer cells. Down‐regulation of Oct4 leads to cancer cell apoptosis 70. Thus, Oct4 is also considered as a hallmark of cancer stem‐like cells (CSC). Recent studies indicate that it is highly expressed in several particular types of cancer tissues in which existence of cancer stem cells has been affirmed, such as lung cancer 71, hepatocellular carcinoma 72 and prostate cancer 70. It has been shown that Oct4 is strongly expressed in primary tumour cells and cancer cells in cervical cancer tissues but has relatively low level of expression in normal cervical epithelial cells 73. One recent study confirmed that cervical cancers contain a subpopulation of stem‐like cancer cells expressing Oct4 protein 74. Previous studies have also confirmed that levels of both Oct4 mRNA and protein in HPV16‐positive cervical cancer cells (Caski cells) were higher than in HPV‐negative cervical cancer cells (C‐33A cells) 75. These results suggest that HPV16 infection might be associated with up‐regulation of Oct4 expression in the process of cervical carcinogenesis.
Aldehyde dehydrogenase 1 (ALDH1)
The aldehyde dehydrogenase (ALDH) family consists of cytosolic isoenzymes responsible for oxidizing intracellular aldehydes contributing to oxidation of retinol to retinoic acid in early stem cell differentiation. ALDH1 gene is located on chromosome 9q21 and contains 13 exons encoding a 501 amino acid polypeptide. The human ALDH1 family includes ALDH1A1, ALDH1A2, ALDH1A3, ALDH1B1 and ALDH1L1; gene sequences of ALDH1A2, ALDH1A3, ALDH1B1 and ALDH1L1 are currently unknown. Human ALDH‐1 plays a role in biological synthesis of retinoic acid and is involved in early differentiation of stem cells by mediating oxidation of retinol into retinoic acid. One previous study indicated that retinoic acid is involved in cell differentiation and proliferation via binding to the retinoic acid receptor. Increased ALDH1 activation has been found in myelomatosis and acute myelocytic leukaemia; a clinical study has also indicated that some breast cancer cells also express ALDH1. Prognosis in patients with positive ALDH1 was poor, and ALDH1 expression was closely correlated to tumour grade, state of oestrogen receptor (ER)/progesterone receptor (PR), overexpression of ERBB2 and formation of CK 76, 77. Our group found that higher ALDH1 expression correlates with significantly higher rates of cell proliferation, microsphere formation and migration. We also demonstrated that SiHa‐ALDH1‐positive cells are significantly more tumourigenic compared to SiHa‐ALDH1‐negative cells. Similarly, SiHa cells that overexpress ALDH1 are significantly more tumourigenic and exhibit higher rates of cell proliferation and migration compared to SiHa cells, when ALDH1 expression was knocked down using a lentivirus vector. Our data suggested ALDH1 to be a marker of cervical cancer stem cells, which expands our understanding of its functional role 78, 79.
Nucleostemin and Musashi1
Nucleostemin (NS) is a putative GTPase that binds to p53 and is highly expressed in nucleoli of ES cells and cancer cell lines. Musashi1 (Msil1) is an RNA‐binding protein and is highly expressed in neural progenitor cells. These two proteins also play important roles in carcinogenesis of embryonic malignancies. A study by Ye et al. also demonstrates that NS and Msil1 are abundantly expressed in CIN‐II–III and cervical carcinoma tissues 48.
p16
p16 is an important tumour suppressor gene first identified in 1993. It is located on chromosome 9 its full length being 8.5 kb. Encoded products of p16, p16INK4A and p14ARF, play significant roles in oncogenesis. p16INK4A is a cyclin‐dependent kinase inhibitor that negatively regulates cell proliferation by inhibiting hyperphosphorylation of the retinoblastoma tumour suppressor (Rb) by cyclin D–CDK4/CDK6 complexes 80. p14ARF is a small basic arginine‐rich nuclear/nucleolar protein that can arrest cell growth in G1–S and G2–M in a p53‐dependent manner; its expression is increased after oncogenic stress or cell cycle deregulation 81. As a tumour suppressor gene, p16 silenced by any mechanism causes promotion of carcinogenesis.
Some studies have indicated that p16 is closely related to cervical carcinoma 82, 83, 84 and further study has demonstrated that p16INK4A and p14ARF are overexpressed in high‐grade CIN and invasive cancer, relative to low‐grade CIN or normal epithelium 85. Moreover, previous investigations have shown that methylation of p16 is a frequent event in cervical carcinoma samples, and early promoter methylation of the p16 gene may be a crucial event in malignant progression in the cervix 86. It has been reported p16INK4A is required for DDP resistance in cervical carcinoma SiHa cells 87. Thus, p16 is suggested to be a potential utility in diagnosis and prognosis 88, 89, 90.
p63
p63, a homologue of tumour suppressor gene p53, has been described to be a transcription factor operating mainly in the embryonic stage of development 91. Existence of p63 has recently been demonstrated in bronchial, prostate and cervical reserve tissues using immunohistochemical techniques, and is believed to play a role in regulation and maturation of epithelium in the adult phase 92, 93, 94, 95. Several studies have shown (by immunostaining) that p63 is strongly expressed in the basal layer of ectocervical epithelia and in basal layers of CIN lesions, irrespective of their grade 39, 53. Nevertheless, whether p63 can be a biomarker of cervical stem cells remains questionable. Some researchers have suggested that p63 can be detected in all cancer cells in the uterine cervix and are not unique to cervical cancer stem cells 73.
However, there are shortcomings and difficulties in using several sorting markers addressed to isolate CCSCs. For some inconsistencies present in published data, the difficulties can be ascribed to i) differential glycosylation, ii) epigenetic modifications and iii) tumour microenvironment.
The CSC pathway
SHH signalling pathway
Hh (Hedgehog) is one of the core developmental signalling pathways in which mutation during development causes congenital malformations 96. During embryology, this pathway controls cell proliferation and differentiation and can be activated by binding sonic hedgehog (SHH), Indian hedgehog (IHH) desert Hedgehog, and patched hedgehog (PTCH). In the absence of the Hh ligand, PTCH functions as an inhibitor of smoothened (SMO), a transmembrane protein with homology to G‐coupled protein. Activation of SMO may initiate processes during which transcription factors belonging to the Gli family, modify transcription of Ptc, WNT and Noggin, an inhibitor of BMP‐4 (bone morphogenetic protein) belonging to the TGF‐β superfamily 97, 98. In mammals, SHH regulates proliferation of primitive haematopoietic cells with BMP‐4 98. Errors in the pathway can be a pathogenic factor for development of medulloblastoma and basal cell carcinoma 17. SHH signalling has also been implicated in regulation of self‐renewal by the finding that populations highly enriched for human HSCs (haematopoietic stem cell) exhibit increased self‐renewal in response to SHH stimulation in vitro, albeit in combination with other growth factors 98 (Fig. 5).
Figure 5.
SHH pathway (cited from Sheridan C (2009) Genentech obtains proof of concept for hedgehog inhibition. Nat. Biotechnol. 27 , 968–969).
There are two different scenarios in which this pathway may be involved in carcinogenesis. One is by mutation of pathway components, as exemplified by mutation of PTCH1 and SMO; the other is autocrine requirement for an Hh ligand. It has previously been reported that components of Hh signalling pathway are gradually up‐regulated as normal epithelium progresses to squamous cell carcinoma 99, suggesting a role for Hh pathway molecules in this development. Based on this, Samarzija et al. found that an autocrine Hh signalling loop could exist in cervical cancer cell lines 100.
Concerning HPV, there are two contrary results. One indicates that Hh signalling is not induced directly by HPV‐encoded proteins but rather that Hh‐activating mutations are selected in cells initially immortalized by HPV 100. The other is activation of the Hedgehog pathway being due to PTCH1 inactivation, along with HPV infection as important in CACX development 101.
The Hedgehog signalling pathway is associated with poor outcome in women with node negative cervical cancer, treated with radiation 102. Thus some researchers support a role for the Hh pathway in repopulation after chemoradiation and suggest that SMO may be a valid therapeutic target 103. Recently, miRNAs have become a focus; Gli3 may be a target of miR‐506 exerting its anti‐proliferative function in cervical neoplasia 104.
Notch signalling
Notch is a transmembrane receptor tethered to the plasma membrane as a heterodimeric protein. There are four mammalian Notch receptors, Notch 1–4 and they bind to two distinct families of Notch ligands, Delta‐like (DLL1, DLL3 and DLL4) and Jagged‐like (JAG1, JAG2). Signals are transduced by the Notch intracellular domain (NICD) produced by ligand‐induced cleavage, which translocates to the nucleus and directly induces transcription of downstream targets by forming a transactivation complex with several cofactors 105 (Fig. 6).
Figure 6.
Receptors and ligands of Notch pathway (cited from Maillard I, Fang T, Pear WS (2005) Regulation of lymphoid development, differentiation, and function by the Notch pathway. Annu. Rev. Immunol. 23, 945–974).
Notch is a cell to cell signalling pathway communication system, known to play a critical role in regulating cell proliferation and differentiation, during embryogenesis and in normal adult stem cells 106. Notch signalling determines which progenitor cells will be committed to a certain developmental route and which remain uncommitted and capable of differentiating into various cell types 107, 108. It is also involved in regulation of a set of neural stem cells 106.
Notch pathways are known to be abnormal in several cancer subtypes 109, 110, 111, 112, 113, 114 and its activation in HSCs in culture by Jagged‐1 ligand, consistently increases amounts of primitive progenitor activity observed in vitro and in vivo, suggesting that Notch activation promotes stem cell self‐renewal, or at least maintenance of multipotentiality 98, 115. It has also been suggested that interference in function of the Notch system is responsible for T‐cell leukaemia and breast cancers 107.
Notch signalling may have different roles during early and late stages of cervical cancer development 116. Reduction in Notch1 expression directs basal cells to cease terminal differentiation and to form immature epithelium, thereby playing a major role in histopathogenesis of epithelial dysplasia 117. Furthermore, down‐regulation of Notch1 expression seems to be an inherent mechanism for switching epithelium from the normal mature state to an activated immature one, suggesting its essential role in maintaining epithelial integrity 118. Notch‐1 receptor expression increases with progression of cervical cancer 119. Regulation of the Notch signalling pathway by HPV E6 has been demonstrated in cervical cancer cell lines. At late passage of W12 cell line, Jagged‐1 is up‐regulated, while Manic Fringe is reduced 120; silencing Jagged‐1 inhibits tumourigenicity of CaSki. However, a controversial report has shown that Notch 1 is low in HPV‐positive cervical cancer cell lines HeLa, C40I, C4‐II, SiHa and Caski compared to HPV‐negative C33a and primary keratinocytes 121, 122.
Wnt signalling
Wnts compose a family of 19 glycoproteins that act as ligands for the Frizzled (Fz) transmembrane receptor. Binding of Wnt ligands to Fz receptors activates two distinct signal transduction pathways, known as the ‘canonical’ and ‘non‐canonical’ Wnt pathways. The canonical pathway causes accumulation of β‐catenin (a downstream activator of the Wnt signalling pathway) in the cell nucleus and consequent transcription of Wnt target genes. Ordinarily, β‐catenin levels are maintained at low levels in the cytoplasm, but activation of the Wnt/ β‐catenin pathway causes translocation and accumulation of β‐catenin in the nucleus, thereby promoting transcription of Wnt target genes (Fig. 7).
Figure 7.
Relationship between cervical cancer and Wnt pathway (cited from Rath G, Jawanjal P, Salhan S, Nalliah M, Dhawan I (2015) Clinical significance of inactivated glycogen synthase kinase 3beta in HPV‐associated cervical cancer: Relationship with Wnt/beta‐catenin pathway activation. Am. J. Reprod. Immunol. 73, 460–478).
The Wnt signalling pathway is known to play key roles in controlling cell proliferation and differentiation in both embryogenesis and in regulating homoeostasis in normal adult tissues 123, 124, 125. Reya et al. demonstrated that overexpression of activated β‐catenin in long‐term cultures of transplantable HSCs, and ectopic expression of axin, lead to inhibition of HSC proliferation, their increased death and reduced reconstitution in vivo 126. In separate studies, soluble Wnt proteins from conditioned supernatants have also been shown to influence proliferation of hematopoietic progenitors from mouse foetal liver and human bone marrow 127, 128, and studies of epidermal and gut progenitors suggest that the Wnt signalling pathway may contribute to regulation of stem cell/progenitor cell self‐renewal in other tissues. Cultured human keratinocytes with higher proliferative potential have higher levels of β‐catenin compared to keratinocytes, which have a lower proliferative capacity. Moreover, retroviral transduction of activated β‐catenin results in increased epidermal stem cell self‐renewal and reduced differentiation 129. In vivo data from transgenic mice suggest that activation of the Wnt signalling pathway in epidermal stem cells can give rise to epithelial cancers 130. Furthermore, mice lacking TCF‐4, one of the transcriptional mediators of the Wnt signalling pathway, quickly exhaust undifferentiated progenitors in crypts of gut epithelium during foetal development 131, suggesting that this pathway is required for maintenance or self‐renewal of gut epithelial stem cells.
In addition, Wnt/β‐catenin signalling has been shown to be of importance in CSCs in a number of malignancies, such as colon cancer 132 and cutaneous squamous cell carcinoma 133.
Multiple therapeutic interventions have been proposed related to cervical cancer and the Wnt pathway. Activation of canonical Wnt at multiple levels (plasma, membrane, cytoplasm or nucleus) specifically supports transformation of HPV‐infected primary human keratinocytes 134. Wnt inhibitory factor 1 induces apoptosis and inhibits cervical cancer growth, invasion and angiogenesis in vivo 135, 136, and expression of Wnt7A 137, SOX1 138,Wnt‐11 139,Wnt5A 140 and nuclear β‐catenin 141 are related to cervical cancer survival and chemo‐/ radioresistance. Generation of HPV‐16‐associated cervical tumourigenesis synergizes with overactivation of the Wnt/ β‐catenin pathway 142. The mechanism of this has been associated with inactivation of negative regulators such as APC or axin 143, and overexpression of Dishevelled 144, frizzled receptors and Wnt ligands 145, 146.
These reports demonstrate that Wnt and Notch signalling are frequently co‐activated in human CC cells and specimens, and resveratrol can inhibit signalling activation, meanwhile leading to cervical squamous cell carcinoma and adenocarcinoma cells growth arrest and apoptosis 147.
The cancer stem cell niche
The ‘stem cell niche’ was proposed by Schofield in 1978 during research on processes of HSC self‐renewal and differentiation. The neighbouring physiological microenvironment of specialized cells, physically anchors stem cells and provides factors necessary for maintenance of stem cell characteristics 148.
Anatomical structure of the stem cell niche was first described in the study of germline stem cell (GSC) niches in adult Drosophila testis and ovary 149. In the testis, GSCs adhering to niche components receive signals supporting self‐renewal, while those further away are initiated to differentiation 150. Niches have also been identified in mammalian stem cells in the intestinal, neural, epidermal and hematopoietic systems. It has been widely acknowledged that HSCs may have two niches, osteoblast and vascular 151. The major signal pathways that have been verified in niches are Hedgehog, Wnt, BMPs, FGF (fibroblast growth factor) and Notch.
In the light of findings of stem cell niches, existence of cancer stem cell niches has been proposed. Putative existence of a cancer stem cell niche consisting of bi‐directional stromal and stem cell secreting factors that trigger cancer stem cell growth and proliferation, has been hypothesized in the nervous and hematopoietic systems 152, 153. Interactions between cancer stem cells and their niches may initiate some typical attributes of tumour cells. The surrounding microenvironment of cancer stem cells is composed of stromal fibroblasts, adipocytes, endothelial cells, extracellular matrix (ECM) and the immune system.
The microenvironment of cancer stem cells may be the key to maintaining their self‐renewal. Specialized microenvironments of bone marrow endothelial cells have proved to be crucial to homing and engraftment of both normal HSCs and leukaemic cells 148, 154. Extracellular matrix components and signalling molecules in the HSC microenvironment promote cell survival in AML, providing resistance to chemotherapeutic treatments 148, 155.
A more significant example comes from the glioma stem cell and its microenvironment. It has been verified that intrinsic properties of glioma stem cells are tightly regulated by specific signals from the niches 156. The signals help to maintain number of glioma stem cells and their undifferentiated state. Hypoxia is a major characteristic of the glioma stem cell niche. A number of studies has demonstrated that a hypoxic niche may play an important role in maintaining stemness of glioma stem cells and in promoting the glioma stem cell pool. Moreover, relationships between cancer stem cells and their niches can be bi‐directional. Glioma stem cells do not simply exploit pre‐existing microenvironments, but are also actively involved in shaping and generation of their niches through intricate crosstalk with various components of both surrounding and distant tissues 156.
Components of cancer stem cell niches may be highly related to metastatic potential of cancer stem cells 148. For example, MMP9 is induced in clusters of pre‐metastatic lung endothelial cells through VEGF Receptor 1 (VEGFR1) signalling from distant primary tumours 151. In addition, adhesion molecules and integrins may be associated with migration of CSCs 157.
At the moment, the transformation zone of cervix is considered to be a baseline lymphangiogenic niche in the cervical neoplastic process 157.
Cancer stem cell‐directed therapies
Resistance of CSCs to conventional cancer treatments such as chemotherapy and radiatiotherapy is considered to be a formidable problem as residual CSCs presumably trigger relapse after termination of treatment. Development of new therapeutic strategies based on the CSC model has therefore become a key goal in the challenge to achieve complete eradication of cancer. To date, four strategies have been considered.
The first is to attack CSCs directly. CSCs can be targeted by agents that kill them specifically, or that promote their differentiation into non‐CSCs, which in turn undergo apoptosis, senescence or terminal differentiation.The second strategy for CSC eradication is to promote their exit from the quiescent state and entry into the cycling state, and the third is to attack the CSC niche, which provides the supportive environment for maintenance of CSCs.The fourth strategy is to inhibit transitions between CSCs and non‐CSCs 158.
Considering cervical carcinoma, sorting markers or signalling pathway has not been applied in clinical practice. In addition, gene silencing of E6 with a lentiviral short hairpin RNA (shRNA) was found to profoundly inhibit HeLa‐SFC sphere formation and cell growth 65.
Conclusion
Study of cervical cancer stem cells is still at an early stage, and many problems remain to be solved. For example, there are abundant matrix and fibrous components of cervical tissue, which render it difficult to isolate tumour cells. The study of cervical stem cells is limited at present, and there are no internationally recognized markers of cervical stem cells. Finally, cervical cancer stem cells have not been isolated, and identifying their specific membrane markers is key to their isolation.
Conflicts of interest
The authors declare no conflicts of interest.
Acknowledgements
This work was supported by National Natural Science Foundation of China (81101979 and 81572575), Guangdong province Natural Scientific Grant (S2011040004639, S2012020010913 and 2014A030313109), Science and Technology Planning Key Project of Guangdong Province (2013B021800234), Guangdong College Students' innovation and entrepreneurship training program (1055813194), National College Students' innovation and entrepreneurship training program (201310558097), PhD Program Foundation of China education minister (20120171110097).
References
- 1. Al‐Hajj M, Wicha MS, Benito‐Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proc. Natl Acad. Sci. USA 100, 3983–3988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ (2005) Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 65, 10946–10951. [DOI] [PubMed] [Google Scholar]
- 3. Friel AM, Sergent PA, Patnaude C, Szotek PP, Oliva E, Scadden DT et al (2008) Functional analyses of the cancer stem cell‐like properties of human endometrial tumor initiating cells. Cell Cycle 7, 242–249. [DOI] [PubMed] [Google Scholar]
- 4. Ho MM, Ng AV, Lam S, Hung JY (2007) Side population in human lung cancer cell lines and tumors is enriched with stem‐like cancer cells. Cancer Res. 67, 4827–4833. [DOI] [PubMed] [Google Scholar]
- 5. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres‐Cortes J et al (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645–648. [DOI] [PubMed] [Google Scholar]
- 6. Marx J (2003) Cancer research. Mutant stem cells may seed cancer. Science 301, 1308–1310. [DOI] [PubMed] [Google Scholar]
- 7. Matsui W, Huff CA, Wang Q, Malehorn MT, Barber J, Tanhehco Y et al (2004) Characterization of clonogenic multiple myeloma cells. Blood 103, 2332–2336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Pardal R, Clarke MF, Morrison SJ (2003) Applying the principles of stem‐cell biology to cancer. Nat. Rev. Cancer 3, 895–902. [DOI] [PubMed] [Google Scholar]
- 9. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, cancer, and cancer stem cells. Nature 414, 105–111. [DOI] [PubMed] [Google Scholar]
- 10. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T et al (2004) Identification of human brain tumour initiating cells. Nature 432, 396–401. [DOI] [PubMed] [Google Scholar]
- 11. Szotek PP, Pieretti‐Vanmarcke R, Masiakos PT, Dinulescu DM, Connolly D, Foster R et al (2006) Ovarian cancer side population defines cells with stem cell‐like characteristics and Mullerian Inhibiting Substance responsiveness. Proc. Natl Acad. Sci. USA 103, 11154–11159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Zhang S, Balch C, Chan MW, Lai H‐C, Matei D, Schilder JM et al (2008) Identification and characterization of ovarian cancer‐initiating cells from primary human tumors. Cancer Res. 68, 4311–4320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL et al (2006) Cancer stem cells–perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 66, 9339–9344. [DOI] [PubMed] [Google Scholar]
- 14. Saini V, Shoemaker RH (2010) Potential for therapeutic targeting of tumor stem cells. Cancer Sci. 101, 16–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Al‐Hajj M, Becker MW, Wicha M, Weissman I, Clarke MF (2004) Therapeutic implications of cancer stem cells. Curr. Opin. Genet. Dev. 14, 43–47. [DOI] [PubMed] [Google Scholar]
- 16. Clarke RB, Anderson E, Howell A, Potten CS (2003) Regulation of human breast epithelial stem cells. Cell Prolif. 36(Suppl 1), 45–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Taipale J, Beachy PA (2001) The Hedgehog and Wnt signalling pathways in cancer. Nature 411, 349–354. [DOI] [PubMed] [Google Scholar]
- 18. Gournay J, Auvigne I, Pichard V, Ligeza C, Bralet MP, Ferry N (2002) In vivo cell lineage analysis during chemical hepatocarcinogenesis in rats using retroviral‐mediated gene transfer: evidence for dedifferentiation of mature hepatocytes. Lab. Invest. 82, 781–788. [DOI] [PubMed] [Google Scholar]
- 19. Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, Vogel S et al (2005) Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 121, 823–835. [DOI] [PubMed] [Google Scholar]
- 20. Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, Hotz S et al (2005) A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res. 65, 9328–9337. [DOI] [PubMed] [Google Scholar]
- 21. Hirschmann‐Jax C, Foster AE, Wulf GG, Nuchtern JG, Jax TW, Gobel U et al (2004) A distinct “side population” of cells with high drug efflux capacity in human tumor cells. Proc. Natl Acad. Sci. USA 101, 14228–14233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Kondo T, Setoguchi T, Taga T (2004) Persistence of a small subpopulation of cancer stem‐like cells in the C6 glioma cell line. Proc. Natl Acad. Sci. USA 101, 781–786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Delvenne P, Herman L, Kholod N, Caberg JH, Herfs M, Boniver J et al (2007) Role of hormone cofactors in the human papillomavirus‐induced carcinogenesis of the uterine cervix. Mol. Cell. Endocrinol. 264, 1–5. [DOI] [PubMed] [Google Scholar]
- 24. Martens JE, Smedts FM, Ploeger D, Helmerhorst TJ, Ramaekers FC, Arends JW et al (2009) Distribution pattern and marker profile show two subpopulations of reserve cells in the endocervical canal. Int. J. Gynecol. Pathol. 28, 381–388. [DOI] [PubMed] [Google Scholar]
- 25. Martens JE, Smedts F, van Muyden RC, Schoots C, Helmerhorst TJ, Hopman A et al (2007) Reserve cells in human uterine cervical epithelium are derived from mullerian epithelium at midgestational age. Int. J. Gynecol. Pathol. 26, 463–468. [DOI] [PubMed] [Google Scholar]
- 26. Herfs M, Yamamoto Y, Laury A, Wang X, Nucci MR, McLaughlin‐Drubin ME et al (2012) A discrete population of squamocolumnar junction cells implicated in the pathogenesis of cervical cancer. Proc. Natl Acad. Sci. USA 109, 10516–10521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Lopez J, Ruiz G, Organista‐Nava J, Gariglio P, Garcia‐Carranca A (2012) Human papillomavirus infections and cancer stem cells of tumors from the uterine cervix. Open Virol. J. 6, 232–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Herfs M, Vargas SO, Yamamoto Y, Howitt BE, Nucci MR, Hornick JL et al (2013) A novel blueprint for ‘top down’ differentiation defines the cervical squamocolumnar junction during development, reproductive life, and neoplasia. J. Pathol. 229, 460–468. [DOI] [PubMed] [Google Scholar]
- 29. Organista‐Nava J, Gomez‐Gomez Y, Gariglio P (2014) Embryonic stem cell‐specific signature in cervical cancer. Tumour Biol. 35, 1727–1738. [DOI] [PubMed] [Google Scholar]
- 30. Brankovic I, Verdonk P, Klinge I (2013) Applying a gender lens on human papillomavirus infection: cervical cancer screening, HPV DNA testing, and HPV vaccination. Int. J. Equity Health 12, 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Rijkaart DC, Berkhof J, van Kemenade FJ, Coupe VM, Rozendaal L, Heideman DA et al (2012) HPV DNA testing in population‐based cervical screening (VUSA‐Screen study): results and implications. Br. J. Cancer 106, 975–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Melsheimer P, Vinokurova S, Wentzensen N, Bastert G, von Knebel Doeberitz M (2004) DNA aneuploidy and integration of human papillomavirus type 16 e6/e7 oncogenes in intraepithelial neoplasia and invasive squamous cell carcinoma of the cervix uteri. Clin. Cancer Res. 10, 3059–3063. [DOI] [PubMed] [Google Scholar]
- 33. Fisher JW, Brundage SI (2009) The challenge of eliminating cervical cancer in the United States: a story of politics, prudishness, and prevention. Women Health 49, 246–261. [DOI] [PubMed] [Google Scholar]
- 34. Peterson NB, Murff HJ, Cui Y, Hargreaves M, Fowke JH (2008) Papanicolaou testing among women in the southern United States. J. Womens Health (Larchmt) 17, 939–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Eggleston KS, Coker AL, Williams M, Tortolero‐Luna G, Martin JB, Tortolero SR (2006) Cervical cancer survival by socioeconomic status, race/ethnicity, and place of residence in Texas, 1995‐2001. J. Womens Health (Larchmt) 15, 941–951. [DOI] [PubMed] [Google Scholar]
- 36. Coker AL, Desimone CP, Eggleston KS, White AL, Williams M (2009) Ethnic disparities in cervical cancer survival among Texas women. J. Womens Health (Larchmt) 18, 1577–1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Raychaudhuri S, Mandal S (2012) Current status of knowledge, attitude and practice (KAP) and screening for cervical cancer in countries at different levels of development. Asian Pac. J. Cancer Prev. 13, 4221–4227. [DOI] [PubMed] [Google Scholar]
- 38. Khan S, Jaffer NN, Khan MN, Rai MA, Shafiq M, Ali A et al (2007) Human papillomavirus subtype 16 is common in Pakistani women with cervical carcinoma. Int. J. Infect. Dis. 11, 313–317. [DOI] [PubMed] [Google Scholar]
- 39. Martens JE, Arends J, Van der Linden PJ, De Boer BA, Helmerhorst TJ (2004) Cytokeratin 17 and p63 are markers of the HPV target cell, the cervical stem cell. Anticancer Res. 24, 771–775. [PubMed] [Google Scholar]
- 40. Patanwala IY, Bauer HM, Miyamoto J, Park IU, Huchko MJ, Smith‐McCune KK (2013) A systematic review of randomized trials assessing human papillomavirus testing in cervical cancer screening. Am. J. Obstet. Gynecol. 208, 343–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S et al (2003) Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell 113, 643–655. [DOI] [PubMed] [Google Scholar]
- 42. Hyslop L, Stojkovic M, Armstrong L, Walter T, Stojkovic P, Przyborski S et al (2005) Downregulation of NANOG induces differentiation of human embryonic stem cells to extraembryonic lineages. Stem Cells 23, 1035–1043. [DOI] [PubMed] [Google Scholar]
- 43. Armstrong L, Hughes O, Yung S, Hyslop L, Stewart R, Wappler I et al (2006) The role of PI3K/AKT, MAPK/ERK and NFkappabeta signalling in the maintenance of human embryonic stem cell pluripotency and viability highlighted by transcriptional profiling and functional analysis. Hum. Mol. Genet. 15, 1894–1913. [DOI] [PubMed] [Google Scholar]
- 44. Takaishi S, Okumura T, Wang TC (2008) Gastric cancer stem cells. J. Clin. Oncol. 26, 2876–2882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Sato A, Okada M, Shibuya K, Watanabe E, Seino S, Suzuki K et al (2013) Resveratrol promotes proteasome‐dependent degradation of Nanog via p53 activation and induces differentiation of glioma stem cells. Stem Cell. Res. 11, 601–610. [DOI] [PubMed] [Google Scholar]
- 46. Nirasawa S, Kobayashi D, Tsuji N, Kuribayashi K, Watanabe N (2009) Diagnostic relevance of overexpressed Nanog gene in early lung cancers. Oncol. Rep. 22, 587–591. [DOI] [PubMed] [Google Scholar]
- 47. Fujii H, Honoki K, Tsujiuchi T, Kido A, Yoshitani K, Takakura Y (2009) Sphere‐forming stem‐like cell populations with drug resistance in human sarcoma cell lines. Int. J. Oncol. 34, 1381–1386. [PubMed] [Google Scholar]
- 48. Ye F, Zhou C, Cheng Q, Shen J, Chen H (2008) Stem‐cell‐abundant proteins Nanog, Nucleostemin and Musashi1 are highly expressed in malignant cervical epithelial cells. BMC Cancer 8, 108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Gu TT, Liu SY, Zheng PS (2012) Cytoplasmic NANOG‐positive stromal cells promote human cervical cancer progression. Am. J. Pathol. 181, 652–661. [DOI] [PubMed] [Google Scholar]
- 50. Ali N, Allam H, May R, Sureban SM, Bronze MS, Bader T et al (2011) Hepatitis C virus‐induced cancer stem cell‐like signatures in cell culture and murine tumor xenografts. J. Virol. 85, 12292–12303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Ikeda K, Tate G, Suzuki T, Mitsuya T (2008) Coordinate expression of cytokeratin 8 and cytokeratin 17 immunohistochemical staining in cervical intraepithelial neoplasia and cervical squamous cell carcinoma: an immunohistochemical analysis and review of the literature. Gynecol. Oncol. 108, 598–602. [DOI] [PubMed] [Google Scholar]
- 52. Smedts F, Ramaekers F, Leube RE, Keijser K, Link M, Vooijs P (1993) Expression of keratins 1, 6, 15, 16, and 20 in normal cervical epithelium, squamous metaplasia, cervical intraepithelial neoplasia, and cervical carcinoma. Am. J. Pathol. 142, 403–412. [PMC free article] [PubMed] [Google Scholar]
- 53. Smedts F, Ramaekers FC, Vooijs PG (1993) The dynamics of keratin expression in malignant transformation of cervical epithelium: a review. Obstet. Gynecol. 82, 465. [PubMed] [Google Scholar]
- 54. Wang HY, Sun JM, Lu HF, Shi DR, Ou ZL, Ren YL et al (2006) Micrometastases detected by cytokeratin 19 expression in sentinel lymph nodes of patients with early‐stage cervical cancer. Int. J. Gynecol. Cancer 16, 643–648. [DOI] [PubMed] [Google Scholar]
- 55. Kurahashi T, Hara I, Oka N, Kamidono S, Eto H, Miyake H (2005) Detection of micrometastases in pelvic lymph nodes in patients undergoing radical cystectomy for locally invasive bladder cancer by real‐time reverse transcriptase‐PCR for cytokeratin 19 and uroplakin II. Clin. Cancer Res. 11, 3773–3777. [DOI] [PubMed] [Google Scholar]
- 56. Nagano O, Saya H (2004) Mechanism and biological significance of CD44 cleavage. Cancer Sci. 95, 930–935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Cheng C, Sharp PA (2006) Regulation of CD44 alternative splicing by SRm160 and its potential role in tumor cell invasion. Mol. Cell. Biol. 26, 362–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Vigetti D, Viola M, Karousou E, Rizzi M, Moretto P, Genasetti A et al (2008) Hyaluronan‐CD44‐ERK1/2 regulate human aortic smooth muscle cell motility during aging. J. Biol. Chem. 283, 4448–4458. [DOI] [PubMed] [Google Scholar]
- 59. Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B (1990) CD44 is the principal cell surface receptor for hyaluronate. Cell 61, 1303–1313. [DOI] [PubMed] [Google Scholar]
- 60. Patrawala L, Calhoun T, Schneider‐Broussard R, Li H, Bhatia B, Tang S et al (2006) Highly purified CD44 + prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene 25, 1696–1708. [DOI] [PubMed] [Google Scholar]
- 61. Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW et al (2007) Phenotypic characterization of human colorectal cancer stem cells. Proc. Natl Acad. Sci. USA 104, 10158–10163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Yeung TM, Gandhi SC, Wilding JL, Muschel R, Bodmer WF (2010) Cancer stem cells from colorectal cancer‐derived cell lines. Proc. Natl Acad. Sci. USA 107, 3722–3727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Charafe‐Jauffret E, Ginestier C, Iovino F, Wicinski J, Cervera N, Finetti P et al (2009) Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. Cancer Res. 69, 1302–1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Li F (2009) Every single cell clones from cancer cell lines growing tumors in vivo may not invalidate the cancer stem cell concept. Mol. Cells 27, 491–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Gu W, Yeo E, McMillan N, Yu C (2011) Silencing oncogene expression in cervical cancer stem‐like cells inhibits their cell growth and self‐renewal ability. Cancer Gene Ther. 18, 897–905. [DOI] [PubMed] [Google Scholar]
- 66. Ben‐Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A et al (2008) An embryonic stem cell‐like gene expression signature in poorly differentiated aggressive human tumors. Nat. Genet. 40, 499–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Friedman S, Lu M, Schultz A, Thomas D, Lin RY (2009) CD133 + anaplastic thyroid cancer cells initiate tumors in immunodeficient mice and are regulated by thyrotropin. PLoS ONE 4, e5395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Du Z, Jia D, Liu S, Wang F, Li G, Zhang Y et al (2009) Oct4 is expressed in human gliomas and promotes colony formation in glioma cells. Glia 57, 724–733. [DOI] [PubMed] [Google Scholar]
- 69. Hu T, Liu S, Breiter DR, Wang F, Tang Y, Sun S (2008) Octamer 4 small interfering RNA results in cancer stem cell‐like cell apoptosis. Cancer Res. 68, 6533–6540. [DOI] [PubMed] [Google Scholar]
- 70. de Resende MF, Chinen LT, Vieira S, Jampietro J, da Fonseca FP, Vassallo J et al (2013) Prognostication of OCT4 isoform expression in prostate cancer. Tumour Biol. 34, 2665–2673. [DOI] [PubMed] [Google Scholar]
- 71. Wang P, Gao Q, Suo Z, Munthe E, Solberg S, Ma L et al (2013) Identification and characterization of cells with cancer stem cell properties in human primary lung cancer cell lines. PLoS ONE 8, e57020. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 72. Dong Z, Zeng Q, Luo H, Zou J, Cao C, Liang J et al (2012) Increased expression of OCT4 is associated with low differentiation and tumor recurrence in human hepatocellular carcinoma. Pathol. Res. Pract. 208, 527–533. [DOI] [PubMed] [Google Scholar]
- 73. Cheung AN, Tsun KL, Ng KM, Szeto E, Siu MK, Wong ES et al (2010) P634A4 and TAp73 immunocytochemistry in liquid‐based cervical cytology–potential biomarkers for diagnosis and progress prediction of cervical neoplasia. Mod. Pathol. 23, 559–566. [DOI] [PubMed] [Google Scholar]
- 74. Feng D, Peng C, Li C, Zhou Y, Li M, Ling B et al (2009) Identification and characterization of cancer stem‐like cells from primary carcinoma of the cervix uteri. Oncol. Rep. 22, 1129–1134. [DOI] [PubMed] [Google Scholar]
- 75. Liu D, Zhou P, Zhang L, Wu G, Zheng Y, He F (2011) Differential expression of Oct4 in HPV‐positive and HPV‐negative cervical cancer cells is not regulated by DNA methyltransferase 3A. Tumour Biol. 32, 941–950. [DOI] [PubMed] [Google Scholar]
- 76. Ginestier C, Hur MH, Charafe‐Jauffret E, Monville F, Dutcher J, Brown M et al (2007) 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Tanei T, Morimoto K, Shimazu K, Kim SJ, Tanji Y, Taguchi T et al (2009) Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin‐based chemotherapy for breast cancers. Clin. Cancer Res. 15, 4234–4241. [DOI] [PubMed] [Google Scholar]
- 78. Rao QX, Yao TT, Zhang BZ, Lin RC, Chen ZL, Zhou H et al (2012) Expression and functional role of ALDH1 in cervical carcinoma cells. Asian Pac. J. Cancer Prev. 13, 1325–1331. [DOI] [PubMed] [Google Scholar]
- 79. Yao T, Chen Q, Zhang B, Zhou H, Lin Z (2011) The expression of ALDH1 in cervical carcinoma. Med. Sci. Monit. 17, Hy21–Hy26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Serrano M (1997) The tumor suppressor protein p16INK4a. Exp. Cell Res. 237, 7–13. [DOI] [PubMed] [Google Scholar]
- 81. Stott FJ, Bates S, James MC, McConnell BB, Starborg M, Brookes S et al (1998) The alternative product from the human CDKN2A locus, p14(ARF), participates in a regulatory feedback loop with p53 and MDM2. EMBO J. 17, 5001–5014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Herman JG, Merlo A, Mao L, Lapidus RG, Issa JP, Davidson NE et al (1995) Inactivation of the CDKN2/p16/MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers. Cancer Res. 55, 4525–4530. [PubMed] [Google Scholar]
- 83. Liu HQ, Wang YH, Wang LL, Hao M (2015) P16INK4A and survivin: Diagnostic and prognostic markers in cervical intraepithelial neoplasia and cervical squamous cell carcinoma. Exp. Mol. Pathol. 99, 44–49. [DOI] [PubMed] [Google Scholar]
- 84. Bergeron C, Ikenberg H, Sideri M, Denton K, Bogers J, Schmidt D et al (2015) Prospective evaluation of p16/Ki‐67 dual‐stained cytology for managing women with abnormal Papanicolaou cytology: PALMS study results. Cancer Cytopathol. 123, 373–381. [DOI] [PubMed] [Google Scholar]
- 85. Wang JL, Zheng BY, Li XD, Nokelainen K, Angstrom T, Lindstrom MS et al (2005) p16INK4A and p14ARF expression pattern by immunohistochemistry in human papillomavirus‐related cervical neoplasia. Mod. Pathol. 18, 629–637. [DOI] [PubMed] [Google Scholar]
- 86. Huang LW, Pan HS, Lin YH, Seow KM, Chen HJ, Hwang JL (2011) P16 methylation is an early event in cervical carcinogenesis. Int. J. Gynecol. Cancer 21, 452–456. [DOI] [PubMed] [Google Scholar]
- 87. Li Y, Xiao S, Dan L, Xue M (2015) P16 is required for cisplatin resistance in cervical carcinoma SiHa cells. Oncol Lett. 9, 1104–1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Alshenawy HA (2014) Evaluation of p16, human papillomavirus capsid protein L1 and Ki‐67 in cervical intraepithelial lesions: potential utility in diagnosis and prognosis. Pathol. Res. Pract. 210, 916–921. [DOI] [PubMed] [Google Scholar]
- 89. Liao GD, Kang LN, Chen W, Zhang X, Liu XY, Zhao FH et al (2014) p16 Immunohistochemistry Interpretation by Nonpathologists as an Accurate Method for Diagnosing Cervical Precancer and Cancer. J. Low Genit. Tract. Dis. 19, 207–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Demir F, Kimiloglu E, Igdem AA, Ayanoglu YT, Erdogan N (2014) High risk HPV in situ hybridization, p16 INK 4A, and survivin expressions in cervical carcinomas and intraepithelial neoplasms: evaluation of prognostic factors. Eur. J. Gynaecol. Oncol. 35, 708–717. [PubMed] [Google Scholar]
- 91. Yang A, Kaghad M, Wang Y, Gillett E, Fleming MD, Dotsch V et al (1998) p63, a p53 homolog at 3q27‐29, encodes multiple products with transactivating, death‐inducing, and dominant‐negative activities. Mol. Cell 2, 305–316. [DOI] [PubMed] [Google Scholar]
- 92. Quade BJ, Yang A, Wang Y, Sun D, Park J, Sheets EE et al (2001) Expression of the p53 homologue p63 in early cervical neoplasia. Gynecol. Oncol. 80, 24–29. [DOI] [PubMed] [Google Scholar]
- 93. Yang A, Schweitzer R, Sun D, Kaghad M, Walker N, Bronson RT et al (1999) p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development. Nature 398, 714–718. [DOI] [PubMed] [Google Scholar]
- 94. Wang BY, Gil J, Kaufman D, Gan L, Kohtz DS, Burstein DE (2002) P63 in pulmonary epithelium, pulmonary squamous neoplasms, and other pulmonary tumors. Hum. Pathol. 33, 921–926. [DOI] [PubMed] [Google Scholar]
- 95. Shah RB, Zhou M, LeBlanc M, Snyder M, Rubin MA (2002) Comparison of the basal cell‐specific markers, 34betaE12 and p63, in the diagnosis of prostate cancer. Am. J. Surg. Pathol. 26, 1161–1168. [DOI] [PubMed] [Google Scholar]
- 96. Jiang J, Hui CC (2008) Hedgehog signaling in development and cancer. Dev. Cell 15, 801–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Ingham PW (1998) Transducing Hedgehog: the story so far. EMBO J. 17, 3505–3511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Bhardwaj G, Murdoch B, Wu D, Baker DP, Williams KP, Chadwick K et al (2001) Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation. Nat. Immunol. 2, 172–180. [DOI] [PubMed] [Google Scholar]
- 99. Xuan YH, Jung HS, Choi YL, Shin YK, Kim HJ, Kim KH et al (2006) Enhanced expression of hedgehog signaling molecules in squamous cell carcinoma of uterine cervix and its precursor lesions. Mod. Pathol. 19, 1139–1147. [DOI] [PubMed] [Google Scholar]
- 100. Samarzija I, Beard P (2012) Hedgehog pathway regulators influence cervical cancer cell proliferation, survival and migration. Biochem. Biophys. Res. Commun. 425, 64–69. [DOI] [PubMed] [Google Scholar]
- 101. Chakraborty C, Dutta S, Mukherjee N, Samadder S, Roychowdhury A, Roy A et al (2015) Inactivation of PTCH1 is associated with the development of cervical carcinoma: clinical and prognostic implication. Tumour Biol. 36, 1143–1154. [DOI] [PubMed] [Google Scholar]
- 102. Rofstad EK, Sundfor K, Lyng H, Trope CG (2000) Hypoxia‐induced treatment failure in advanced squamous cell carcinoma of the uterine cervix is primarily due to hypoxia‐induced radiation resistance rather than hypoxia‐induced metastasis. Br. J. Cancer 83, 354–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Chaudary N, Pintilie M, Hedley D, Fyles AW, Milosevic M, Clarke B et al (2012) Hedgehog pathway signaling in cervical carcinoma and outcome after chemoradiation. Cancer 118, 3105–3115. [DOI] [PubMed] [Google Scholar]
- 104. Wen SY, Lin Y, Yu YQ, Cao SJ, Zhang R, Yang XM et al (2015) miR‐506 acts as a tumor suppressor by directly targeting the hedgehog pathway transcription factor Gli3 in human cervical cancer. Oncogene 34, 717–725. [DOI] [PubMed] [Google Scholar]
- 105. Gordon WR, Vardar‐Ulu D, Histen G, Sanchez‐Irizarry C, Aster JC, Blacklow SC (2007) Structural basis for autoinhibition of Notch. Nat. Struct. Mol. Biol. 14, 295–300. [DOI] [PubMed] [Google Scholar]
- 106. Hitoshi S, Alexson T, Tropepe V, Donoviel D, Elia AJ, Nye JS et al (2002) Notch pathway molecules are essential for the maintenance, but not the generation, of mammalian neural stem cells. Genes Dev. 16, 846–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Kojika S, Griffin JD (2001) Notch receptors and hematopoiesis. Exp. Hematol. 29, 1041–1052. [DOI] [PubMed] [Google Scholar]
- 108. Artavanis‐Tsakonas S, Rand MD, Lake RJ (1999) Notch signaling: cell fate control and signal integration in development. Science 284, 770–776. [DOI] [PubMed] [Google Scholar]
- 109. Rizzo P, Miao H, D'Souza G, Osipo C, Song LL, Yun J et al (2008) Cross‐talk between notch and the estrogen receptor in breast cancer suggests novel therapeutic approaches. Cancer Res. 68, 5226–5235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Weng AP, Lau A (2005) Notch signaling in T‐cell acute lymphoblastic leukemia. Future Oncol. 1, 511–519. [DOI] [PubMed] [Google Scholar]
- 111. Salcido CD, Larochelle A, Taylor BJ, Dunbar CE, Varticovski L (2010) Molecular characterisation of side population cells with cancer stem cell‐like characteristics in small‐cell lung cancer. Br. J. Cancer 102, 1636–1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Rizzo P, Osipo C, Foreman K, Golde T, Osborne B, Miele L (2008) Rational targeting of Notch signaling in cancer. Oncogene 27, 5124–5131. [DOI] [PubMed] [Google Scholar]
- 113. Qiao L, Wong BC (2009) Role of Notch signaling in colorectal cancer. Carcinogenesis 30, 1979–1986. [DOI] [PubMed] [Google Scholar]
- 114. Sullivan JP, Spinola M, Dodge M, Raso MG, Behrens C, Gao B et al (2010) Aldehyde dehydrogenase activity selects for lung adenocarcinoma stem cells dependent on notch signaling. Cancer Res. 70, 9937–9948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Karanu FN, Murdoch B, Gallacher L, Wu DM, Koremoto M, Sakano S et al (2000) The notch ligand jagged‐1 represents a novel growth factor of human hematopoietic stem cells. J. Exp. Med. 192, 1365–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Wu L, Griffin JD (2004) Modulation of Notch signaling by mastermind‐like (MAML) transcriptional co‐activators and their involvement in tumorigenesis. Semin. Cancer Biol. 14, 348–356. [DOI] [PubMed] [Google Scholar]
- 117. Franko‐Tobin LG, Mackey LV, Huang W, Song X, Jin B, Luo J et al (2012) Notch1‐mediated tumor suppression in cervical cancer with the involvement of SST signaling and its application in enhanced SSTR‐targeted therapeutics. Oncologist 17, 220–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Sakamoto K, Fujii T, Kawachi H, Miki Y, Omura K, Morita K et al (2012) Reduction of NOTCH1 expression pertains to maturation abnormalities of keratinocytes in squamous neoplasms. Lab. Invest. 92, 688–702. [DOI] [PubMed] [Google Scholar]
- 119. Daniel B, Rangarajan A, Mukherjee G, Vallikad E, Krishna S (1997) The link between integration and expression of human papillomavirus type 16 genomes and cellular changes in the evolution of cervical intraepithelial neoplastic lesions. J. Gen. Virol. 78(Pt 5), 1095–1101. [DOI] [PubMed] [Google Scholar]
- 120. Veeraraghavalu K, Pett M, Kumar RV, Nair P, Rangarajan A, Stanley MA et al (2004) Papillomavirus‐mediated neoplastic progression is associated with reciprocal changes in JAGGED1 and manic fringe expression linked to notch activation. J. Virol. 78, 8687–8700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Talora C, Sgroi DC, Crum CP, Dotto GP (2002) Specific down‐modulation of Notch1 signaling in cervical cancer cells is required for sustained HPV‐E6/E7 expression and late steps of malignant transformation. Genes Dev. 16, 2252–2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Chen J (2015) Signaling pathways in HPV‐associated cancers and therapeutic implications. Rev. Med. Virol. 25(Suppl 1), 24–53. [DOI] [PubMed] [Google Scholar]
- 123. van der Flier LG, Clevers H (2009) Stem cells, self‐renewal, and differentiation in the intestinal epithelium. Annu. Rev. Physiol. 71, 241–260. [DOI] [PubMed] [Google Scholar]
- 124. Blanpain C, Horsley V, Fuchs E (2007) Epithelial stem cells: turning over new leaves. Cell 128, 445–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Andrade AC, Nilsson O, Barnes KM, Baron J (2007) Wnt gene expression in the post‐natal growth plate: regulation with chondrocyte differentiation. Bone 40, 1361–1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Reya T, O'Riordan M, Okamura R, Devaney E, Willert K, Nusse R et al (2000) Wnt signaling regulates B lymphocyte proliferation through a LEF‐1 dependent mechanism. Immunity 13, 15–24. [DOI] [PubMed] [Google Scholar]
- 127. Austin TW, Solar GP, Ziegler FC, Liem L, Matthews W (1997) A role for the Wnt gene family in hematopoiesis: expansion of multilineage progenitor cells. Blood 89, 3624–3635. [PubMed] [Google Scholar]
- 128. Van Den Berg DJ, Sharma AK, Bruno E, Hoffman R (1998) Role of members of the Wnt gene family in human hematopoiesis. Blood 92, 3189–3202. [PubMed] [Google Scholar]
- 129. Zhu AJ, Watt FM (1999) beta‐catenin signalling modulates proliferative potential of human epidermal keratinocytes independently of intercellular adhesion. Development 126, 2285–2298. [DOI] [PubMed] [Google Scholar]
- 130. Gat U, DasGupta R, Degenstein L, Fuchs E (1998) De Novo hair follicle morphogenesis and hair tumors in mice expressing a truncated beta‐catenin in skin. Cell 95, 605–614. [DOI] [PubMed] [Google Scholar]
- 131. Korinek V, Barker N, Moerer P, van Donselaar E, Huls G, Peters PJ et al (1998) Depletion of epithelial stem‐cell compartments in the small intestine of mice lacking Tcf‐4. Nat. Genet. 19, 379–383. [DOI] [PubMed] [Google Scholar]
- 132. Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H, van den Born M et al (2009) Crypt stem cells as the cells‐of‐origin of intestinal cancer. Nature 457, 608–611. [DOI] [PubMed] [Google Scholar]
- 133. Malanchi I, Peinado H, Kassen D, Hussenet T, Metzger D, Chambon P et al (2008) Cutaneous cancer stem cell maintenance is dependent on beta‐catenin signalling. Nature 452, 650–653. [DOI] [PubMed] [Google Scholar]
- 134. Bulut G, Fallen S, Beauchamp EM, Drebing LE, Sun J, Berry DL et al (2011) Beta‐catenin accelerates human papilloma virus type‐16 mediated cervical carcinogenesis in transgenic mice. PLoS ONE 6, e27243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Delmas AL, Riggs BM, Pardo CE, Dyer LM, Darst RP, Izumchenko EG et al (2011) WIF1 is a frequent target for epigenetic silencing in squamous cell carcinoma of the cervix. Carcinogenesis 32, 1625–1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Ramachandran I, Thavathiru E, Ramalingam S, Natarajan G, Mills WK, Benbrook DM et al (2012) Wnt inhibitory factor 1 induces apoptosis and inhibits cervical cancer growth, invasion and angiogenesis in vivo. Oncogene 31, 2725–2737. [DOI] [PubMed] [Google Scholar]
- 137. Ramos‐Solano M, Meza‐Canales ID, Torres‐Reyes LA, Alvarez‐Zavala M, Alvarado‐Ruiz L, Rincon‐Orozco B et al (2015) Expression of WNT genes in cervical cancer‐derived cells: Implication of WNT7A in cell proliferation and migration. Exp. Cell Res. 335, 39–50. [DOI] [PubMed] [Google Scholar]
- 138. Lin YW, Tsao CM, Yu PN, Shih YL, Lin CH, Yan MD (2013) SOX1 suppresses cell growth and invasion in cervical cancer. Gynecol. Oncol. 131, 174–181. [DOI] [PubMed] [Google Scholar]
- 139. Wei H, Wang N, Zhang Y, Wang S, Pang X, Zhang J et al (2014) Clinical significance of Wnt‐11 and squamous cell carcinoma antigen expression in cervical cancer. Med. Oncol. 31, 933. [DOI] [PubMed] [Google Scholar]
- 140. Lin L, Liu Y, Zhao W, Sun B, Chen Q (2014) Wnt5A expression is associated with the tumor metastasis and clinical survival in cervical cancer. Int. J. Clin. Exp. Pathol. 7, 6072–6078. [PMC free article] [PubMed] [Google Scholar]
- 141. Zhang Y, Liu B, Zhao Q, Hou T, Huang X (2014) Nuclear localizaiton of beta‐catenin is associated with poor survival and chemo‐/radioresistance in human cervical squamous cell cancer. Int. J. Clin. Exp. Pathol. 7, 3908–3917. [PMC free article] [PubMed] [Google Scholar]
- 142. Rath G, Jawanjal P, Salhan S, Nalliah M, Dhawan I (2015) Clinical significance of inactivated glycogen synthase kinase 3beta in HPV‐associated cervical cancer: Relationship with Wnt/beta‐catenin pathway activation. Am. J. Reprod. Immunol. 73, 460–478. [DOI] [PubMed] [Google Scholar]
- 143. Yang HJ, Liu VW, Wang Y, Tsang PC, Ngan HY (2006) Differential DNA methylation profiles in gynecological cancers and correlation with clinico‐pathological data. BMC Cancer 6, 212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Okino K, Nagai H, Hatta M, Nagahata T, Yoneyama K, Ohta Y et al (2003) Up‐regulation and overproduction of DVL‐1, the human counterpart of the Drosophila dishevelled gene, in cervical squamous cell carcinoma. Oncol. Rep. 10, 1219–1223. [DOI] [PubMed] [Google Scholar]
- 145. Kirikoshi H, Katoh M (2002) Expression and regulation of WNT10B in human cancer: up‐regulation of WNT10B in MCF‐7 cells by beta‐estradiol and down‐regulation of WNT10B in NT2 cells by retinoic acid. Int. J. Mol. Med. 10, 507–511. [PubMed] [Google Scholar]
- 146. Perez‐Plasencia C, Vazquez‐Ortiz G, Lopez‐Romero R, Pina‐Sanchez P, Moreno J, Salcedo M (2007) Genome wide expression analysis in HPV16 cervical cancer: identification of altered metabolic pathways. Infect. Agent Cancer 2, 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Zhang P, Li H, Yang B, Yang F, Zhang LL, Kong QY et al (2014) Biological significance and therapeutic implication of resveratrol‐inhibited Wnt, Notch and STAT3 signaling in cervical cancer cells. Genes Cancer. 5, 154–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Sneddon JB, Werb Z (2007) Location, location, location: the cancer stem cell niche. Cell Stem Cell 1, 607–611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Voog J, Jones DL (2010) Stem cells and the niche: a dynamic duo. Cell Stem Cell 6, 103–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Li L, Xie T (2005) Stem cell niche: structure and function. Annu. Rev. Cell Dev. Biol. 21, 605–631. [DOI] [PubMed] [Google Scholar]
- 151. Kaplan RN, Psaila B, Lyden D (2007) Niche‐to‐niche migration of bone‐marrow‐derived cells. Trends Mol. Med. 13, 72–81. [DOI] [PubMed] [Google Scholar]
- 152. Ruiz‐Vela A, Aguilar‐Gallardo C, Simon C (2009) Building a framework for embryonic microenvironments and cancer stem cells. Stem Cell Rev. 5, 319–327. [DOI] [PubMed] [Google Scholar]
- 153. Perry JM, Li L (2007) Disrupting the stem cell niche: good seeds in bad soil. Cell 129, 1045–1047. [DOI] [PubMed] [Google Scholar]
- 154. Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B et al (2007) A perivascular niche for brain tumor stem cells. Cancer Cell 11, 69–82. [DOI] [PubMed] [Google Scholar]
- 155. De Toni F, Racaud‐Sultan C, Chicanne G, Mas VM, Cariven C, Mesange F et al (2006) A crosstalk between the Wnt and the adhesion‐dependent signaling pathways governs the chemosensitivity of acute myeloid leukemia. Oncogene 25, 3113–3122. [DOI] [PubMed] [Google Scholar]
- 156. Filatova A, Acker T, Garvalov BK (2013) The cancer stem cell niche(s): the crosstalk between glioma stem cells and their microenvironment. Biochim. Biophys. Acta 1830, 2496–2508. [DOI] [PubMed] [Google Scholar]
- 157. Balsat C, Signolle N, Goffin F, Delbecque K, Plancoulaine B, Sauthier P et al (2014) Improved computer‐assisted analysis of the global lymphatic network in human cervical tissues. Mod. Pathol. 27, 887–898. [DOI] [PubMed] [Google Scholar]
- 158. Sugihara E, Saya H (2013) Complexity of cancer stem cells. Int. J. Cancer 132, 1249–1259. [DOI] [PubMed] [Google Scholar]