Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

ISSN (Print): 1574-888X
ISSN (Online): 2212-3946

Review Article

Molecular Mechanisms Underlying the Functions of Cellular Markers Associated with the Phenotype of Cancer Stem Cells

Author(s): Eduardo Alvarado-Ortiz, Miguel Á. Sarabia-Sánchez and Alejandro García-Carrancá*

Volume 14, Issue 5, 2019

Page: [405 - 420] Pages: 16

DOI: 10.2174/1574888X13666180821154752

Price: $65

Abstract

Cancer Stem Cells (CSC) generally constitute a minor cellular population within tumors that exhibits some capacities of normal Stem Cells (SC). The existence of CSC, able to self-renew and differentiate, influences central aspects of tumor biology, in part because they can continue tumor growth, give rise to metastasis, and acquire drug and radioresistance, which open new avenues for therapeutics. It is well known that SC constantly interacts with their niche, which includes mesenchymal cells, extracellular ligands, and the Extra Cellular Matrix (ECM). These interactions regularly lead to homeostasis and maintenance of SC characteristics. However, the exact participation of each of these components for CSC maintenance is not clear, as they appear to be context- or cell-specific. In the recent past, surface cellular markers have been fundamental molecular tools for identifying CSC and distinguishing them from other tumor cells. Importantly, some of these cellular markers have been shown to possess functional roles that affect central aspects of CSC. Likewise, some of these markers can participate in regulating the interaction of CSC with their niche, particularly the ECM. We focused this review on the molecular mechanisms of surface cellular markers commonly employed to identify CSC, highlighting the signaling pathways and mechanisms involved in CSC-ECM interactions, through each of the cellular markers commonly used in the study of CSC, such as CD44, CD133, CD49f, CD24, CXCR4, and LGR5. Their presence does not necessarily implicate them in CSC biology.

Keywords: Cancer, Extra Cellular Matrix (ECM), phenotype, self-renewal, signaling pathways, stemness, surface markers.

[1]
Gupta PB, Chaffer CL, Weinberg RA. Cancer stem cells: Mirage or reality? Nat Med 2009; 15(9): 1010-2.
[2]
Batlle E, Clevers H. Cancer stem cells revisited. Nat Med 2017; 23(10): 1124-34.
[3]
Chen S, Lewallen M, Xie T. Adhesion in the stem cell niche: biological roles and regulation. Development 2013; 140(2): 255-65.
[4]
Zhu TS, Costello MA, Talsma CE, et al. Endothelial cells create a stem cell niche in glioblastoma by providing NOTCH ligands that nurture self-renewal of cancer stem-like cells. Cancer Res 2011; 71(18): 6061-72.
[5]
Mani SA, Guo W, Liao M-J, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008; 133(4): 704-15.
[6]
Bourguignon LYW, Spevak CC, Wong G, Xia W, Gilad E. Hyaluronan-CD44 Interaction with Protein Kinase Cϵ Promotes Oncogenic Signaling by the Stem Cell Marker Nanog and the Production of MicroRNA-21, Leading to Down-regulation of the Tumor Suppressor Protein PDCD4, Anti-apoptosis, and Chemotherapy Resistance in Breast Tumor Cells. J Biol Chem 2009; 284(39): 26533-46.
[7]
Pietras A, Katz AM, Ekström EJ, et al. Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth. Cell Stem Cell 2014; 14(3): 357-69.
[8]
Prochazka L, Tesarik R, Turanek J. Regulation of alternative splicing of CD44 in cancer. Cell Signal 2014; 26(10): 2234-9.
[9]
Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 2003; 100(7): 3983-8.
[10]
Ju S-Y, Chiou S-H, Su Y. Maintenance of the stemness in CD44+ HCT-15 and HCT-116 human colon cancer cells requires miR-203 suppression. Stem Cell Res 2014; 12(1): 86-100.
[11]
Marangoni E, Lecomte N, Durand L, et al. CD44 targeting reduces tumour growth and prevents post-chemotherapy relapse of human breast cancers xenografts. Br J Cancer 2009; 100(6): 918-22.
[12]
Naor D, Sionov RV, Ish-Shalom D. CD44: Structure, function and association with the malignant process. Adv Cancer Res 1997; 71: 241-319.
[13]
Racine RR, Manalo NA, Hall JMF, Dibas A, Raffel GD, Mummert ME. CD44 induced enhancement of phosphatase activity and calcium influx: Modifications of EGR-1 expression and cell proliferation. Biochem Biophys Rep 2016; 6: 172-8.
[14]
Bai Y, Liu YJ, Wang H, Xu Y, Stamenkovic I, Yu Q. Inhibition of the hyaluronan-CD44 interaction by merlin contributes to the tumor-suppressor activity of merlin. Oncogene 2007; 26(6): 836-50.
[15]
Morrison H, Sperka T, Manent J, Giovannini M, Ponta H, Herrlich P. Merlin/Neurofibromatosis Type 2 suppresses growth by inhibiting the activation of Ras and Rac. Cancer Res 2007; 67(2): 520-7.
[16]
Morrison H, Sherman LS, Legg J, et al. The NF2 tumor suppressor gene product, merlin, mediates contact inhibition of growth through interactions with CD44. Genes Dev 2001; 15(8): 968-80.
[17]
Orian-Rousseau V, Morrison H, Matzke A, et al. Hepatocyte growth factor-induced Ras activation requires ERM proteins linked to both CD44v6 and F-actin. Mol Biol Cell 2007; 18(1): 76-83.
[18]
Inaba M, Sorenson DR, Kortus M, Salzmann V, Yamashita YM. Merlin is required for coordinating proliferation of two stem cell lineages in the Drosophila testis. Sci Rep 2017; 7(1): 1-12.
[19]
Larsson J, Ohishi M, Garrison B, et al. Nf2/Merlin regulates hematopoietic stem cell behavior by altering microenvironmental architecture. Cell Stem Cell 2008; 3(2): 221-7.
[20]
Schmitt M, Metzger M, Gradl D, Davidson G, Orian-Rousseau V. CD44 functions in Wnt signaling by regulating LRP6 localization and activation. Cell Death Differ 2015; 22(4): 677-89.
[21]
Miller JR, Hocking AM, Brown JD, Moon RT. Mechanism and function of signal transduction by the Wnt/β-catenin and Wnt/Ca2+ pathways. Oncogene 2000; 18(55): 7860-72.
[22]
Cheng C, Sharp PA. Regulation of CD44 Alternative splicing by srm160 and its potential role in tumor cell invasion. Mol Cell Biol 2006; 26(1): 362-70.
[23]
Brown RL, Reinke LM, Damerow MS, et al. CD44 splice isoform switching in human and mouse epithelium is essential for epithelial-mesenchymal transition and breast cancer progression. J Clin Invest 2011; 121(3): 1064-74.
[24]
Zeilstra J, Joosten SPJ, van Andel H, et al. Stem cell CD44v isoforms promote intestinal cancer formation in Apc(min) mice downstream of Wnt signaling. Oncogene 2014; 33(5): 665-70.
[25]
Harn HJ, Shen KL, Liu CA, Ho LI, Yang LS, Yueh KC. Hyaluronate binding assay study of transfected CD44 V4-V7 isoforms into the human gastric carcinoma cell line SC-M1. J Pathol 1998; 184(3): 291-6.
[26]
Wallach-Dayan SB, Grabovsky V, Moll J, et al. CD44-dependent lymphoma cell dissemination: a cell surface CD44 variant, rather than standard CD44, supports in vitro lymphoma cell rolling on hyaluronic acid substrate and its in vivo accumulation in the peripheral lymph nodes. J Cell Sci 2001; 114(19): 3463-77.
[27]
Nagano O, Okazaki S, Saya H. Redox regulation in stem-like cancer cells by CD44 variant isoforms. Oncogene 2013; 32(44): 5191-8.
[28]
Ishimoto T, Nagano O, Yae T, et al. CD44 Variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc− and thereby promotes tumor growth. Cancer Cell 2011; 19(3): 387-400.
[29]
Todaro M, Gaggianesi M, Catalano V, et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell 2014; 14(3): 342-56.
[30]
Fargeas CA, Florek M, Huttner WB, Corbeil D. Characterization of prominin-2, a new member of the prominin family of pentaspan membrane glycoproteins. J Biol Chem 2003; 278(10): 8586-96.
[31]
Liu Y, Ren S, Xie L, et al. Mutation of N-linked glycosylation at Asn548 in CD133 decreases its ability to promote hepatoma cell growth. Oncotarget 2015; 6(24): 20650.
[32]
Shmelkov SV, St.Clair R, Lyden D, Rafii S. AC133/ CD133/Prominin-1. Int J Biochem Cell Biol 2005; 37(4): 715-9.
[33]
Mehra N. Progenitor Marker CD133 mRNA Is elevated in peripheral blood of cancer patients with bone metastases. Clin Cancer Res 2006; 12(16): 4859-66.
[34]
Wu Y, Wu PY. CD133 as a marker for cancer stem cells: progresses and concerns. Stem Cells Dev 2009; 18(8): 1127-34.
[35]
Huang J, Li C, Wang Y, et al. Cytokine-induced killer (CIK) cells bound with anti-CD3/anti-CD133 bispecific antibodies target CD133high cancer stem cells in vitro and in vivo. Clin Immunol 2013; 149(1): 156-68.
[36]
Zhao L, Yang Y, Zhou P, et al. Targeting CD133high colorectal cancer cells in vitro and in vivo with an asymmetric bispecific antibody. J Immunother 2015; 38(6): 217-28.
[37]
Olweus J, Kearney J, Buck DW, et al. AC133, a novel marker for human hematopoietic stem and progenitor. Blood 1997; 90(12): 5002-12.
[38]
Peichev M, Naiyer AJ, Pereira D, et al. Expression of VEGFR-2 and AC133 by circulating human CD34+ cells identifies a population of functional endothelial precursors. Blood 2000; 95(3): 952-8.
[39]
Uchida N, Buck DW, He D, et al. Direct isolation of human central nervous system stem cells. Proc Natl Acad Sci 2000; 97(26): 14720-5.
[40]
Singh SK, Clarke ID, Terasaki M, et al. Identification of a cancer stem cell in human brain tumors. Cancer Res 2003; 63(18): 5821-8.
[41]
Ma L, Liu T, Jin Y, Wei J, Yang Y, Zhang H. ABCG2 is required for self-renewal and chemoresistance of CD133-positive human colorectal cancer cells. Tumour Biol 2016; 37(9): 12889-96.
[42]
Borrego-Diaz E, Terai K, Lialyte K, et al. Overactivation of Ras signaling pathway in CD133+ MPNST cells. J Neurooncol 2012; 108(3): 423-34.
[43]
Wang YK, Zhu YL, Qiu FM, et al. Activation of Akt and MAPK pathways enhances the tumorigenicity of CD133+ primary colon cancer cells. Carcinogenesis 2010; 31(8): 1376-80.
[44]
Tang KH, Ma S, Lee TK, et al. CD133+ liver tumor-initiating cells promote tumor angiogenesis, growth, and self-renewal through neurotensin/interleukin-8/CXCL1 signaling. Hepatology 2012; 55(3): 807-20.
[45]
Ginestier C, Liu S, Diebel ME, et al. CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts. J Clin Invest 2010; 120(2): 485-97.
[46]
Korkaya H, Liu S, Wicha MS. Regulation of cancer stem cells by cytokine networks: Attacking cancer’s inflammatory Roots. Clin Cancer Res 2011; 17(19): 6125-9.
[47]
Charafe-Jauffret E, Ginestier C, Iovino F, et al. Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. Cancer Res 2009; 69(4): 1302-13.
[48]
Mak AB, Nixon AML, Kittanakom S, et al. Regulation of CD133 by HDAC6 promotes β-catenin signaling to suppress cancer cell differentiation. Cell Rep 2012; 2(4): 951-63.
[49]
Li Y, Zhang X, Polakiewicz RD, Yao T-P, Comb MJ. HDAC6 is required for epidermal growth factor-induced β-catenin nuclear localization. J Biol Chem 2008; 283(19): 12686-90.
[50]
Suzuki M. Hypoxia induces CD133 expression in human lung cancer cells by up-regulation of OCT3/4 and SOX2. Int J Oncol 2011; 40(1): 71-9.
[51]
Irollo E, Pirozzi G. CD133: to be or not to be, is this the real question? Am J Transl Res 2013; 5(6): 563-81.
[52]
Neuzil J, Stantic M, Zobalova R, et al. Tumour-initiating cells vs. cancer ‘stem’ cells and CD133: What’s in the name? Biochem Biophys Res Commun 2007; 355(4): 855-9.
[53]
Kemper K, Sprick MR, de Bree M, et al. The AC133 Epitope, but not the CD133 protein, is lost upon cancer stem cell differentiation. Cancer Res 2010; 70(2): 719-29.
[54]
Fargeas CA, Huttner WB, Corbeil D. Nomenclature of prominin-1 (CD133) splice variants? an update. Tissue Antigens 2007; 69(6): 602-6.
[55]
Tabu K, Bizen N, Taga T, Tanaka S. Gene Regulation of Prominin-1 (CD133) in Normal and Cancerous Tissues. In: Corbeil D, editor. Prominin-1 (CD133): New Insights on Stem & Cancer Stem Cell Biology. New York: NY Springer New York 2013; pp. 73-85.
[56]
Wei Y, Jiang Y, Zou F, et al. Activation of PI3K/Akt pathway by CD133-p85 interaction promotes tumorigenic capacity of glioma stem cells. Proc Natl Acad Sci 2013; 110(17): 6829-34.
[57]
Watt FM. Role of integrins in regulating epidermal adhesion, growth and differentiation. EMBO J 2002; 21(15): 3919-26.
[58]
Friedrichs K, Ruiz P, Franke F, Gille I, Terpe HJ, Imhof BA. High expression level of alpha 6 integrin in human breast carcinoma is correlated with reduced survival. Cancer Res 1995; 55(4): 901-6.
[59]
Lathia JD, Gallagher J, Heddleston JM, et al. Integrin alpha 6 regulates glioblastoma stem Cells. Cell Stem Cell 2010; 6(5): 421-32.
[60]
Hou T, Zhang W, Tong C, et al. Putative stem cell markers in cervical squamous cell carcinoma are correlated with poor clinical outcome. BMC Cancer 2015; 15(1): 1-12.
[61]
Lim S-T, Chen XL, Lim Y, et al. Nuclear FAK Promotes Cell Proliferation and Survival through FERM-Enhanced p53 Degradation. Mol Cell 2008; 29(1): 9-22.
[62]
Mitra SK, Mikolon D, Molina JE, et al. Intrinsic FAK activity and Y925 phosphorylation facilitate an angiogenic switch in tumors. Oncogene 2006; 25(44): 5969-84.
[63]
Yu K-R, Yang S-R, Jung J-W, et al. CD49f Enhances Multipotency and Maintains Stemness Through the Direct Regulation of OCT4 and SOX2. Stem Cells 2012; 30(5): 876-87.
[64]
Guan J-L. Integrin signaling through FAK in the regulation of mammary stem cells and breast cancer. IUBMB Life 2010; 62(4): 268-76.
[65]
Villa-Diaz LG, Kim JK, Laperle A, Palecek SP, Krebsbach PH. Inhibition of focal adhesion kinase signaling by integrin α6β1 supports human pluripotent stem cell self-renewal: Integrin-FAK Involvement in Self-Renewal of hPSCs. Stem Cells 2016; 34(7): 1753-64.
[66]
Begum A, Ewachiw T, Jung C, et al. The extracellular matrix and focal adhesion kinase signaling regulate cancer stem cell function in pancreatic ductal adenocarcinoma. PLoS One 2017; 12(7)e0180181
[67]
Kolev VN, Tam WF, Wright QG, et al. Inhibition of FAK kinase activity preferentially targets cancer stem cells. Oncotarget 2017; 8(31): 51733.
[68]
Chang C, Goel HL, Gao H, et al. A laminin 511 matrix is regulated by TAZ and functions as the ligand for the α6Bβ1 integrin to sustain breast cancer stem cells. Genes Dev 2015; 29(1): 1-6.
[69]
Goel HL, Gritsko T, Pursell B, et al. Regulated splicing of the α6 integrin cytoplasmic domain determines the fate of breast cancer stem cells. Cell Rep 2014; 7(3): 747-61.
[70]
Hu T, Zhou R, Zhao Y, Wu G. Integrin α6/Akt/Erk signaling is essential for human breast cancer resistance to radiotherapy. Sci Rep 2016; 6(1): 1-10.
[71]
López J, Poitevin A, Mendoza-Martínez V, Pérez-Plasencia C, García-Carrancá A. Cancer-initiating cells derived from established cervical cell lines exhibit stem-cell markers and increased radioresistance. BMC Cancer 2012; 12(48): 1-14.
[72]
Culp TD, Budgeon LR, Marinkovich MP, Meneguzzi G, Christensen ND. Keratinocyte-secreted laminin 5 can function as a transient receptor for human papillomaviruses by binding virions and transferring them to adjacent cells. J Virol 2006; 80(18): 8940-50.
[73]
Yoon C-S, Kim K-D, Park S-N, Cheong S-W. α6 Integrin Is the Main Receptor of Human Papillomavirus Type 16 VLP. Biochem Biophys Res Commun 2001; 283(3): 668-73.
[74]
Ortiz-Sánchez E, Santiago-López L, Cruz-Domínguez VB, et al. Characterization of cervical cancer stem cell-like cells: Phenotyping, stemness, and human papilloma virus co-receptor expression. Oncotarget 2016; 7(22): 31943-54.
[75]
Bretz NP, Salnikov AV, Perne C, et al. CD24 controls Src/STAT3 activity in human tumors. Cell Mol Life Sci 2012; 69(22): 3863-79.
[76]
Ono YJ, Tanabe A, Tanaka T, et al. Met signaling cascade is amplified by the recruitment of phosphorylated met to lipid rafts via CD24 and leads to drug resistance in endometrial cancer cell lines. Mol Cancer Ther 2015; 14(10): 2353-63.
[77]
Runz S, Mierke CT, Joumaa S, Behrens J, Fabry B, Altevogt P. CD24 induces localization of β1 integrin to lipid raft domains. Biochem Biophys Res Commun 2008; 365(1): 35-41.
[78]
Ayre DC, Pallegar NK, Fairbridge NA, Canuti M, Lang AS, Christian SL. Analysis of the structure, evolution, and expression of CD24, an important regulator of cell fate. Gene 2016; 590(2): 324-37.
[79]
Zheng J, Li Y, Yang J, et al. NDRG2 inhibits hepatocellular carcinoma adhesion, migration and invasion by regulating CD24 expression. BMC Cancer 2011; 11(1): 1-9.
[80]
Wang X, Zhang Y, Zhao Y, et al. CD24 promoted cancer cell angiogenesis via Hsp90-mediated STAT3/VEGF signaling pathway in colorectal cancer. Oncotarget 2016; 7(34): 55663-76.
[81]
Shah M, Patel K, Fried VA, Sehgal PB. Interactions of STAT3 with Caveolin-1 and Heat Shock Protein 90 in Plasma Membrane Raft and Cytosolic Complexes: Preservation of cytokine signaling during fever. J Biol Chem 2002; 277(47): 45662-9.
[82]
Baumann P, Cremers N, Kroese F, et al. CD24 expression causes the acquisition of multiple cellular properties associated with tumor growth and metastasis. Cancer Res 2005; 65(23): 10783-93.
[83]
Ricardo S, Vieira AF, Gerhard R, et al. Breast cancer stem cell markers CD44, CD24 and ALDH1: expression distribution within intrinsic molecular subtype. J Clin Pathol 2011; 64(11): 937-46.
[84]
Honeth G, Bendahl P-O, Ringnér M, et al. The CD44+/CD24-phenotype is enriched in basal-like breast tumors. Breast Cancer Res 2008; 10(3): 1-12.
[85]
Mylona E, Giannopoulou I, Fasomytakis E, et al. The clinicopathologic and prognostic significance of CD44+/CD24−/low and CD44−/CD24+ tumor cells in invasive breast carcinomas. Hum Pathol 2008; 39(7): 1096-102.
[86]
Cho RW, Wang X, Diehn M, et al. Isolation and Molecular Characterization of Cancer Stem Cells in MMTV- Wnt-1 Murine Breast Tumors. Stem Cells 2008; 26(2): 364-71.
[87]
Vaillant F, Asselin-Labat M-L, Shackleton M, Forrest NC, Lindeman GJ, Visvader JE. The mammary progenitor marker CD61/ 3 integrin identifies cancer stem cells in mouse models of mammary tumorigenesis. Cancer Res 2008; 68(19): 7711-7.
[88]
Zhang C, Li C, He F, Cai Y, Yang H. Identification of CD44+CD24+ gastric cancer stem cells. J Cancer Res Clin Oncol 2011; 137(11): 1679-86.
[89]
Stuelten CH, Mertins SD, Busch JI, et al. Complex Display of Putative Tumor Stem Cell Markers in the NCI60 Tumor Cell Line Panel. Stem Cells 2010; 28(4): 649-60.
[90]
Overdevest JB, Thomas S, Kristiansen G, Hansel DE, Smith SC, Theodorescu D. CD24 offers a therapeutic target for control of bladder cancer metastasis based on a requirement for lung colonization. Cancer Res 2011; 71(11): 3802-11.
[91]
Yang C-H, Wang H-L, Lin Y-S, et al. Identification of CD24 as a cancer stem cell marker in human nasopharyngeal carcinoma. Seno M. PLoS One 2014; 9(6)e99412
[92]
Yeung TM, Gandhi SC, Wilding JL, Muschel R, Bodmer WF. Cancer stem cells from colorectal cancer-derived cell lines. Proc Natl Acad Sci 2010; 107(8): 3722-7.
[93]
Li C, Lee CJ, Simeone DM. Identification of Human Pancreatic Cancer Stem Cells. In: Yu JS, editor. Cancer Stem Cells. Totowa, NJ: Humana Press 2009; pp. 161-73.
[94]
Gao M-Q, Choi Y-P, Kang S, Youn J, Cho N-H. CD24+ cells from hierarchically organized ovarian cancer are enriched in cancer stem cells. Oncogene 2010; 29(18): 2672-80.
[95]
Vermeulen L, Todaro M, de Sousa Mello F, et al. Single-cell cloning of colon cancer stem cells reveals a multi-lineage differentiation capacity. Proc Natl Acad Sci 2008; 105(36): 13427-32.
[96]
Burgos-Ojeda D, Wu R, McLean K, et al. CD24+ Ovarian Cancer Cells Are Enriched for Cancer-Initiating Cells and Dependent on JAK2 Signaling for Growth and Metastasis. Mol Cancer Ther 2015; 14(7): 1717-27.
[97]
Lee TKW, Castilho A, Cheung VCH, Tang KH, Ma S, Ng IOL. CD24+ Liver Tumor-Initiating Cells Drive Self-Renewal and Tumor Initiation through STAT3-Mediated NANOG Regulation. Cell Stem Cell 2011; 9(1): 50-63.
[98]
Shen Y-A, Wang C-Y, Chuang H-Y, et al. CD44 and CD24 coordinate the reprogramming of nasopharyngeal carcinoma cells towards a cancer stem cell phenotype through STAT3 activation. Oncotarget 2016; 7(36): 58351-66.
[99]
Lim J, Lee K, Shim J, Shin I. CD24 regulates stemness and the epithelial to mesenchymal transition through modulation of Notch1 mRNA stability by p38MAPK. Arch Biochem Biophys 2014; 558: 120-6.
[100]
Nakamura K, Terai Y, Tanabe A, et al. CD24 expression is a marker for predicting clinical outcome and regulates the epithelial-mesenchymal transition in ovarian cancer via both the Akt and ERK pathways. Oncol Rep 2017 Jun; 37(6): 3189-200.
[101]
Lo H-W, Zhu H, Cao X, Aldrich A, Ali-Osman F. A Novel Splice Variant of GLI1 That Promotes Glioblastoma Cell Migration and Invasion. Cancer Res 2009; 69(17): 6790-8.
[102]
Schabath H. CD24 affects CXCR4 function in pre-B lymphocytes and breast carcinoma cells. J Cell Sci 2006; 119(2): 314-25.
[103]
Ju J, Jang K, Lee K, et al. CD24 enhances DNA damage-induced apoptosis by modulating NF-κB signaling in CD44-expressing breast cancer cells. Carcinogenesis 2011; 32(10): 1474-83.
[104]
Kristiansen G, Sammar M, Altevogt P. Tumour Biological Aspects of CD24, A Mucin-Like Adhesion Molecule. J Mol Histol 2003; 35(3): 255-62.
[105]
Teicher BA, Fricker SP. CXCL12 (SDF-1)/CXCR4 Pathway in Cancer. Clin Cancer Res 2010; 16(11): 2927-31.
[106]
Burger JA. CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood 2006; 107(5): 1761-7.
[107]
Barbero S, Bonavia R, Bajetto A, et al. Stromal cell-derived factor 1α stimulates human glioblastoma cell growth through the activation of both extracellular signal-regulated kinases 1/2 and Akt. Cancer Res 2003; 63(8): 1969-74.
[108]
Zhou Y, Larsen PH, Hao C, Yong VW. CXCR4 Is a Major Chemokine Receptor on Glioma Cells and Mediates Their Survival. J Biol Chem 2002; 277(51): 49481-7.
[109]
Kijima T, Maulik G, Ma PC, et al. Regulation of cellular proliferation, cytoskeletal function, and signal transduction through CXCR4 and c-Kit in small cell lung cancer cells. Cancer Res 2002; 62(21): 6304-11.
[110]
Dubrovska A, Hartung A, Bouchez LC, et al. CXCR4 activation maintains a stem cell population in tamoxifen-resistant breast cancer cells through AhR signalling. Br J Cancer 2012; 107(1): 43-52.
[111]
Dubrovska A, Elliott J, Salamone RJ, et al. CXCR4 Expression in Prostate Cancer Progenitor Cells. Bielenberg DR. PLoS One 2012; 7(2)e31226
[112]
Engl T, Relja B, Marian D, et al. CXCR4 Chemokine Receptor Mediates Prostate Tumor Cell Adhesion through α5 and β3 Integrins. Neoplasia 2006; 8(4): 290-301.
[113]
Furusato B, Mohamed A, Uhlén M, Rhim JS. CXCR4 and cancer: CXCR4 and cancer. Pathol Int 2010; 60(7): 497-505.
[114]
Gatti M, Pattarozzi A, Bajetto A, et al. Inhibition of CXCL12/CXCR4 autocrine/paracrine loop reduces viability of human glioblastoma stem-like cells affecting self-renewal activity. Toxicology 2013; 314(2-3): 209-20.
[115]
Ueda Y, Neel NF, Schutyser E, Raman D, Richmond A. Deletion of the COOH-Terminal Domain of CXC Chemokine Receptor 4 Leads to the Down-regulation of Cell-to-Cell Contact, Enhanced Motility and Proliferation in Breast Carcinoma Cells. Cancer Res 2006; 66(11): 5665-75.
[116]
Mathieu M-C, Lapierre I, Brault K, Raymond M. Aromatic Hydrocarbon Receptor (AhR)·AhR Nuclear Translocator- and p53-mediated Induction of the Murine Multidrug Resistance mdr1 Gene by 3-Methylcholanthrene and Benzo(a)pyrene in Hepatoma Cells. J Biol Chem 2001; 276(7): 4819-27.
[117]
Jung M-J, Rho J-K, Kim Y-M, et al. Upregulation of CXCR4 is functionally crucial for maintenance of stemness in drug-resistant non-small cell lung cancer cells. Oncogene 2013; 32(2): 209-21.
[118]
Fuchs K, Hippe A, Schmaus A, Homey B, Sleeman JP, Orian-Rousseau V. Opposing effects of high- and low-molecular weight hyaluronan on CXCL12-induced CXCR4 signaling depend on CD44. Cell Death Dis 2013; 4(10): 1-11.
[119]
Carmon KS, Gong X, Lin Q, Thomas A, Liu Q. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/ -catenin signaling. Proc Natl Acad Sci 2011; 108(28): 11452-7.
[120]
Sato T, van Es JH, Snippert HJ, et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 2011; 469(7330): 415-8.
[121]
Barker N, van Es JH, Kuipers J, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007; 449(7165): 1003-7.
[122]
Shimokawa M, Ohta Y, Nishikori S, et al. Visualization and targeting of LGR5+ human colon cancer stem cells. Nature 2017; 545(7653): 187-92.
[123]
de Sousa e Melo F, Kurtova AV, Harnoss JM, et al.A distinct role for Lgr5+ stem cells in primary and metastatic colon cancer. Nature 2017; 543(7647): 676-80.
[124]
Chen Q, Cao H-Z, Zheng P-S. LGR5 promotes the proliferation and tumor formation of cervical cancer cells through the Wnt/β-catenin signaling pathway. Oncotarget 2014; 5(19): 9092-105.
[125]
Seino T, Kawasaki S, Shimokawa M, et al. Human pancreatic tumor organoids reveal loss of stem cell niche factor dependence during disease progression. Cell Stem Cell 2018; 22(3): 454-67.
[126]
Carmon KS, Gong X, Yi J, et al. LGR5 receptor promotes cell-cell adhesion in stem cells and colon cancer cells via the IQGAP1-Rac1 pathway. J Biol Chem 2017; 292(36): 14989-5001.
[127]
Yang L, Tang H, Kong Y, et al. LGR5 Promotes Breast Cancer Progression and Maintains Stem-Like Cells Through Activation of Wnt/β-Catenin signaling: LGR5 activation in breast cancer stem cells. Stem Cells 2015; 33(10): 2913-24.
[128]
Carmon KS, Gong X, Yi J, Thomas A, Liu Q. RSPO-LGR4 functions via IQGAP1 to potentiate Wnt signaling. Proc Natl Acad Sci 2014; 111(13): E1221-9.
[129]
Carmon KS, Lin Q, Gong X, Thomas A, Liu Q. LGR5 Interacts and Cointernalizes with Wnt Receptors To Modulate Wnt/ -Catenin Signaling. Mol Cell Biol 2012; 32(11): 2054-64.
[130]
Snyder JC, Rochelle LK, Barak LS, Caron MG. The Stem Cell-Expressed Receptor Lgr5 Possesses Canonical and Functionally Active Molecular Determinants Critical to β-arrestin-2 Recruitment. Porter J PLoS ONE 2013; 8(12)e84476
[131]
Kim K-A, Kakitani M, Zhao J, et al. Mitogenic Influence of Human R-Spondin1 on the Intestinal Epithelium. Science 2005; 309(5738): 1256-9.
[132]
Cao H-Z, Liu X-F, Yang W-T, Chen Q, Zheng P-S. LGR5 promotes cancer stem cell traits and chemoresistance in cervical cancer. Cell Death Dis 2017; 8(9)e3039
[133]
Baker A-M, Graham TA, Elia G, Wright NA, Rodriguez-Justo M. Characterization of LGR5 stem cells in colorectal adenomas and carcinomas. Sci Rep 2015; 5(1): 1-8.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy