Ovarian cancer spheroid cells with stem cell-like properties contribute to tumor generation, metastasis and chemotherapy resistance through hypoxia-resistant metabolism

Jianqun Liao, Feng Qian, Nana Tchabo, Paulette Mhawech-Fauceglia, Amy Beck, Zikun Qian, Xinhui Wang, Wendy J Huss, Shashikant B Lele, Carl D Morrison, Kunle Odunsi, Jianqun Liao, Feng Qian, Nana Tchabo, Paulette Mhawech-Fauceglia, Amy Beck, Zikun Qian, Xinhui Wang, Wendy J Huss, Shashikant B Lele, Carl D Morrison, Kunle Odunsi

Abstract

Cells with sphere forming capacity, spheroid cells, are present in the malignant ascites of patients with epithelial ovarian cancer (EOC) and represent a significant impediment to efficacious treatment due to their putative role in progression, metastasis and chemotherapy resistance. The exact mechanisms that underlie EOC metastasis and drug resistance are not clear. Understanding the biology of sphere forming cells may contribute to the identification of novel therapeutic opportunities for metastatic EOC. Here we generated spheroid cells from human ovarian cancer cell lines and primary ovarian cancer. Xenoengraftment of as few as 2000 dissociated spheroid cells into immune-deficient mice allowed full recapitulation of the original tumor, whereas >10(5) parent tumor cells remained non-tumorigenic. The spheroid cells were found to be enriched for cells with cancer stem cell-like characteristics such as upregulation of stem cell genes, self-renewal, high proliferative and differentiation potential, and high aldehyde dehydrogenase (ALDH) activity. Furthermore, spheroid cells were more aggressive in growth, migration, invasion, scratch recovery, clonogenic survival, anchorage-independent growth, and more resistant to chemotherapy in vitro. (13)C-glucose metabolic studies revealed that spheroid cells route glucose predominantly to anaerobic glycolysis and pentose cycle to the detriment of re-routing glucose for anabolic purposes. These metabolic properties of sphere forming cells appear to confer increased resistance to apoptosis and contribute to more aggressive tumor growth. Collectively, we demonstrated that spheroid cells with cancer stem cell-like characteristics contributed to tumor generation, progression and chemotherapy resistance. This study provides insight into the relationship between tumor dissemination and metabolic attributes of human cancer stem cells and has clinical implications for cancer therapy.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Spheroid cells were generated from…
Figure 1. Spheroid cells were generated from primary and established EOC cells.
(A) Three primary cell lines generated from EOC patients show epithelial cells morphology. (B) One of primary EOC cell line generated 3-D independent, self-renewing spheroid cells under stem-cell selection media. (C) Primary cell lines express ovarian carcinoma marker CA-125 and epithelial marker CK-7, as shown by IHC for RP-OV17534. (D) EOC cell line OV2774 is able to generate OV2774 spheroid cells (E). All scale bar unit in this figure and following figures are in µm.
Figure 2. RP-OV17534 Spheroid cells generated tumors…
Figure 2. RP-OV17534 Spheroid cells generated tumors in immune-deficient mice.
(A) Different amount of spheroid cells s.c. injected into SCID mice formed tumors. (B) Spheroid cells i.p. injected into SCID mice formed bloody ascites and tumors in different organs as indicated by arrows. (C) Representative H&E staining sections (upper panel) shows RP-OV17534 primary tumor, subcutaneous graft tumor from spheroids, intraperitoneal graft tumor from spheroids, and serial intraperitoneal graft tumor from SCID mouse ascites. Histological analysis (lower panel) shows frequent metastasis of tumor cells into various organs of spheroid cells recipients.
Figure 3. Spheroid cells overexpressed stem cell…
Figure 3. Spheroid cells overexpressed stem cell genes.
(A) Total RNA isolated from spheroid cells and parent cells were examined for their gene expression by a human stem cell markers cDNA array and stem cell-genes showed higher expression levels in spheroid cells to non-spheroid cells. (B) The overexpression levels of Notch1, Nanog, Cdcp1, CD34, and Myc in spheroid cells compared to non-spheroid cells were confirmed by quantitative real-time PCR. These stem cell-genes’ expression were lost or down-regulated when spheroid cells were differentiated by culturing in CM without growth factors for 14 days. Expression levels are represented as fold changes compared to these of non-spheroid cells. (C) FACS examination of CD117 expression on spheroid cells and parent cells showing high CD117 expression in spheroid cells. Some spheroid cells were treated with Cisplatin for 24 hrs before surface staining by CD117 Abs. Error bars: SD, N = 3. *: p

Figure 4. Increased ALDH activity in spheroid…

Figure 4. Increased ALDH activity in spheroid cells.

(A) The ALDEFLOUR kit labels the population…

Figure 4. Increased ALDH activity in spheroid cells.
(A) The ALDEFLOUR kit labels the population with a high ALDH enzymatic activity in spheroid cells. An aliquot of each sample of cells was treated with ALDH inhibitor DEAB as negative control for setting FACS gate. (B) The BioVision ALDH colorimetric assay detected enhanced ALDH activity in spheroid cell lysate. Error bars: SD, N = 2. *: p

Figure 5. Spheroid cells show a more…

Figure 5. Spheroid cells show a more aggressive growth pattern.

(A) Spheroid cells had higher…

Figure 5. Spheroid cells show a more aggressive growth pattern.
(A) Spheroid cells had higher proliferation potential than their parent cells. Both spheroid cells and parental cells were cultured in CM for 24 h, then 5000/well cells were cultured in CM in 96 well plates. Half of media was changed every 3 days. Cell proliferations were detected from day 0 to day 17 by CellTiter-Glo Luminescent Cell Viability Assay. (B) Spheroid cells had higher migration ability than parent cells. Parental and spheroid EOC cells were treated with RPMI only medium for 24 hrs. In a transwell plate, 2×105/well cells were put into insert with 0.2%BSA-RPMI medium, the lower chamber contains no cells except RPMI medium with 2.5% FBS. After culture for 1, 2, and 3 days, the numbers of cells that passed through membrane as well as cells fall into chamber were calculated under microscope. (C) Spheroid cells recovered ‘scratch’ made by 200-µL pipette tip after 24 hours more efficient than their parent cells. Experiments were repeated three times with similar results. (D) More spheroid cells invaded into matrigel than parent cells as shown by microscopy. Spheroid cells and parental cells were treated with serum-free medium RPMI for 24 h. In a transwell plate, 40000/well of cells were put into insert coated with matrigel with 0.2% BSA, while the chamber had only media with 5% FBS, after culture for 1, 2, and 3 days, The number of cells invading into matrigel were counted after 0.1% crystal blue staining. (E) Invasion tests were done in duplicate and average and SD were shown. *,p<0.01. (F) Spheroid cells formed colonies in top agar detected in anchorage-independent growth assay (AIG) as shown under microscopy. 500 cells were seeded in 6 well plates containing a top layer of 0.3% soft agar and 0.5% agar base in DMEM, 10% FBS. 24 hrs later, the number of cells seeded was determined by counting cells under the light microscope. After 3 weeks of growth in soft agar, the number of colonies (Colonies >5 cells) were counted; 10 different fields were quantified per well and the average number of colonies per field was calculated. The AIG index was expressed relative to the number of cells seeded. More than 70% of seeded spheroid cells formed colonies while none of seeded parent cells formed colonies. Experiments were repeated twice with similar results. *,p<0.01. Error bars: SD.

Figure 6. Spheroid cells are resistant to…

Figure 6. Spheroid cells are resistant to chemotherapy in vitro .

(A) More spheroid cells were…

Figure 6. Spheroid cells are resistant to chemotherapy in vitro.
(A) More spheroid cells were killed by Cisplatin than parent cells. The same numbers of spheroid and parent cells were cultured in CM with indicated amount of Cisplatin for 2 days and numbers of cells from different time points were determined. (B) More parent cells survived after Cisplatin treatment for 48 h (Left). However, 8 days later, only spheroid cells were growing while parent cells disappeared (Right). (C) Spheroid and parent cells were treated with indicated amount of Cisplatin for 24 h, washed, and 300 of survived cells/well were cultured in CM for another 10 days, cell colonies were shown after crystal violet staining. The same clonogenic survival assays were repeated twice with similar results. Colonies formed (>5 cells) after 10 days of growth were counted and average and SD were shown. *,p

Figure 7. Spheroid cells show a hypoxia-resistant…

Figure 7. Spheroid cells show a hypoxia-resistant metabolism profile.

Metabolic profiles of a normal ovarian…

Figure 7. Spheroid cells show a hypoxia-resistant metabolism profile.
Metabolic profiles of a normal ovarian cell line RPNLOv78 as control, EOC cells RP-OV17534, and spheroid RP-OV17534 cells cultures in the presence of [U-13C6]glucose for 24 hrs (n = 3) showing (A) percentages of m3-13C lactate derived directed from glycolysis; (B) percentages of m2-13C lactate derived from pentose cycle; (C) ratio of m2 to m3 lactate; (D) percentage of 13C-labeled lactate of total lactate; (E) percentages of 13C-double-labeled glutamate in total 13C-labeled glutamate; (F) 13CO2 release from [U-13C6]glucose; (G) The fraction of newly synthesized palmitate and the 13C enrichment of acetyl units (H). Error bars: SD; *,p<0.01, compared with parental EOC cells. (I) EOC spheroid cells show increased anaerobic glycolysis, increased direct glucose oxidation in the pentose cycle, but low TCA cycle carbon flux, low anaplerotic flux, and de novo fatty acid synthesis. In contrast, parent EOC cells show increased TCA cycle and high anaplerotic flux. G6P, glucose-6-P; F6P, fructose-6-P; GAP, glyceraldehyde-3-P; 6PG, 6-P-gluconate; R5P, ribulose-5-P. Various thickness of lines represent different strengths of metabolic flux.
All figures (7)
Similar articles
Cited by
References
    1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, et al. (2009) Cancer statistics, 2009. CA Cancer J Clin 59: 225–249. - PubMed
    1. Armstrong D (2010) Update on treatment options for newly diagnosed ovarian cancer. Clin Adv Hematol Oncol 8: 675–678. - PubMed
    1. Colombo N, Van Gorp T, Parma G, Amant F, Gatta G, et al. (2006) Ovarian cancer. Crit Rev Oncol Hematol 60: 159–179. - PubMed
    1. Cannistra SA (2004) Cancer of the ovary. N Engl J Med 351: 2519–2529. - PubMed
    1. Burleson KM, Boente MP, Pambuccian SE, Skubitz AP (2006) Disaggregation and invasion of ovarian carcinoma ascites spheroids. J Transl Med 4: 6. - PMC - PubMed
Show all 59 references
Publication types
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 4. Increased ALDH activity in spheroid…
Figure 4. Increased ALDH activity in spheroid cells.
(A) The ALDEFLOUR kit labels the population with a high ALDH enzymatic activity in spheroid cells. An aliquot of each sample of cells was treated with ALDH inhibitor DEAB as negative control for setting FACS gate. (B) The BioVision ALDH colorimetric assay detected enhanced ALDH activity in spheroid cell lysate. Error bars: SD, N = 2. *: p

Figure 5. Spheroid cells show a more…

Figure 5. Spheroid cells show a more aggressive growth pattern.

(A) Spheroid cells had higher…

Figure 5. Spheroid cells show a more aggressive growth pattern.
(A) Spheroid cells had higher proliferation potential than their parent cells. Both spheroid cells and parental cells were cultured in CM for 24 h, then 5000/well cells were cultured in CM in 96 well plates. Half of media was changed every 3 days. Cell proliferations were detected from day 0 to day 17 by CellTiter-Glo Luminescent Cell Viability Assay. (B) Spheroid cells had higher migration ability than parent cells. Parental and spheroid EOC cells were treated with RPMI only medium for 24 hrs. In a transwell plate, 2×105/well cells were put into insert with 0.2%BSA-RPMI medium, the lower chamber contains no cells except RPMI medium with 2.5% FBS. After culture for 1, 2, and 3 days, the numbers of cells that passed through membrane as well as cells fall into chamber were calculated under microscope. (C) Spheroid cells recovered ‘scratch’ made by 200-µL pipette tip after 24 hours more efficient than their parent cells. Experiments were repeated three times with similar results. (D) More spheroid cells invaded into matrigel than parent cells as shown by microscopy. Spheroid cells and parental cells were treated with serum-free medium RPMI for 24 h. In a transwell plate, 40000/well of cells were put into insert coated with matrigel with 0.2% BSA, while the chamber had only media with 5% FBS, after culture for 1, 2, and 3 days, The number of cells invading into matrigel were counted after 0.1% crystal blue staining. (E) Invasion tests were done in duplicate and average and SD were shown. *,p<0.01. (F) Spheroid cells formed colonies in top agar detected in anchorage-independent growth assay (AIG) as shown under microscopy. 500 cells were seeded in 6 well plates containing a top layer of 0.3% soft agar and 0.5% agar base in DMEM, 10% FBS. 24 hrs later, the number of cells seeded was determined by counting cells under the light microscope. After 3 weeks of growth in soft agar, the number of colonies (Colonies >5 cells) were counted; 10 different fields were quantified per well and the average number of colonies per field was calculated. The AIG index was expressed relative to the number of cells seeded. More than 70% of seeded spheroid cells formed colonies while none of seeded parent cells formed colonies. Experiments were repeated twice with similar results. *,p<0.01. Error bars: SD.

Figure 6. Spheroid cells are resistant to…

Figure 6. Spheroid cells are resistant to chemotherapy in vitro .

(A) More spheroid cells were…

Figure 6. Spheroid cells are resistant to chemotherapy in vitro.
(A) More spheroid cells were killed by Cisplatin than parent cells. The same numbers of spheroid and parent cells were cultured in CM with indicated amount of Cisplatin for 2 days and numbers of cells from different time points were determined. (B) More parent cells survived after Cisplatin treatment for 48 h (Left). However, 8 days later, only spheroid cells were growing while parent cells disappeared (Right). (C) Spheroid and parent cells were treated with indicated amount of Cisplatin for 24 h, washed, and 300 of survived cells/well were cultured in CM for another 10 days, cell colonies were shown after crystal violet staining. The same clonogenic survival assays were repeated twice with similar results. Colonies formed (>5 cells) after 10 days of growth were counted and average and SD were shown. *,p

Figure 7. Spheroid cells show a hypoxia-resistant…

Figure 7. Spheroid cells show a hypoxia-resistant metabolism profile.

Metabolic profiles of a normal ovarian…

Figure 7. Spheroid cells show a hypoxia-resistant metabolism profile.
Metabolic profiles of a normal ovarian cell line RPNLOv78 as control, EOC cells RP-OV17534, and spheroid RP-OV17534 cells cultures in the presence of [U-13C6]glucose for 24 hrs (n = 3) showing (A) percentages of m3-13C lactate derived directed from glycolysis; (B) percentages of m2-13C lactate derived from pentose cycle; (C) ratio of m2 to m3 lactate; (D) percentage of 13C-labeled lactate of total lactate; (E) percentages of 13C-double-labeled glutamate in total 13C-labeled glutamate; (F) 13CO2 release from [U-13C6]glucose; (G) The fraction of newly synthesized palmitate and the 13C enrichment of acetyl units (H). Error bars: SD; *,p<0.01, compared with parental EOC cells. (I) EOC spheroid cells show increased anaerobic glycolysis, increased direct glucose oxidation in the pentose cycle, but low TCA cycle carbon flux, low anaplerotic flux, and de novo fatty acid synthesis. In contrast, parent EOC cells show increased TCA cycle and high anaplerotic flux. G6P, glucose-6-P; F6P, fructose-6-P; GAP, glyceraldehyde-3-P; 6PG, 6-P-gluconate; R5P, ribulose-5-P. Various thickness of lines represent different strengths of metabolic flux.
All figures (7)
Similar articles
Cited by
References
    1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, et al. (2009) Cancer statistics, 2009. CA Cancer J Clin 59: 225–249. - PubMed
    1. Armstrong D (2010) Update on treatment options for newly diagnosed ovarian cancer. Clin Adv Hematol Oncol 8: 675–678. - PubMed
    1. Colombo N, Van Gorp T, Parma G, Amant F, Gatta G, et al. (2006) Ovarian cancer. Crit Rev Oncol Hematol 60: 159–179. - PubMed
    1. Cannistra SA (2004) Cancer of the ovary. N Engl J Med 351: 2519–2529. - PubMed
    1. Burleson KM, Boente MP, Pambuccian SE, Skubitz AP (2006) Disaggregation and invasion of ovarian carcinoma ascites spheroids. J Transl Med 4: 6. - PMC - PubMed
Show all 59 references
Publication types
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 5. Spheroid cells show a more…
Figure 5. Spheroid cells show a more aggressive growth pattern.
(A) Spheroid cells had higher proliferation potential than their parent cells. Both spheroid cells and parental cells were cultured in CM for 24 h, then 5000/well cells were cultured in CM in 96 well plates. Half of media was changed every 3 days. Cell proliferations were detected from day 0 to day 17 by CellTiter-Glo Luminescent Cell Viability Assay. (B) Spheroid cells had higher migration ability than parent cells. Parental and spheroid EOC cells were treated with RPMI only medium for 24 hrs. In a transwell plate, 2×105/well cells were put into insert with 0.2%BSA-RPMI medium, the lower chamber contains no cells except RPMI medium with 2.5% FBS. After culture for 1, 2, and 3 days, the numbers of cells that passed through membrane as well as cells fall into chamber were calculated under microscope. (C) Spheroid cells recovered ‘scratch’ made by 200-µL pipette tip after 24 hours more efficient than their parent cells. Experiments were repeated three times with similar results. (D) More spheroid cells invaded into matrigel than parent cells as shown by microscopy. Spheroid cells and parental cells were treated with serum-free medium RPMI for 24 h. In a transwell plate, 40000/well of cells were put into insert coated with matrigel with 0.2% BSA, while the chamber had only media with 5% FBS, after culture for 1, 2, and 3 days, The number of cells invading into matrigel were counted after 0.1% crystal blue staining. (E) Invasion tests were done in duplicate and average and SD were shown. *,p<0.01. (F) Spheroid cells formed colonies in top agar detected in anchorage-independent growth assay (AIG) as shown under microscopy. 500 cells were seeded in 6 well plates containing a top layer of 0.3% soft agar and 0.5% agar base in DMEM, 10% FBS. 24 hrs later, the number of cells seeded was determined by counting cells under the light microscope. After 3 weeks of growth in soft agar, the number of colonies (Colonies >5 cells) were counted; 10 different fields were quantified per well and the average number of colonies per field was calculated. The AIG index was expressed relative to the number of cells seeded. More than 70% of seeded spheroid cells formed colonies while none of seeded parent cells formed colonies. Experiments were repeated twice with similar results. *,p<0.01. Error bars: SD.
Figure 6. Spheroid cells are resistant to…
Figure 6. Spheroid cells are resistant to chemotherapy in vitro.
(A) More spheroid cells were killed by Cisplatin than parent cells. The same numbers of spheroid and parent cells were cultured in CM with indicated amount of Cisplatin for 2 days and numbers of cells from different time points were determined. (B) More parent cells survived after Cisplatin treatment for 48 h (Left). However, 8 days later, only spheroid cells were growing while parent cells disappeared (Right). (C) Spheroid and parent cells were treated with indicated amount of Cisplatin for 24 h, washed, and 300 of survived cells/well were cultured in CM for another 10 days, cell colonies were shown after crystal violet staining. The same clonogenic survival assays were repeated twice with similar results. Colonies formed (>5 cells) after 10 days of growth were counted and average and SD were shown. *,p

Figure 7. Spheroid cells show a hypoxia-resistant…

Figure 7. Spheroid cells show a hypoxia-resistant metabolism profile.

Metabolic profiles of a normal ovarian…

Figure 7. Spheroid cells show a hypoxia-resistant metabolism profile.
Metabolic profiles of a normal ovarian cell line RPNLOv78 as control, EOC cells RP-OV17534, and spheroid RP-OV17534 cells cultures in the presence of [U-13C6]glucose for 24 hrs (n = 3) showing (A) percentages of m3-13C lactate derived directed from glycolysis; (B) percentages of m2-13C lactate derived from pentose cycle; (C) ratio of m2 to m3 lactate; (D) percentage of 13C-labeled lactate of total lactate; (E) percentages of 13C-double-labeled glutamate in total 13C-labeled glutamate; (F) 13CO2 release from [U-13C6]glucose; (G) The fraction of newly synthesized palmitate and the 13C enrichment of acetyl units (H). Error bars: SD; *,p<0.01, compared with parental EOC cells. (I) EOC spheroid cells show increased anaerobic glycolysis, increased direct glucose oxidation in the pentose cycle, but low TCA cycle carbon flux, low anaplerotic flux, and de novo fatty acid synthesis. In contrast, parent EOC cells show increased TCA cycle and high anaplerotic flux. G6P, glucose-6-P; F6P, fructose-6-P; GAP, glyceraldehyde-3-P; 6PG, 6-P-gluconate; R5P, ribulose-5-P. Various thickness of lines represent different strengths of metabolic flux.
All figures (7)
Figure 7. Spheroid cells show a hypoxia-resistant…
Figure 7. Spheroid cells show a hypoxia-resistant metabolism profile.
Metabolic profiles of a normal ovarian cell line RPNLOv78 as control, EOC cells RP-OV17534, and spheroid RP-OV17534 cells cultures in the presence of [U-13C6]glucose for 24 hrs (n = 3) showing (A) percentages of m3-13C lactate derived directed from glycolysis; (B) percentages of m2-13C lactate derived from pentose cycle; (C) ratio of m2 to m3 lactate; (D) percentage of 13C-labeled lactate of total lactate; (E) percentages of 13C-double-labeled glutamate in total 13C-labeled glutamate; (F) 13CO2 release from [U-13C6]glucose; (G) The fraction of newly synthesized palmitate and the 13C enrichment of acetyl units (H). Error bars: SD; *,p<0.01, compared with parental EOC cells. (I) EOC spheroid cells show increased anaerobic glycolysis, increased direct glucose oxidation in the pentose cycle, but low TCA cycle carbon flux, low anaplerotic flux, and de novo fatty acid synthesis. In contrast, parent EOC cells show increased TCA cycle and high anaplerotic flux. G6P, glucose-6-P; F6P, fructose-6-P; GAP, glyceraldehyde-3-P; 6PG, 6-P-gluconate; R5P, ribulose-5-P. Various thickness of lines represent different strengths of metabolic flux.

References

    1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, et al. (2009) Cancer statistics, 2009. CA Cancer J Clin 59: 225–249.
    1. Armstrong D (2010) Update on treatment options for newly diagnosed ovarian cancer. Clin Adv Hematol Oncol 8: 675–678.
    1. Colombo N, Van Gorp T, Parma G, Amant F, Gatta G, et al. (2006) Ovarian cancer. Crit Rev Oncol Hematol 60: 159–179.
    1. Cannistra SA (2004) Cancer of the ovary. N Engl J Med 351: 2519–2529.
    1. Burleson KM, Boente MP, Pambuccian SE, Skubitz AP (2006) Disaggregation and invasion of ovarian carcinoma ascites spheroids. J Transl Med 4: 6.
    1. L’Esperance S, Bachvarova M, Tetu B, Mes-Masson AM, Bachvarov D (2008) Global gene expression analysis of early response to chemotherapy treatment in ovarian cancer spheroids. BMC Genomics 9: 99.
    1. Shield K, Riley C, Quinn MA, Rice GE, Ackland ML, et al. (2007) Alpha2beta1 integrin affects metastatic potential of ovarian carcinoma spheroids by supporting disaggregation and proteolysis. J Carcinog 6: 11.
    1. Zietarska M, Maugard CM, Filali-Mouhim A, Alam-Fahmy M, Tonin PN, et al. (2007) Molecular description of a 3D in vitro model for the study of epithelial ovarian cancer (EOC). Mol Carcinog 46: 872–885.
    1. Shield K, Ackland ML, Ahmed N, Rice GE (2009) Multicellular spheroids in ovarian cancer metastases: Biology and pathology. Gynecol Oncol 113: 143–148.
    1. Boros LG, Lapis K, Szende B, Tomoskozi-Farkas R, Balogh A, et al. (2001) Wheat germ extract decreases glucose uptake and RNA ribose formation but increases fatty acid synthesis in MIA pancreatic adenocarcinoma cells. Pancreas 23: 141–147.
    1. Boros LG, Lee PW, Brandes JL, Cascante M, Muscarella P, et al. (1998) Nonoxidative pentose phosphate pathways and their direct role in ribose synthesis in tumors: is cancer a disease of cellular glucose metabolism? Med Hypotheses 50: 55–59.
    1. Dang CV, Semenza GL (1999) Oncogenic alterations of metabolism. Trends Biochem Sci 24: 68–72.
    1. Boros LG, Cascante M, Lee WN (2002) Metabolic profiling of cell growth and death in cancer: applications in drug discovery. Drug Discov Today 7: 364–372.
    1. Raamsdonk LM, Teusink B, Broadhurst D, Zhang N, Hayes A, et al. (2001) A functional genomics strategy that uses metabolome data to reveal the phenotype of silent mutations. Nat Biotechnol 19: 45–50.
    1. Bertozzi CC, Chang CY, Jairaj S, Shan X, Huang J, et al. (2006) Multiple initial culture conditions enhance the establishment of cell lines from primary ovarian cancer specimens. In Vitro Cell Dev Biol Anim 42: 58–62.
    1. Shepherd TG, Theriault BL, Campbell EJ, Nachtigal MW (2006) Primary culture of ovarian surface epithelial cells and ascites-derived ovarian cancer cells from patients. Nat Protoc 1: 2643–2649.
    1. Zhang S, Balch C, Chan MW, Lai HC, Matei D, et al. (2008) Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res 68: 4311–4320.
    1. Crouch SP, Kozlowski R, Slater KJ, Fletcher J (1993) The use of ATP bioluminescence as a measure of cell proliferation and cytotoxicity. J Immunol Methods 160: 81–88.
    1. Pejovic T, Pande NT, Mori M, Mhawech-Fauceglia P, Harrington C, et al. (2009) Expression profiling of the ovarian surface kinome reveals candidate genes for early neoplastic changes. Transl Oncol 2: 341–349.
    1. Vizan P, Boros LG, Figueras A, Capella G, Mangues R, et al. (2005) K-ras codon-specific mutations produce distinctive metabolic phenotypes in NIH3T3 mice [corrected] fibroblasts. Cancer Res 65: 5512–5515.
    1. Lee WN, Boros LG, Puigjaner J, Bassilian S, Lim S, et al. (1998) Mass isotopomer study of the nonoxidative pathways of the pentose cycle with [1,2–13C2]glucose. Am J Physiol 274: E843–851.
    1. Katz J, Lee WN, Wals PA, Bergner EA (1989) Studies of glycogen synthesis and the Krebs cycle by mass isotopomer analysis with [U-13C]glucose in rats. J Biol Chem 264: 12994–13004.
    1. Kasho VN, Cheng S, Jensen DM, Ajie H, Lee WN, et al. (1996) Feasibility of analysing [13C]urea breath tests for Helicobacter pylori by gas chromatography-mass spectrometry in the selected ion monitoring mode. Aliment Pharmacol Ther 10: 985–995.
    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 U S A 100: 3983–3988.
    1. Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ (2005) Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 65: 10946–10951.
    1. Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, et al. (2007) Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci U S A 104: 10158–10163.
    1. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, et al. (2007) Identification of pancreatic cancer stem cells. Cancer Res 67: 1030–1037.
    1. Patrawala L, Calhoun T, Schneider-Broussard R, Li H, Bhatia B, 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.
    1. Hill RP, Perris R (2007) “Destemming” cancer stem cells. J Natl Cancer Inst 99: 1435–1440.
    1. Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser A (2007) Tumor growth need not be driven by rare cancer stem cells. Science 317: 337.
    1. Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, et al. (2006) Cancer stem cells–perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res 66: 9339–9344.
    1. Loh YH, Wu Q, Chew JL, Vega VB, Zhang W, et al. (2006) The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nat Genet 38: 431–440.
    1. Mohle R, Kanz L (2007) Hematopoietic growth factors for hematopoietic stem cell mobilization and expansion. Semin Hematol 44: 193–202.
    1. Wiese C, Rolletschek A, Kania G, Blyszczuk P, Tarasov KV, et al. (2004) Nestin expression–a property of multi-lineage progenitor cells? Cell Mol Life Sci 61: 2510–2522.
    1. Bapat SA, Mali AM, Koppikar CB, Kurrey NK (2005) Stem and progenitor-like cells contribute to the aggressive behavior of human epithelial ovarian cancer. Cancer Res 65: 3025–3029.
    1. Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, 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.
    1. Carpentino JE, Hynes MJ, Appelman HD, Zheng T, Steindler DA, et al. (2009) Aldehyde dehydrogenase-expressing colon stem cells contribute to tumorigenesis in the transition from colitis to cancer. Cancer Res 69: 8208–8215.
    1. van den Hoogen C, van der Horst G, Cheung H, Buijs JT, Lippitt JM, et al. (2010) High aldehyde dehydrogenase activity identifies tumor-initiating and metastasis-initiating cells in human prostate cancer. Cancer Res 70: 5163–5173.
    1. Charafe-Jauffret E, Ginestier C, Bertucci F, Cabaud O, Wicinski J, et al... (2013) ALDH1-positive cancer stem cells predict engraftment of primary breast tumors and are governed by a common stem cell program. Cancer Res.
    1. Puiffe ML, Le Page C, Filali-Mouhim A, Zietarska M, Ouellet V, et al. (2007) Characterization of ovarian cancer ascites on cell invasion, proliferation, spheroid formation, and gene expression in an in vitro model of epithelial ovarian cancer. Neoplasia 9: 820–829.
    1. Warburg O (1956) On the origin of cancer cells. Science 123: 309–314.
    1. Lyssiotis CA, Vander-Heiden MG, Munoz-Pinedo C, Emerling BM (2012) Emerging concepts: linking hypoxic signaling and cancer metabolism. Cell Death Dis 3: e303.
    1. Kondoh H (2008) Cellular life span and the Warburg effect. Exp Cell Res 314: 1923–1928.
    1. Naora H, Montell DJ (2005) Ovarian cancer metastasis: integrating insights from disparate model organisms. Nat Rev Cancer 5: 355–366.
    1. Allen HJ, Porter C, Gamarra M, Piver MS, Johnson EA (1987) Isolation and morphologic characterization of human ovarian carcinoma cell clusters present in effusions. Exp Cell Biol 55: 194–208.
    1. Aaberg-Jessen C, Norregaard A, Christensen K, Pedersen CB, Andersen C, et al. (2013) Invasion of primary glioma- and cell line-derived spheroids implanted into corticostriatal slice cultures. Int J Clin Exp Pathol 6: 546–560.
    1. Luca AC, Mersch S, Deenen R, Schmidt S, Messner I, et al. (2013) Impact of the 3D Microenvironment on Phenotype, Gene Expression, and EGFR Inhibition of Colorectal Cancer Cell Lines. PLoS One 8: e59689.
    1. Vang S, Wu HT, Fischer A, Miller DH, Maclaughlan S, et al. (2013) Identification of Ovarian Cancer Metastatic miRNAs. PLoS One 8: e58226.
    1. Burleson KM, Hansen LK, Skubitz AP (2004) Ovarian carcinoma spheroids disaggregate on type I collagen and invade live human mesothelial cell monolayers. Clin Exp Metastasis 21: 685–697.
    1. Burleson KM, Casey RC, Skubitz KM, Pambuccian SE, Oegema TR Jr, et al. (2004) Ovarian carcinoma ascites spheroids adhere to extracellular matrix components and mesothelial cell monolayers. Gynecol Oncol 93: 170–181.
    1. Chiaradonna F, Moresco RM, Airoldi C, Gaglio D, Palorini R, et al. (2012) From cancer metabolism to new biomarkers and drug targets. Biotechnol Adv 30: 30–51.
    1. Ward PS, Thompson CB (2012) Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell 21: 297–308.
    1. Chen CT, Hsu SH, Wei YH (2010) Upregulation of mitochondrial function and antioxidant defense in the differentiation of stem cells. Biochim Biophys Acta 1800: 257–263.
    1. Prigione A, Fauler B, Lurz R, Lehrach H, Adjaye J (2010) The senescence-related mitochondrial/oxidative stress pathway is repressed in human induced pluripotent stem cells. Stem Cells 28: 721–733.
    1. Zhou Y, Zhou Y, Shingu T, Feng L, Chen Z, et al. (2011) Metabolic alterations in highly tumorigenic glioblastoma cells: preference for hypoxia and high dependency on glycolysis. J Biol Chem 286: 32843–32853.
    1. Yuan S, Wang F, Chen G, Zhang H, Feng L, et al. (2013) Effective elimination of cancer stem cells by a novel drug combination strategy. Stem Cells 31: 23–34.
    1. Nakano A, Tsuji D, Miki H, Cui Q, El Sayed SM, et al. (2011) Glycolysis inhibition inactivates ABC transporters to restore drug sensitivity in malignant cells. PLoS One 6: e27222.
    1. Liu PP, Liao J, Tang ZJ, Wu WJ, Yang J, et al... (2013) Metabolic regulation of cancer cell side population by glucose through activation of the Akt pathway. Cell Death Differ.
    1. Keith B, Simon MC (2007) Hypoxia-inducible factors, stem cells, and cancer. Cell 129: 465–472.

Source: PubMed

3
Předplatit