Brain metastases are among the most feared complications in breast cancer, as no therapy exists that prevents or eliminates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties that allow breast cancer cell growth within the brain tissue. To provide information in this direction, we established a human breast cancer cell model for brain metastasis based on circulating tumor cells from a breast cancer patient and variants of these cells derived from bone or brain lesions in immunodeficient mice. The brain-derived cells showed an increased potential for brain metastasis in vivo and exhibited a unique protein expression profile identified by large-scale proteomic analysis. This protein profile is consistent with either a selection of predisposed cells or bioenergetic adaptation of the tumor cells to the unique energy metabolism of the brain. Increased expression of enzymes involved in glycolysis, tricarboxylic acid cycle, and oxidative phosphorylation pathways suggests that the brain metastatic cells derive energy from glucose oxidation. The cells further showed enhanced activation of the pentose phosphate pathway and the glutathione system, which can minimize production of reactive oxygen species resulting from an enhanced oxidative metabolism. These changes promoted resistance of brain metastatic cells to drugs that affect the cellular redox balance. Importantly, the metabolic alterations are associated with strongly enhanced tumor cell survival and proliferation in the brain microenvironment. Thus, our data support the hypothesis that predisposition or adaptation of the tumor cell energy metabolism is a key element in breast cancer brain metastasis, and raise the possibility of targeting the functional differentiation in breast cancer brain lesions as a novel therapeutic strategy. [Cancer Res 2007;67(4):1472–86]

Brain metastases are the most feared complication in breast cancer. Nearly 20% of patients with advanced breast cancer are eventually diagnosed with brain lesions, making breast tumors the main source of metastatic brain disease in women (13). The incidence of brain metastases increases as patients live longer in response to improved cancer therapy, but no current regimen significantly affects breast cancer brain metastases. Palliative treatment extends survival in most cases for only a few weeks or months, but often with severe impact on the quality of life (35). Therefore, it is imperative to gain a better understanding of the nature and functionality of breast cancer cells that cause brain metastases for development of effective regimens to prevent and control this stage of the disease.

In the past, much attention focused on invasive properties of metastatic cancer cells and their ability to attract new blood vessels (68). However, intrinsic mechanisms allowing metastatic tumor cells to survive and proliferate in preferred target organs are poorly understood. Thus, the goal of this study was to analyze protein expression profiles of tumor cells from breast cancer brain metastases and gain insight into cellular properties that promote tumor cell survival in the unique brain microenvironment. Metastatic breast cancer cells colonize the brain mainly from the bloodstream (9). We therefore isolated circulating tumor cells from a stage IV breast cancer patient, reintroduced the cells into the bloodstream of immunodeficient mice, and recovered tumor cells from the brain, or long bone for comparison. To define determinants of breast cancer cell growth in the brain, we examined the protein expression profiles of the parental cell line and its brain or bone homing variants by multidimensional proteomic analysis, MudPIT (10). More than 300 proteins were found uniquely regulated in the brain metastatic cells. Most of these proteins are involved in cellular metabolism and cell stress response. Our proteomic results, transcriptional validation, and cell function analyses in vitro indicate that brain metastatic cells use enhanced mitochondrial respiratory pathways for energy production and antioxidant defense mechanisms. The metabolic changes identified in the brain metastatic cells may reflect a predisposition or adaptation of the tumor cells to the brain microenvironment where a constant high energy demand is met almost entirely by glucose oxidation (1114). Our results provide new evidence that brain metastatic breast cancer cells use energy metabolism pathways that are distinct from anaerobic glycolysis, which is predominant in most cancer cells in oxygen-poor tumor microenvironments. Furthermore, the redox state of brain metastatic breast cancer cells may provide a link between their energy metabolism and gene regulation. Our results show the significance of understanding the metabolic state of metastatic cells and support the rationale of targeting tumor energy metabolism as a potential therapy for breast cancer brain metastasis.

Real-time PCR primers. Real-time PCR was done with the following gene-specific primers:

Gene nameForwardReverse
MDH2 GCAGCCACTTTCACTTCTC ACTCCAGCCGGAATAACTAC 
TPI1 CACTGAGAAGGTTGTTTTCG TAAATGATACGGGTGCTCTG 
PCK2 ATCCACATCTGTGATGGAAC CGTCTTGCTCTCTACTCGTG 
ERRα TTCTCATCGCTGTCGCTGTCT CAGCCGCCGCACTAGTTG 
PGC-1α CTGGAGAGCCCCTGTGAGAGT GTGGGCTTGTACGGTGGTGT 
PGC-1β GTTTCACCTCCAGCCTCAGAG CCAGGCAGGCCTCAGATCTA 
IDH3A ATTGATCGGAGGTCTCGGTGT CAGGAGGGCTGTGGGATTC 
ACO2 CCCGAGGTGAAGAATGTCATC GAAGCCCGTTGTACCAGC 
PGLS TGAGGACTACGCCAAGAAG AGTTGCCACAAAGATGACAG 
ACAA2 ACAGACAATGCAGGTAGACG GCCAGTGGTGTGAAGTTATG 
GAPDH GAAGGTGAAGGTCGGAGTC GAAGATGGTGATGGGATTTC 
Gene nameForwardReverse
MDH2 GCAGCCACTTTCACTTCTC ACTCCAGCCGGAATAACTAC 
TPI1 CACTGAGAAGGTTGTTTTCG TAAATGATACGGGTGCTCTG 
PCK2 ATCCACATCTGTGATGGAAC CGTCTTGCTCTCTACTCGTG 
ERRα TTCTCATCGCTGTCGCTGTCT CAGCCGCCGCACTAGTTG 
PGC-1α CTGGAGAGCCCCTGTGAGAGT GTGGGCTTGTACGGTGGTGT 
PGC-1β GTTTCACCTCCAGCCTCAGAG CCAGGCAGGCCTCAGATCTA 
IDH3A ATTGATCGGAGGTCTCGGTGT CAGGAGGGCTGTGGGATTC 
ACO2 CCCGAGGTGAAGAATGTCATC GAAGCCCGTTGTACCAGC 
PGLS TGAGGACTACGCCAAGAAG AGTTGCCACAAAGATGACAG 
ACAA2 ACAGACAATGCAGGTAGACG GCCAGTGGTGTGAAGTTATG 
GAPDH GAAGGTGAAGGTCGGAGTC GAAGATGGTGATGGGATTTC 

Breast cancer cell model. BCM2 Parent cells were established from blood of a stage IV breast cancer patient with widespread metastasis and cultured in EMEM with 10% fetal bovine serum (FBS) after isolation with antiepithelial immunomagnetic beads (monoclonal antibody BerEP4; Dynal, Lake Success, NY). BCM2 parent cells (2.5 × 105) were i.v. injected into 5- to 6-week-old female severe combined immunodeficient (SCID) mice (C.B17/lcTac-Prkdc scid) and tumor cells were recovered from the brain (BCM2 BrainG1) or femur (BCM2 Bone) 6 weeks later. BCM2 BrainG2 cells were from a brain lesion after i.v. injection of BCM2 BrainG1 cells. For in vivo tracking, the cell lines were transduced with firefly luciferase (F-luc) in a lentiviral vector system with cytomegalovirus promoter, injected i.v. into female SCID mice and followed by repeated noninvasive bioluminescence imaging with an IVIS 200 system (Xenogen, Alameda, CA). For ex vivo organ imaging, mice were injected i.p. with 100 mg/kg d-luciferin 5 min before necropsy and the excised organs were incubated with 100 μg/mL d-luciferin (5 min). Bioluminescence signal was quantified as photons per second per square centimeter in defined regions of interest using Living Image software (Xenogen). Where appropriate, signal was normalized to the bioluminescence expression of each cell variant. Background luminescence in vivo was 1 × 105 to 2 × 105 photons/s. To measure tumor cell growth in the brain directly, 1 × 104 tumor cells in 2 μL were implanted into the forebrain of female SCID mice (2 mm lateral, 1 mm anterior to bregma, 3-mm depth from dura). All animal work was in accordance with The Scripps Research Institute Animal Resources (Association for Assessment and Accreditation of Laboratory Animal Care accredited).

Protein fractionation and preparation. BCM2 Parent, BCM2 Bone, and BCM2 BrainG1 cells were seeded at the same density and harvested at ∼80% confluency. Whole-cell protein extracts were prepared with the TotalProteinExtraction kit (BioChain, Hayward, CA) and protein concentration was determined by bicinchoninic acid (BCA) assay (Pierce, Rockford, IL). For each cell line, 1 mg of total lysate was resuspended in starting buffer following the buffer exchange method of the ProteomeLab PF2D kit from Beckman Coulter (Fullerton, CA). Cell lysates were resolved based on their isoelectric point (pI) using the method of Beckman Coulter (15) and fractions collected in intervals of 0.3 pH unit using the chromatofocusing separation of the ProteomeLab PF2D system. Eleven fractions were generated for each cell line, and each fraction was precipitated with trichloroacetic acid/acetone before in-solution digest with trypsin. Protein pellets from each fraction were resuspended in trypsin digestion buffer (50 mmol/L ammonium bicarbonate + 0.1% Rapigest; Waters Corp., Milford, MA) and digested with trypsin overnight at 37°C.

Multidimensional chromatography and tandem mass spectrometry. Peptide mixtures were resolved by strong cation exchange liquid chromatography upstream of reverse-phase liquid chromatography as described (16). Eluted peptides were electrosprayed directly into an LTQ ion trap mass spectrometer equipped with a nano-liquid chromatography electrospray ionization source (ThermoFinnigan, San Jose, CA). Full mass spectra were recorded over a 400 to 1,600 m/z range, followed by three tandem mass spectrometry (MS/MS) events sequentially generated in a data-dependent manner on the first, second, and third most intense ions selected from the full MS spectrum (at 35% collision energy). Mass spectrometer scan functions and high-performance liquid chromatography solvent gradients were controlled by the Xcalibur data system (ThermoFinnigan).

Interpretation of MS/MS data sets. SEQUEST (17) was used to match MS/MS spectra. The validity of peptide/spectrum matches was assessed using SEQUEST-defined parameters, the cross-correlation score (XCorr) and normalized difference in cross-correlation scores (ΔCn).

Distribution of XCorr and ΔCn values for (a) direct and (b) decoy database hits was obtained, and the two subsets were separated by quadratic discriminant analysis. Full separation of the direct and decoy subsets is not generally possible. Therefore, the discriminant score was set such that a false-positive rate of 5% was determined based on the number of accepted decoy database peptides. This procedure was independently done on data subsets for charge states +1, +2, and +3. In addition, spectra were only retained if they had a ΔCn of at least 0.08 and minimum XCorr of 1.8 for +1, 2.5 for +2, and 3.5 for +3 spectra. In addition, the minimum sequence length was seven amino acid residues. DTASelect (18) was used to select and sort peptide/spectrum matches passing this set of criteria. The human protein database used was downloaded from the International Protein Index (IPI) version 3.12 in November 2005 and reverse protein sequences of the IPI database were used as the decoy database. Peptide hits from multiple runs were compared using CONTRAST (18). Proteins were considered detected if they were identified by at least half tryptic status and more than two peptides. A semiquantitative comparison of protein expression was obtained based on the ratio of spectra counts (the number of spectra collected per protein; refs. 16, 19).

Western blot analysis. Whole-cell protein extracts were prepared with the TotalProteinExtraction kit (BioChain) and protein concentrations determined by BCA assay (Pierce). Total cell lysates (50 μg) were used to compare protein expression. Immunoblotting assays were carried out by standard procedures using AMP-activated protein kinase (AMPK)-α and phospho-AMPK-α(Thr172) antibodies from Cell Signaling Technology (Beverly, MA); anti–β-actin was used for equal loading control (Sigma, St. Louis, MO). Bands were detected using horseradish peroxidase–labeled secondary antibodies and enhanced chemiluminescence (Amersham Pharmacia, Piscataway, NJ).

Determination of cellular ATP. Cellular ATP levels were measured in 96-well plates with the CellTiter Glo luminescence ATP assay (Promega) according to the manufacturer's instructions. Cells (2.5 × 104 per well) in growth medium containing 10% FBS were seeded in quadruplicates and the assays repeated thrice with similar results. The cells in this assay were not genetically tagged with luciferase. ATP-dependent luminescence was related to an ATP standard curve ranging from 10 nmol/L to 1 μmol/L.

Real-time quantitative PCR. Changes in mRNA expression of identified metabolic enzymes were examined by real-time PCR after reverse transcription of 400-ng RNA from each sample with SuperScript II reverse transcriptase (Invitrogen Life Technologies, Inc., Carlsbad, CA). cDNA was diluted 20-fold before PCR amplification using a 2720 Thermal Cycler and SYBR Green mix (Applied Biosystems, Foster City, CA). A typical protocol involved 10-min denaturation at 95°C, 40 cycles with denaturation at 95°C for 20 s, annealing at 60°C for 20 s, and extension at 72°C for 20 s, and final elongation at 72°C for 5 min. Melting curve analysis verified that all primers yielded a single PCR product. Priming specificity for human genes was established with mouse brain cDNA. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) primers were used for mRNA normalization and quantitative real-time PCR was done twice in triplicates for each gene.

Cytotoxicity assay with bortezomib and 2-deoxy-d-glucose. Cells (1 × 104 per well in 96-well plates) were seeded in growth medium and allowed to attach overnight before three gentle washes with PBS and addition of bortezomib or 2-deoxy-d-glucose (2DG). Bortezomib (VELCADE, Millenium Pharmaceuticals, Inc., Cambridge, MA) was reconstituted in DMSO and 2DG in water. Bortezomib dilutions from 40 μmol/L to 0.04 μmol/L were made with growth medium containing 2% FBS, and 2DG ranging from 20 mmol/L to 0.31 mmol/L with growth medium containing 2% dialyzed FBS (Invitrogen). Drug-treated cells were compared with cells treated with the respective diluent medium. Cytotoxic effects of each drug were assessed after 24 h using the Cell Counting-8 viability assay (Dojindo Molecular Technologies, Inc., Gaithersburg, MD). Each experiment was repeated twice.

Determination of total glutathione. Total glutathione was determined in cell lysates as described (20). Cell pellets were resuspended in 2 volumes of PBS, lysed by freezing and thawing in liquid nitrogen, sonicated twice for 10 s, and centrifuged at 11,800 × g at 4°C for 10 min. Ten microliters of supernatant were analyzed in 0.5-mL phosphate buffer [143 mmol/L sodium phosphate, 6 mmol/L EDTA (pH 7.5) containing 0.6 mmol/L 5,5′-dithiobis(2-nitrobenzoic acid), 0.5 units/mL glutathione reductase, and 0.3 mmol/L NADPH]. Glutathione reductase was produced recombinantly as described (21). Reduction of 5,5′-dithiobis(2-nitrobenzoic acid) was measured at 25°C by light absorbance at 412 nm. Glutathione contents were determined against a calibration curve and corrected for total protein content. Protein concentration of cell extracts was determined by the Bradford method.

Establishing a tumor cell model for breast cancer brain metastasis. It is generally accepted that metastatic breast cancer cells reach the brain primarily from the bloodstream (4). We therefore reasoned that blood-borne tumor cells could serve as a source for brain homing breast cancer cells and as a control against which cells derived from brain metastases can be compared with defined determinants of metastatic growth in the brain. We thus isolated circulating tumor cells from a stage IV breast cancer patient, established the tumor cells in culture, reintroduced them into the bloodstream of immunodeficient mice by tail-vein injection, and recovered tumor cells that had colonized the brain, or long bone for comparison. The cell line representing the circulating tumor cells is named BCM2 Parent (22), and their derivatives from bone lesion or brain lesions BCM2 Bone or BCM2 BrainG1, respectively. To enrich for a brain metastatic phenotype, BCM2 BrainG1 cells were subjected to an additional round of in vivo selection and the resulting cells were named BCM2 BrainG2. To establish the brain homing properties of these cells, they were genetically tagged with F-luc and their fate and distribution followed in female SCID mice after tail-vein injection by noninvasive bioluminescence imaging. BCM2 Parent cells colonized the brain and all other major target organs of breast cancer metastasis as seen in the clinic (Fig. 1A). A similar spectrum of organ colonization was observed for the in vivo selected cell variants derived from bone (BCM2 Bone) or brain metastases (BCM2 BrainG2), indicating a pluripotential of these cells on injection into the tail vein. Despite the fact that this route favors lung colonization, because the pulmonary vasculature is the first capillary bed that the cells encounter, the incidence of brain metastases in mice injected with the brain lesion-derived cells (83%) was significantly higher than that seen for BCM2 Parent (22%) or bone-derived cells (20%; Fig. 1B). Furthermore, tumor burden in the brain caused by the brain metastatic cells was >36-fold larger than that induced by the parental cells, and >17-fold larger than that caused by the bone metastatic cells. Measurements were based on total photon flux in the excised brains (Fig. 1C and D). Thus, BCM2 Parent cells represent a circulating breast cancer cell population that can efficiently colonize the brain from the bloodstream. Importantly, brain homing descendants of these cells show an increased propensity to survive and proliferate within the brain microenvironment. These results indicate a stable change in the functional phenotype of brain metastatic breast cancer cells and provide the basis for a molecular characterization of this specialized cell type.

Figure 1.

Human tumor cell model for breast cancer brain metastasis. Metastatic profiles of BCM2 cell variants. BCM2 Parent cells were established from a blood sample of a stage IV breast cancer patient. Bone and brain metastatic variants of these cells, designated as BCM2 Bone and BCM2 BrainG2, respectively, were derived from metastatic bone or brain lesions in female SCID mice after injecting BCM2 Parent cells into the tail vein. A, widespread metastatic activity in female SCID mice measured by noninvasive bioluminescence imaging 56 d after tail-vein injection of 2.5 × 105 F-luc–tagged tumor cells. Three representative mice are shown for each experimental group (n = 15). B, increased incidence of brain metastases in mice injected with the in vivo selected brain metastatic cells (BCM2 BrainG2). *, P < 0.0001, Fisher's exact test. Brain metastasis was measured by ex vivo bioluminescence imaging of the excised brains 8 wk after tail-vein injection of 2.5 × 105 BCM2 Parent cells (22% incidence; n = 18), BCM2 BrainG2 cells (83% incidence; n = 23), or BCM2 Bone cells (20% incidence; n = 15). C, metastatic burden in the brain induced by BCM2 BrainG2 cells is >36-fold larger than that induced by BCM2 Parent cells and >17-fold larger than that by BCM2 Bone cells. Total metastatic load was measured ex vivo in the excised brains 56 d after tail-vein injection of the tumor cells. Brain tumor burden was determined by photon flux (photons/s/cm2) in defined regions of interest covering the entire brain and normalized to the bioluminescence activity of each cell line. D, representative images of excised brains from each group shown on an identical signal scale to illustrate the differences in metastatic burden.

Figure 1.

Human tumor cell model for breast cancer brain metastasis. Metastatic profiles of BCM2 cell variants. BCM2 Parent cells were established from a blood sample of a stage IV breast cancer patient. Bone and brain metastatic variants of these cells, designated as BCM2 Bone and BCM2 BrainG2, respectively, were derived from metastatic bone or brain lesions in female SCID mice after injecting BCM2 Parent cells into the tail vein. A, widespread metastatic activity in female SCID mice measured by noninvasive bioluminescence imaging 56 d after tail-vein injection of 2.5 × 105 F-luc–tagged tumor cells. Three representative mice are shown for each experimental group (n = 15). B, increased incidence of brain metastases in mice injected with the in vivo selected brain metastatic cells (BCM2 BrainG2). *, P < 0.0001, Fisher's exact test. Brain metastasis was measured by ex vivo bioluminescence imaging of the excised brains 8 wk after tail-vein injection of 2.5 × 105 BCM2 Parent cells (22% incidence; n = 18), BCM2 BrainG2 cells (83% incidence; n = 23), or BCM2 Bone cells (20% incidence; n = 15). C, metastatic burden in the brain induced by BCM2 BrainG2 cells is >36-fold larger than that induced by BCM2 Parent cells and >17-fold larger than that by BCM2 Bone cells. Total metastatic load was measured ex vivo in the excised brains 56 d after tail-vein injection of the tumor cells. Brain tumor burden was determined by photon flux (photons/s/cm2) in defined regions of interest covering the entire brain and normalized to the bioluminescence activity of each cell line. D, representative images of excised brains from each group shown on an identical signal scale to illustrate the differences in metastatic burden.

Close modal

Establishing a molecular profile of brain metastatic breast cancer cells. To define determinants of metastatic growth of breast cancer cells in the brain, we examined the protein expression profiles of the parental cell line and its brain or bone homing descendants by shotgun proteomic analyses. Total protein lysates were fractionated by chromatofocusing using the PF2D ProteomeLab System (Beckman Coulter) and the fractions analyzed by multidimensional protein identification technology, MudPIT, to establish comprehensive protein profiles of BCM2 Parent, BCM2 BrainG1, and BCM2 Bone cells. More than 300 identified proteins were found ≥2-fold up-regulated or down-regulated in the brain metastatic cells compared with the parental cells and bone metastatic variant. Functional classification of the proteins found differentially expressed in the brain metastatic cells revealed that 50% belong to the functional category of cell metabolism. Proteins involved in cell stress response comprise the second largest category (8%). Given the strong predominance of alterations in the metabolic protein profile of the brain metastatic cells, we focused our attention on proteins involved in cell metabolism and identified 63 proteins, whose differential expression particularly marks the brain-derived cells. These proteins were further clustered into three subcategories: glucose oxidation, fatty acid oxidation, and cellular redox-active proteins (Table 1).

Table 1.

Proteins identified by proteomic analysis as differentially expressed in brain metastatic breast cancer cells

LocusDescriptionLMWpIP SeqCountP SpecCountP SeqCovBo SeqCountBo SpecCountBo SeqCovBr SeqCountBr SpecCountBr SeqCovCellular componentBiological process
Energy pathway                
    IPI00019383 Galactokinase 392 42,272 6.5 × × × 6.9 12 40 32.7 cytoplasm galactose metabolism; metabolism 
    IPI00294380 Phosphoenolpyruvate carboxykinase, mitochondrial precursor 640 70,637 7.6 × × × × × × 15 11.7 mitochondrion gluconeogenesis 
    IPI00465028 Triose phosphate isomerase 1 variant 249 26,713 7.4 13 79 57 15 77 50.6 35 339 65.9 none gluconeogenesis; glycolysis 
    IPI00017895 Glycero l-3-phosphate dehydrogenase, mitochondrial precursor 727 80,815 7.4 × × × × × × 5.5 mitochondrion glucose catabolism 
    IPI00383237 Pyruvate kinase M2 530 57,781 7.7 × × × × × × none glycolysis 
    IPI00549725 Phosphoglycerate mutase 1 266 30,048 24 19.2 11.3 76 25.9 cytosol glycolysis 
    IPI00025366 Citrate synthase, mitochondrial precursor 466 51,712 8.3 6.2 × × 14 12.2 mitochondrion tricarboxylic acid cycle 
    IPI00017855 Aconitate hydratase, mitochondrial precursor 780 85,425 7.6 11 17.7 11 7.6 22 75 33.7 mitochondrion tricarboxylic acid cycle 
    IPI00305166 Succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial precursor 664 72,692 7.4 × × × × × × 13 4.4 mitochondrion tricarboxylic acid cycle 
    IPI00296053 Fumarate hydratase, mitochondrial precursor 510 54637 8.8 11 35 20 17 17.8 12 72 24.9 mitochondrion; TCA cycle enzyme complex tricarboxylic acid cycle 
    IPI00011107 Isocitrate dehydrogenase [NADP], mitochondrial precursor 452 50,909 8.7 7.3 × × × 23 18.8 mitochondrion tricarboxylic acid cycle 
    IPI00291006 Malate dehydrogenase, mitochondrial precursor 338 35,531 8.7 12.1 × × × 40 26.3 mitochondrial matrix tricarboxylic acid cycle 
    IPI00219217 l-lactate dehydrogenase B chain (LDH-H) 333 36,507 6.1 12.9 × × × 16 15.6 cytoplasm l-lactate dehydrogenase activity; oxidoreductase activity tricarboxylic acid cycle; anaerobic glycolysis 
    IPI00176698 Cytochrome c 105 11,966 9.5 10 40 × × × × × × none electron transport 
    IPI00021793 Cytochrome c oxidase polypeptide VIa-liver, mitochondrial precursor; COX6A1 109 12,155 9.3 × × × × × × 24.8 mitochondrial membrane; inner membrane electron transport 
    IPI00006579 Cytochrome c oxidase subunit IV isoform 1, mitochondrial precursor 169 19,577 9.5 12 20.1 × × × × × × mitochondrion; inner membrane electron transport 
    IPI00008398 Cytochrome P450 26B1 512 57,513 8.5 8.4 × × × × × × endoplasmic reticulum; microsome; membrane electron transport 
    IPI00010810 Electron transfer flavoprotein α-subunit, mitochondrial precursor 333 35,080 8.4 × × × 11 25 87 58.3 mitochondrial matrix electron transport 
    IPI00513827 Hypothetical protein DKFZp686M24262 454 50,271 7.8 × × × × × × 12.8 none electron transport 
    IPI00032297 Isovaleryl CoA dehydrogenase 426 46,568 17 10.8 mitochondrial matrix electron transport 
    IPI00031109 Mimitin, mitochondrial precursor 169 19,856 × × × × × × 16.6 mitochondrion; mitochondrial inner membrane electron transport 
    IPI00619898 NAD(P)H menadione oxidoreductase 1, dioxin-inducible isoform c 236 26,365 8.8 22 21.6 × × × 11.9 none electron transport 
    IPI00419266 NADH dehydrogenase (ubiquinone) 1α subcomplex, 6, 14 kDa 154 17,871 10.1 17.5 × × × × × × mitochondrion; mitochondrial inner membrane electron transport 
    IPI00026964 Ubiquinol-cytochrome c reductase iron-sulfur subunit, mitochondrial precursor 274 29,652 8.3 × × × × × × 9.9 mitochondrion; respiratory chain complex III (sensu Eukarya) electron transport 
    IPI00011217 NADH-ubiquinone oxidoreductase 18 kDa subunit, mitochondrial precursor 175 20,108 10.3 11 10.3 × × × × × × membrane fraction; mitochondrion “mitochondrial electron transport, NADH to ubiquinone” 
    IPI00025796 NADH-ubiquinone oxidoreductase 30 kDa subunit, mitochondrial precursor 264 30,242 7.5 × × × × × × 17.4 membrane fraction; mitochondrion mitochondrial electron transport, NADH to ubiquinone 
    IPI00028520 NADH-ubiquinone oxidoreductase 51 kDa subunit, mitochondrial precursor 464 50,817 8.2 × × × × × × 14.4 mitochondrion; mitochondrial inner membrane mitochondrial electron transport, NADH to ubiquinone 
    IPI00255052 NADH-ubiquinone oxidoreductase B22 subunit 178 21,700 8.4 × × × × × × 14 mitochondrion; mitochondrial inner membrane mitochondrial electron transport, NADH to ubiquinone 
    IPI00654562 Cytochrome c oxidase polypeptide VIb 115 13,317 5.5 × × × × × × 25.2 mitochondrion electron transport; metabolism 
    IPI00013847 Ubiquinol-cytochrome-c reductase complex core protein I, mitochondrial precursor 480 52,619 6.4 × × × 14 mitochondrion; inner membrane electron transport; oxidative phosphorylation 
    IPI00305383 Ubiquinol-cytochrome-c reductase complex core protein 2, mitochondrial precursor 453 48,443 8.6 11 8.2 × × × 24 20.3 mitochondrial electron transport chain electron transport; oxidative phosphorylation 
    IPI00550882 Pyrroline-5-carboxylate reductase 1 319 33361 7.6 × × × × × × 8.8 none electron transport 
    IPI00025252 Protein disulfide-isomerase A3 precursor 505 56,782 6.4 13 × × × 5.3 endoplasmic reticulum electron transport; protein-nucleus import; 
    IPI00021785 Cytochrome c oxidase polypeptide Vb, mitochondrial precursor 129 13,696 8.8 × × × 12 14 31 32.6 mitochondrial membrane; inner membrane electron transport 
    IPI00012069 NAD(P)H dehydrogenase [quinone] 1 274 30,868 8.9 22 18.6 × × × 10.2 cytoplasm electron transport; xenobiotic metabolism 
    IPI00219381 NADH-ubiquinone oxidoreductase B8 subunit 98 10,790 9.6 31.6 × × × × × × membrane fraction; mitochondrion energy pathways 
    IPI00029561 NADH-ubiquinone oxidoreductase 42 kDa subunit, mitochondrial precursor 355 40,751 8.5 × × × × × × 17.2 membrane fraction; mitochondrion energy pathways 
    IPI00027776 Ferrochelatase, mitochondrial precursor 423 47,862 8.7 × × × × × × 9.2 mitochondrion energy pathways 
    IPI00553153 Hypothetical protein DKFZp564G0422 107 12,405 9.6 12 15.9 × × × 11.2 mitochondrion energy pathways 
    IPI00419255 ATP6V1F protein 119 13,370 5.5 × × × × × × 10 46.2 proton-transporting two-sector ATPase complex ATP synthesis coupled proton transport 
    IPI00303476 ATP synthase β chain, mitochondrial precursor 529 56,560 5.4 × × × 5.5 5.1 mitochondrion; proton-transporting ATP synthase complex ATP synthesis coupled proton transport; proton transport 
    IPI00029133 ATP synthase B chain, mitochondrial precursor 256 28,909 9.4 12.5 × × × × × × mitochondrial matrix ATP synthesis coupled proton transport; proton transport 
    IPI00218848 ATP synthase e chain, mitochondrial 68 7,802 9.4 27 55.9 × × × × × × mitochondrion; proton-transporting two-sector ATPase complex ATP synthesis coupled proton transport; proton transport 
    IPI00003856 Vacuolar ATP synthase subunit E 226 26,145 6.2 × × × × × × plasma membrane; proton-transporting ATPase complex ATP synthesis coupled proton transport; proton transport 
    IPI00642733 NADH:ubiquinone oxidoreductase 206 22,143 9.9 9.7 16 × × × none mitochondrial electron transport, NADH to ubiquinone 
                
Fatty acid β-oxidation                
    IPI00298406 3-Hydroxyacyl-CoA dehydrogenase, isoform 2 390 42,123 9.3 7.2 8.2 33 22.8 none fatty acid metabolism 
    IPI00001539 3-ketoacyl-CoA thiolase, mitochondrial 397 41,924 8.1 × × × × × × 11 14.4 mitochondrion lipid metabolism 
    IPI00005040 Acyl-CoA dehydrogenase, medium-chain specific, mitochondrial precursor 421 46,588 8.4 × × × × × × 13.8 mitochondrial matrix fatty acid β-oxidation 
    IPI00333838 Cytosolic acyl CoA thioester hydrolase, inducible 421 46,277 7.3 × × × × × × 10 24 none lipid metabolism 
    IPI00011416 Δ3,5-δ2,4-dienoyl-CoA isomerase, mitochondrial precursor 328 35,994 7.1 × × × × × × 11 mitochondrion; peroxisome fatty acid β-oxidation 
    IPI00024993 Enoyl-CoA hydratase, mitochondrial precursor 290 31,387 8.1 × × × × × × 11.7 mitochondrion fatty acid β-oxidation 
    IPI00017726 3-hydroxyacyl-CoA dehydrogenase type-2 261 26,923 7.8 × × × × × × 26.4 mitochondrion; plasma membrane lipid metabolism 
    IPI00298202 Peroxisomal acyl-CoA thioester hydrolase 1 319 35,914 7.6 × × × × × × 11 peroxisome lipid metabolism; acyl-CoA metabolism 
    IPI00294398 Short chain 3-hydroxyacyl-CoA dehydrogenase, mitochondrial precursor 314 34,278 8.9 8.9 10.2 33 28.3 mitochondrion fatty acid metabolism 
    IPI00010415 Splice Isoform 1 of Cytosolic acyl CoA thioester hydrolase 380 41,796 8.5 30 11.3 16 13.9 60 22.6 cytoplasm lipid metabolism 
    IPI00220906 Splice Isoform 1 of Peroxisomal acyl-CoA thioester hydrolase 2a 483 53,257 8.7 × × × × × × 10 20.9 peroxisome lipid metabolism; acyl-CoA metabolism 
    IPI00031522 Trifunctional enzyme α subunit, mitochondrial precursor 763 83,000 18 10.7 × × × 10 48 19 mitochondrion fatty acid metabolism 
    IPI00022793 Trifunctional enzyme β subunit, mitochondrial precursor 475 51,396 9.4 7.6 × × × 35 19.4 mitochondrial membrane fatty acid β-oxidation 
                
Cell redox homeostasis                
    IPI00412561 Glutaredoxin family protein 379 42,170 9.2 × × × × × × 11.3 none cell redox homeostasis 
    IPI00333763 Glutaredoxin-related protein C14orf87 157 16,628 6.8 × × × × × × 11 28 mitochondrion cell redox homeostasis 
    IPI00219757 Glutathione S-transferase P 209 23,225 5.6 14.4 × × × 22 27.8 none glutathione transferase activity; transferase activity central nervous system development; metabolism 
    IPI00016862 Glutathione reductase, mitochondrial precursor 522 56,257 8.5 × × × × × × 25 20.3 mitochondrion cell redox homeostasis; glutathione metabolism 
    IPI00465436 Catalase 526 59,625 7.4 10.3 × × × 11 22.6 peroxisome cell redox homeostasis; response to oxidative stress 
    IPI00289800 Glucose-6-phosphate dehydrogenase 515 59,257 6.8 14 17.3 6.6 11 28 30.7 cellular component unknown Hexose Monophosphate shunt 
    IPI00029997 6-phosphogluconolactonase 258 27,547 6.1 × × × × × × 10.9 none Hexose Monophosphate shunt 
LocusDescriptionLMWpIP SeqCountP SpecCountP SeqCovBo SeqCountBo SpecCountBo SeqCovBr SeqCountBr SpecCountBr SeqCovCellular componentBiological process
Energy pathway                
    IPI00019383 Galactokinase 392 42,272 6.5 × × × 6.9 12 40 32.7 cytoplasm galactose metabolism; metabolism 
    IPI00294380 Phosphoenolpyruvate carboxykinase, mitochondrial precursor 640 70,637 7.6 × × × × × × 15 11.7 mitochondrion gluconeogenesis 
    IPI00465028 Triose phosphate isomerase 1 variant 249 26,713 7.4 13 79 57 15 77 50.6 35 339 65.9 none gluconeogenesis; glycolysis 
    IPI00017895 Glycero l-3-phosphate dehydrogenase, mitochondrial precursor 727 80,815 7.4 × × × × × × 5.5 mitochondrion glucose catabolism 
    IPI00383237 Pyruvate kinase M2 530 57,781 7.7 × × × × × × none glycolysis 
    IPI00549725 Phosphoglycerate mutase 1 266 30,048 24 19.2 11.3 76 25.9 cytosol glycolysis 
    IPI00025366 Citrate synthase, mitochondrial precursor 466 51,712 8.3 6.2 × × 14 12.2 mitochondrion tricarboxylic acid cycle 
    IPI00017855 Aconitate hydratase, mitochondrial precursor 780 85,425 7.6 11 17.7 11 7.6 22 75 33.7 mitochondrion tricarboxylic acid cycle 
    IPI00305166 Succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial precursor 664 72,692 7.4 × × × × × × 13 4.4 mitochondrion tricarboxylic acid cycle 
    IPI00296053 Fumarate hydratase, mitochondrial precursor 510 54637 8.8 11 35 20 17 17.8 12 72 24.9 mitochondrion; TCA cycle enzyme complex tricarboxylic acid cycle 
    IPI00011107 Isocitrate dehydrogenase [NADP], mitochondrial precursor 452 50,909 8.7 7.3 × × × 23 18.8 mitochondrion tricarboxylic acid cycle 
    IPI00291006 Malate dehydrogenase, mitochondrial precursor 338 35,531 8.7 12.1 × × × 40 26.3 mitochondrial matrix tricarboxylic acid cycle 
    IPI00219217 l-lactate dehydrogenase B chain (LDH-H) 333 36,507 6.1 12.9 × × × 16 15.6 cytoplasm l-lactate dehydrogenase activity; oxidoreductase activity tricarboxylic acid cycle; anaerobic glycolysis 
    IPI00176698 Cytochrome c 105 11,966 9.5 10 40 × × × × × × none electron transport 
    IPI00021793 Cytochrome c oxidase polypeptide VIa-liver, mitochondrial precursor; COX6A1 109 12,155 9.3 × × × × × × 24.8 mitochondrial membrane; inner membrane electron transport 
    IPI00006579 Cytochrome c oxidase subunit IV isoform 1, mitochondrial precursor 169 19,577 9.5 12 20.1 × × × × × × mitochondrion; inner membrane electron transport 
    IPI00008398 Cytochrome P450 26B1 512 57,513 8.5 8.4 × × × × × × endoplasmic reticulum; microsome; membrane electron transport 
    IPI00010810 Electron transfer flavoprotein α-subunit, mitochondrial precursor 333 35,080 8.4 × × × 11 25 87 58.3 mitochondrial matrix electron transport 
    IPI00513827 Hypothetical protein DKFZp686M24262 454 50,271 7.8 × × × × × × 12.8 none electron transport 
    IPI00032297 Isovaleryl CoA dehydrogenase 426 46,568 17 10.8 mitochondrial matrix electron transport 
    IPI00031109 Mimitin, mitochondrial precursor 169 19,856 × × × × × × 16.6 mitochondrion; mitochondrial inner membrane electron transport 
    IPI00619898 NAD(P)H menadione oxidoreductase 1, dioxin-inducible isoform c 236 26,365 8.8 22 21.6 × × × 11.9 none electron transport 
    IPI00419266 NADH dehydrogenase (ubiquinone) 1α subcomplex, 6, 14 kDa 154 17,871 10.1 17.5 × × × × × × mitochondrion; mitochondrial inner membrane electron transport 
    IPI00026964 Ubiquinol-cytochrome c reductase iron-sulfur subunit, mitochondrial precursor 274 29,652 8.3 × × × × × × 9.9 mitochondrion; respiratory chain complex III (sensu Eukarya) electron transport 
    IPI00011217 NADH-ubiquinone oxidoreductase 18 kDa subunit, mitochondrial precursor 175 20,108 10.3 11 10.3 × × × × × × membrane fraction; mitochondrion “mitochondrial electron transport, NADH to ubiquinone” 
    IPI00025796 NADH-ubiquinone oxidoreductase 30 kDa subunit, mitochondrial precursor 264 30,242 7.5 × × × × × × 17.4 membrane fraction; mitochondrion mitochondrial electron transport, NADH to ubiquinone 
    IPI00028520 NADH-ubiquinone oxidoreductase 51 kDa subunit, mitochondrial precursor 464 50,817 8.2 × × × × × × 14.4 mitochondrion; mitochondrial inner membrane mitochondrial electron transport, NADH to ubiquinone 
    IPI00255052 NADH-ubiquinone oxidoreductase B22 subunit 178 21,700 8.4 × × × × × × 14 mitochondrion; mitochondrial inner membrane mitochondrial electron transport, NADH to ubiquinone 
    IPI00654562 Cytochrome c oxidase polypeptide VIb 115 13,317 5.5 × × × × × × 25.2 mitochondrion electron transport; metabolism 
    IPI00013847 Ubiquinol-cytochrome-c reductase complex core protein I, mitochondrial precursor 480 52,619 6.4 × × × 14 mitochondrion; inner membrane electron transport; oxidative phosphorylation 
    IPI00305383 Ubiquinol-cytochrome-c reductase complex core protein 2, mitochondrial precursor 453 48,443 8.6 11 8.2 × × × 24 20.3 mitochondrial electron transport chain electron transport; oxidative phosphorylation 
    IPI00550882 Pyrroline-5-carboxylate reductase 1 319 33361 7.6 × × × × × × 8.8 none electron transport 
    IPI00025252 Protein disulfide-isomerase A3 precursor 505 56,782 6.4 13 × × × 5.3 endoplasmic reticulum electron transport; protein-nucleus import; 
    IPI00021785 Cytochrome c oxidase polypeptide Vb, mitochondrial precursor 129 13,696 8.8 × × × 12 14 31 32.6 mitochondrial membrane; inner membrane electron transport 
    IPI00012069 NAD(P)H dehydrogenase [quinone] 1 274 30,868 8.9 22 18.6 × × × 10.2 cytoplasm electron transport; xenobiotic metabolism 
    IPI00219381 NADH-ubiquinone oxidoreductase B8 subunit 98 10,790 9.6 31.6 × × × × × × membrane fraction; mitochondrion energy pathways 
    IPI00029561 NADH-ubiquinone oxidoreductase 42 kDa subunit, mitochondrial precursor 355 40,751 8.5 × × × × × × 17.2 membrane fraction; mitochondrion energy pathways 
    IPI00027776 Ferrochelatase, mitochondrial precursor 423 47,862 8.7 × × × × × × 9.2 mitochondrion energy pathways 
    IPI00553153 Hypothetical protein DKFZp564G0422 107 12,405 9.6 12 15.9 × × × 11.2 mitochondrion energy pathways 
    IPI00419255 ATP6V1F protein 119 13,370 5.5 × × × × × × 10 46.2 proton-transporting two-sector ATPase complex ATP synthesis coupled proton transport 
    IPI00303476 ATP synthase β chain, mitochondrial precursor 529 56,560 5.4 × × × 5.5 5.1 mitochondrion; proton-transporting ATP synthase complex ATP synthesis coupled proton transport; proton transport 
    IPI00029133 ATP synthase B chain, mitochondrial precursor 256 28,909 9.4 12.5 × × × × × × mitochondrial matrix ATP synthesis coupled proton transport; proton transport 
    IPI00218848 ATP synthase e chain, mitochondrial 68 7,802 9.4 27 55.9 × × × × × × mitochondrion; proton-transporting two-sector ATPase complex ATP synthesis coupled proton transport; proton transport 
    IPI00003856 Vacuolar ATP synthase subunit E 226 26,145 6.2 × × × × × × plasma membrane; proton-transporting ATPase complex ATP synthesis coupled proton transport; proton transport 
    IPI00642733 NADH:ubiquinone oxidoreductase 206 22,143 9.9 9.7 16 × × × none mitochondrial electron transport, NADH to ubiquinone 
                
Fatty acid β-oxidation                
    IPI00298406 3-Hydroxyacyl-CoA dehydrogenase, isoform 2 390 42,123 9.3 7.2 8.2 33 22.8 none fatty acid metabolism 
    IPI00001539 3-ketoacyl-CoA thiolase, mitochondrial 397 41,924 8.1 × × × × × × 11 14.4 mitochondrion lipid metabolism 
    IPI00005040 Acyl-CoA dehydrogenase, medium-chain specific, mitochondrial precursor 421 46,588 8.4 × × × × × × 13.8 mitochondrial matrix fatty acid β-oxidation 
    IPI00333838 Cytosolic acyl CoA thioester hydrolase, inducible 421 46,277 7.3 × × × × × × 10 24 none lipid metabolism 
    IPI00011416 Δ3,5-δ2,4-dienoyl-CoA isomerase, mitochondrial precursor 328 35,994 7.1 × × × × × × 11 mitochondrion; peroxisome fatty acid β-oxidation 
    IPI00024993 Enoyl-CoA hydratase, mitochondrial precursor 290 31,387 8.1 × × × × × × 11.7 mitochondrion fatty acid β-oxidation 
    IPI00017726 3-hydroxyacyl-CoA dehydrogenase type-2 261 26,923 7.8 × × × × × × 26.4 mitochondrion; plasma membrane lipid metabolism 
    IPI00298202 Peroxisomal acyl-CoA thioester hydrolase 1 319 35,914 7.6 × × × × × × 11 peroxisome lipid metabolism; acyl-CoA metabolism 
    IPI00294398 Short chain 3-hydroxyacyl-CoA dehydrogenase, mitochondrial precursor 314 34,278 8.9 8.9 10.2 33 28.3 mitochondrion fatty acid metabolism 
    IPI00010415 Splice Isoform 1 of Cytosolic acyl CoA thioester hydrolase 380 41,796 8.5 30 11.3 16 13.9 60 22.6 cytoplasm lipid metabolism 
    IPI00220906 Splice Isoform 1 of Peroxisomal acyl-CoA thioester hydrolase 2a 483 53,257 8.7 × × × × × × 10 20.9 peroxisome lipid metabolism; acyl-CoA metabolism 
    IPI00031522 Trifunctional enzyme α subunit, mitochondrial precursor 763 83,000 18 10.7 × × × 10 48 19 mitochondrion fatty acid metabolism 
    IPI00022793 Trifunctional enzyme β subunit, mitochondrial precursor 475 51,396 9.4 7.6 × × × 35 19.4 mitochondrial membrane fatty acid β-oxidation 
                
Cell redox homeostasis                
    IPI00412561 Glutaredoxin family protein 379 42,170 9.2 × × × × × × 11.3 none cell redox homeostasis 
    IPI00333763 Glutaredoxin-related protein C14orf87 157 16,628 6.8 × × × × × × 11 28 mitochondrion cell redox homeostasis 
    IPI00219757 Glutathione S-transferase P 209 23,225 5.6 14.4 × × × 22 27.8 none glutathione transferase activity; transferase activity central nervous system development; metabolism 
    IPI00016862 Glutathione reductase, mitochondrial precursor 522 56,257 8.5 × × × × × × 25 20.3 mitochondrion cell redox homeostasis; glutathione metabolism 
    IPI00465436 Catalase 526 59,625 7.4 10.3 × × × 11 22.6 peroxisome cell redox homeostasis; response to oxidative stress 
    IPI00289800 Glucose-6-phosphate dehydrogenase 515 59,257 6.8 14 17.3 6.6 11 28 30.7 cellular component unknown Hexose Monophosphate shunt 
    IPI00029997 6-phosphogluconolactonase 258 27,547 6.1 × × × × × × 10.9 none Hexose Monophosphate shunt 

NOTE: Protein expression profiles of BCM2 Parent cells and their bone- or brain lesion–derived variants, BCM2 Bone and BCM2 BrainG1, determined by shotgun proteomic analysis using multidimensional protein identification technology, MudPIT. List of proteins found ≥2-fold up-regulated or down-regulated in the brain metastatic cells compared with the parental and bone metastatic cells. The shown proteins represent the main functional categories that were found differentially expressed in the brain metastatic cells.

Abbreviations: SeqCount, sequence count: number of nonredundant peptides used to identify this protein; SpecCount, spectra count: sum of all peptides used to identify this protein; SeqCov, sequence coverage: percent of amino acid sequence covered by the identified peptides; ×, no peptide identification for the listed protein; P, BCM2 Parent; Bo, BCM2 Bone; Br, BCM2 BrainG1.

Brain metastatic cells show enhanced mitochondrial respiratory pathways for energy production. Combining our large-scale expression data and knowledge of known metabolic networks, we were able to construct a metabolic profile of brain metastatic cells based on the differentially expressed proteins listed in Table 1. Proteins up-regulated in the brain metastatic cells indicate three major changes in the energy metabolism of these cells: enhanced glycolysis coupled to increased activity of the tricarboxylic acid (TCA) cycle, increased β-oxidation of fatty acids, and an elevated pentose phosphate pathway. Major up-regulated proteins indicating these changes in the context of their energy metabolism pathways are highlighted in Fig. 2.

Figure 2.

Schematic representation of energy pathways altered in brain metastatic breast cancer cells based on proteomic identification of differentially expressed proteins. Schematic outline of oxidative energy pathways, highlighting proteins in gray italics that were found up-regulated in the brain metastatic cells. The first pathway includes glucose catabolism by glycolysis and the TCA cycle. The second pathway includes fatty acid β-oxidation, which generates acetyl CoA, a substrate for the TCA cycle. The third pathway includes the pentose phosphate pathway, which provides reducing power (NADPH) and can regulate cellular redox homeostasis.

Figure 2.

Schematic representation of energy pathways altered in brain metastatic breast cancer cells based on proteomic identification of differentially expressed proteins. Schematic outline of oxidative energy pathways, highlighting proteins in gray italics that were found up-regulated in the brain metastatic cells. The first pathway includes glucose catabolism by glycolysis and the TCA cycle. The second pathway includes fatty acid β-oxidation, which generates acetyl CoA, a substrate for the TCA cycle. The third pathway includes the pentose phosphate pathway, which provides reducing power (NADPH) and can regulate cellular redox homeostasis.

Close modal

Oxidative metabolism. Instead of generating energy through anaerobic glycolysis, which is often used by fast proliferating cancer cells including BCM2 Parent cells, BCM2 brain-derived cells seem to use primarily aerobic glycolysis, coupled to the TCA cycle and oxidative phosphorylation, to generate energy for cell growth. We found several lines of evidence supporting this hypothesis. First, we detected considerable up-regulation of proteins involved in glycolysis, TCA cycle (marked in gray italics in Fig. 2), and oxidative phosphorylation based on our proteomic analyses (Table 1). Second, we found corresponding increase in the transcription of genes encoding proteins that are involved in oxidative energy metabolism. Quantitative real-time PCR analyses confirmed a trend in gene expression changes consistent with our proteomic data and revealed a specific up-regulation of proteins involved in glycolysis and oxidative metabolism in the brain metastatic cells (Fig. 3). The up-regulated proteins include the glycolytic enzyme triose phosphate isomerase (TPI) and phosphoenolpyruvate carboxylase (PCK2), which can enhance TCA flux via cataplerosis. Also up-regulated were the TCA cycle enzymes, aconitate hydratase (ACO2), isocitric dehydrogenase (IDH3A), and mitochondrial malate dehydrogenase (MDH2; Fig. 3A). Furthermore, we detected increased expression of transcriptional regulators that promote oxidative metabolism, including members of the poly(ADP-ribose) polymerase-γ coactivator-1 (PGC-1) family of transcriptional coactivators, PGC-1α and PGC-1β (23), and the nuclear receptor ERRα (Fig. 3A). ERRα is a downstream effector of PGC-1α in the regulation of mitochondrial energy metabolism and enables PGC-1α to induce target genes such as IDH3A (24). The coordinated up-regulation of all three regulators, PGC-1α, PGC-1β, and ERRα, suggests that the brain metastatic cells have undergone a transcriptional switch that supports expression of oxidative metabolism pathways. Interestingly, PGC-1α, but not PGC-1β or ERRα, was elevated in the bone metastatic cells, suggesting that PGC-1α expression alone was not sufficient to activate the expression of downstream targets such as ACO2 and IDH3A. ERRα is also a regulator of other mitochondrial energy transduction pathways, including fatty acid oxidation and oxidative phosphorylation (2426). Consistent with this role, our proteomic results showed up-regulation of several enzymes involved in fatty acid oxidation, as well as components of the electron transfer chain (Table 1). Similar to the enzymes in the TCA cycle, we found increased mRNA levels for fatty acid oxidation genes, such as β-ketothiolase (ACAA2) in the brain metastatic cells (Fig. 3B). To evaluate an in vivo relevance of our gene and protein expression results obtained with in vitro cultured BCM2 cell variants, we propagated the brain homing metastatic cells in the mouse brain and analyzed gene expression directly in the brain lesions. Consistent with the results in Fig. 3A, to C, several of the genes up-regulated in the cultured brain metastatic cells were found at similarly elevated levels in the brain lesions (Fig. 3D). This finding indicates that the changes in gene regulation seen in the brain homing cells can be maintained when the cells are expanded culture.

Figure 3.

Differential expression of genes involved in energy metabolism in brain metastatic cells. mRNA expression of genes involved in energy metabolism was measured by real-time PCR. mRNA expression in BCM2 Bone, BrainG1, and BrainG2 cells was normalized to mRNA expression measured in BCM2 Parent cells. A, enzymes involved in glycolytic (TPI) or TCA cycle (PCK2, IDH3A, MDH2, and ACO2). B, enzymes involved in fatty acid oxidation (ACAA2) or the pentose phosphate pathway (PGLS). C, transcriptional regulators of oxidative metabolism programs (PGC-1α, PGC-1b, and ERRα). D, genes found up-regulated in the cultured brain metastatic cells were also expressed at similarly elevated levels in brain lesions caused by intracranially injected BCM2 BrainG2 cells.

Figure 3.

Differential expression of genes involved in energy metabolism in brain metastatic cells. mRNA expression of genes involved in energy metabolism was measured by real-time PCR. mRNA expression in BCM2 Bone, BrainG1, and BrainG2 cells was normalized to mRNA expression measured in BCM2 Parent cells. A, enzymes involved in glycolytic (TPI) or TCA cycle (PCK2, IDH3A, MDH2, and ACO2). B, enzymes involved in fatty acid oxidation (ACAA2) or the pentose phosphate pathway (PGLS). C, transcriptional regulators of oxidative metabolism programs (PGC-1α, PGC-1b, and ERRα). D, genes found up-regulated in the cultured brain metastatic cells were also expressed at similarly elevated levels in brain lesions caused by intracranially injected BCM2 BrainG2 cells.

Close modal

In addition to enzymes and transcriptional regulators, our proteomic analyses identified at least one signaling protein important for energy homeostasis, the AMPK, up-regulated in brain metastatic cells (Table 1). AMPK is a key regulator and has been implicated in the control of PGC-1α in muscle (27). We found increased levels of AMPK protein expression and activation, based on Thr172 phosphorylation, in the brain metastatic cells (Fig. 4A). This result supports a possible functional link between AMPK, PGC-1α expression, and the altered energy metabolism in the brain homing cells. Numerous studies have established AMPK as a key regulator of energy homeostasis within the cell. On activation, AMPK switches off ATP consuming biosynthetic pathways (e.g., fatty acid synthesis) and turns on ATP generating metabolic pathways (e.g., fatty acid oxidation and glycolysis) to preserve ATP levels for cell survival (28, 29). The role of fatty oxidation in the altered energy metabolism of brain metastatic breast cancer cells is less clear and may have been influenced by the culture conditions to which the cells were exposed. However, because one of the metabolites of β-ketothiolase is acetyl-CoA, a substrate for the TCA cycle, it is possible that fatty acid oxidation is switched on by AMPK at an increased rate in the brain metastatic cells to further support the enhanced mitochondrial respiratory chain pathways for energy production. In addition to acute effects of AMPK activation, long-term effects can influence the overall regulation of energy metabolism, including increased mitochondrial enzyme content and mitochondrial biogenesis (27, 28, 30, 31). Thus, AMPK can contribute to the maintenance of high ATP levels, which may stay remarkably stable for high energy–dependent molecular activities (32). Consistent with increased glycolysis and oxidative phosphorylation in the brain metastatic cells, indicated by our proteomic and transcriptional analyses, we found elevated levels of cellular ATP in these cells (Fig. 4B). It is possible that long-term effects promoted by constitutive increase in AMPK protein and activation support adaptation of the brain metastatic cells to energy pathways predominant in the brain and contribute to the growth advantage of tumor cells in the brain microenvironment.

Figure 4.

Expression and activation of AMPK in brain metastatic cells. A, AMPK protein expression in BCM2 Parent, BCM2 Bone, BCM2 BrainG1, and BCM2 BrainG2 cells analyzed by Western blot analysis. Activation of AMPK by phosphorylation was examined with the same sample set using specific antibodies against the phosphorylated form of AMPK [pAMPK(T172)]. β-Actin antibody was used to show equal loading of proteins in each lane. B, measurements of ATP pool levels in BCM2 Parent, Bone, and BrainG2 cells. Cellular ATP measured in 2.5 × 104 cells per well in 96-well plates and growth medium containing 10% FBS based on a luminescence ATP assay. Each analysis was done in quadruplicate and repeated twice. Columns, average values of ATP; bars, SD. Brain metastatic cells produced significantly higher ATP levels than the parental and bone metastatic cells. *, P = 0.0005, F = 10.2 (one-way ANOVA followed by Tukey's pairwise comparison). The cells used in this assay were not tagged with luciferase.

Figure 4.

Expression and activation of AMPK in brain metastatic cells. A, AMPK protein expression in BCM2 Parent, BCM2 Bone, BCM2 BrainG1, and BCM2 BrainG2 cells analyzed by Western blot analysis. Activation of AMPK by phosphorylation was examined with the same sample set using specific antibodies against the phosphorylated form of AMPK [pAMPK(T172)]. β-Actin antibody was used to show equal loading of proteins in each lane. B, measurements of ATP pool levels in BCM2 Parent, Bone, and BrainG2 cells. Cellular ATP measured in 2.5 × 104 cells per well in 96-well plates and growth medium containing 10% FBS based on a luminescence ATP assay. Each analysis was done in quadruplicate and repeated twice. Columns, average values of ATP; bars, SD. Brain metastatic cells produced significantly higher ATP levels than the parental and bone metastatic cells. *, P = 0.0005, F = 10.2 (one-way ANOVA followed by Tukey's pairwise comparison). The cells used in this assay were not tagged with luciferase.

Close modal

Pentose phosphate pathway. Besides enhanced mitochondrial respiratory chain pathways, we also detected an up-regulation in the brain metastatic cells of enzymes involved in the oxidative phase of the pentose phosphate pathway (Figs. 2 and 3B). The pentose phosphate pathway serves to generate NADPH and pentose (five-carbon) sugars. This pathway consists of two distinct phases: oxidative and nonoxidative. The oxidative phase generates NADPH and is one of three major ways in which the body creates reducing molecules to prevent oxidative stress (33). We found two key enzymes of the oxidative phase up-regulated in the brain metastatic cells: glucose-6-phosphate dehydrogenase, a NADPH producing enzyme, and 6-phosphogluconolactonase (Table 1). Oxidative stress within cells is controlled primarily by the action of the tripeptide glutathione (GSH), which can reduce peroxides to maintain a reducing milieu within the cell. Regeneration of reduced glutathione by flavoenzyme glutathione reductase uses NADPH as a source of reducing equivalents (34). Consistent with the known metabolic network connecting the pentose phosphate pathway and the glutathione system, we found an up-regulation of glutathione-dependent enzymes in the brain metastatic cells. Prominent examples are glutathione reductase and glutathione S-transferase P. In addition, catalase, another important antioxidant enzyme, was found up-regulated (Table 1; Fig. 2). We believe that the NADPH-producing phase of the pentose phosphate pathway, in combination with the induction of antioxidant enzymes, is crucial in the brain metastatic cells to support the detoxification of oxidative stress. Our results suggest that the brain metastatic cells have an enhanced mitochondrial respiratory metabolism, which may lead to an increased production of reactive oxygen species. We therefore hypothesize that enhanced antioxidative defense mechanisms are crucial in the cells to maintain reduced GSH and to minimize oxidative stress caused by reactive oxygen species in the brain microenvironment.

Brain metastatic cells show reduced susceptibility to 2DG-induced cytotoxicity. It is well established that many cancer cells exhibit increased rates of glycolysis and reduced rates of respiration (35, 36). Fast proliferating cancer cells adopt anaerobic glycolysis to support rapid growth, and this renders the cells sensitive to glucose deprivation (37, 38). Glucose deprivation causes oxidative stress in cancer cells, and a synthetic glucose analogue, 2DG, mimics the effect of glucose deprivation. 2DG has been used to target cancer cells by direct inhibition of glycolysis. Recently, 2DG has been shown to cause selective cytotoxicity in tumor tissue by mechanisms similar to those triggered by glucose deprivation (39, 40). To further confirm the metabolic differences between the originally circulating parental breast cancer cells and their bone- or brain lesion–derived metastatic variants, we studied the effect of 2DG on the growth rate of these cells. We found that the brain metastatic cells, BCM2 BrainG1 and BrainG2, were 2-fold less sensitive to 2DG treatment than the parental and bone metastatic breast cancer cells (Fig. 5A). Pyruvate, a metabolite of glucose provided in the culture medium, may allow the cells to bypass the requirement for glucose and can direct the energy production via oxidative metabolism. It is also possible that the enhanced antioxidant capacity seen in the brain metastatic cells contributes to the partial resistance of these cells to 2DG. Ample evidence suggests that glucose deprivation is sufficient to induce cytotoxicity in cancer cells through metabolic oxidative stress (41, 42). Glucose deprivation can lead to a lack of NADPH and pyruvate and, consequently, to an impairment of the hydroperoxide metabolism (42). Thus, the enhanced pentose phosphate pathway and glutathione system detected in the brain metastatic cells could help the cells to quickly correct any disturbance of the cellular pro-oxidant and antioxidant balance, whereas the parental and bone metastatic breast cancer cells apparently lack this flexibility.

Figure 5.

Differential response of brain metastatic cells to 2-DG and bortezomib. The cytotoxic effects of 2DG and bortezomib on BCM2 Parent, BCM2 Bone, BCM2 BrainG1, and BCM2 BrainG2 cells were measured and compared. The cells were exposed to serial dilutions of 2DG (0–20 mmol/L; A) or bortezomib (0–40 μmol/L; B) for 24 h before measuring cytotoxicity. The experiments were done in triplicate and repeated twice. Points, average values of two experiments; bars, SD. Comparing BCM2 Parent, Bone, BrainG1, and BrainG2 cells, IC50 values for 2DG were 0.97, 1.29, 3.12, and 3.12 mmol/L, respectively. Comparing the cells in the same order, IC50 values for bortezomib were 0.625, 10, ≫40, and ≫40 μmol/L. C, levels of glutathione. Total glutathione (GSH) was measured in BCM2 Parent, Bone, BrainG1, and BrainG2 cells after a 24-h exposure to 25 nmol/L bortezomib or drug-free growth medium. Concentration of GSH from each cell line was normalized to total protein concentration. Percent change in GSH level is shown for each cell line, comparing exposure to bortezomib versus normal growth medium (BCM2 Parent −13%, BCM2 Bone +7%, BCM2 BrainG1 +22%, BCM2 BrainG2 +13%). GSH production in BCM2 BrainG1 and BrainG2 cells in response to bortezomib is significantly higher than in BCM2 Parent cells. P < 0.0001, F = 19.2 (one-way ANOVA followed by Tukey's pairwise comparison).

Figure 5.

Differential response of brain metastatic cells to 2-DG and bortezomib. The cytotoxic effects of 2DG and bortezomib on BCM2 Parent, BCM2 Bone, BCM2 BrainG1, and BCM2 BrainG2 cells were measured and compared. The cells were exposed to serial dilutions of 2DG (0–20 mmol/L; A) or bortezomib (0–40 μmol/L; B) for 24 h before measuring cytotoxicity. The experiments were done in triplicate and repeated twice. Points, average values of two experiments; bars, SD. Comparing BCM2 Parent, Bone, BrainG1, and BrainG2 cells, IC50 values for 2DG were 0.97, 1.29, 3.12, and 3.12 mmol/L, respectively. Comparing the cells in the same order, IC50 values for bortezomib were 0.625, 10, ≫40, and ≫40 μmol/L. C, levels of glutathione. Total glutathione (GSH) was measured in BCM2 Parent, Bone, BrainG1, and BrainG2 cells after a 24-h exposure to 25 nmol/L bortezomib or drug-free growth medium. Concentration of GSH from each cell line was normalized to total protein concentration. Percent change in GSH level is shown for each cell line, comparing exposure to bortezomib versus normal growth medium (BCM2 Parent −13%, BCM2 Bone +7%, BCM2 BrainG1 +22%, BCM2 BrainG2 +13%). GSH production in BCM2 BrainG1 and BrainG2 cells in response to bortezomib is significantly higher than in BCM2 Parent cells. P < 0.0001, F = 19.2 (one-way ANOVA followed by Tukey's pairwise comparison).

Close modal

Brain metastatic cells show enhanced resistance to oxidative damage related to the cellular glutathione status. To further explore the hypothesis that an enhanced redox system in the brain metastatic cells can provide protection against oxidative stress, we studied the stress response of the parental breast cancer cells and their bone or brain metastatic variants to oxidative damage induced by bortezomib. Bortezomib, or PS341, is a specific inhibitor of proteasome activity that targets the protein degradation system. The ubiquitin-proteasome system is responsible for intracellular proteolysis, particularly the degradation of short-lived or oxidized proteins (43). Ample evidence suggests that proteasome inhibition induces mitochondrial dysfunction, increases generation of reactive oxygen species, elevates RNA and DNA oxidation, and promotes protein oxidation (44). We exposed the parental breast cancer cells, as well as their bone- and brain-derived metastatic variants, to increasing concentrations of bortezomib and found that the brain metastatic cells (BCM2 BrainG1 and BrainG2) were >60-fold less sensitive to this treatment than the parental cells (Fig. 5B). Furthermore, we measured the cellular content of total glutathione as an indicator of the cellular redox state under normal culture conditions and in the presence of bortezomib. Under normal growth conditions, which include 10% FBS in the medium, BCM2 Parent cells had the highest cellular glutathione levels. The difference in glutathione between the parental and brain metastatic cells under normal culture conditions might be related to the slower in vitro growth rate of BCM2 BrainG1 and BrainG2 cells, which we consistently observed (data not shown). However, in the presence of bortezomib, the glutathione levels in the brain metastatic cells notably increased (22% in BCM2 BrainG1 cells and 13% in BrainG2 cells) whereas GSH decreased (−13%) in the parental cells (Fig. 5C). This finding is consistent with the reduced sensitivity of the brain metastatic cells to this drug and the higher sensitivity of BCM2 Parent cells, where glutathione is depleted in response to bortezomib (Fig. 5B). This depletion likely reflects enhanced oxidation of glutathione to GSSG, followed by GSSG export. Together, these results support our hypothesis that brain metastatic cells can up-regulate and rapidly adapt their cellular antioxidant defense to protect against oxidative stress. Up-regulation of NADPH production coupled with enhanced glutathione reductase activity allows for the detoxification of higher reactive oxygen species and GSSG fluxes, thus maintaining high GSH levels. The observed changes in energy metabolism and cellular stress response may thus confer a growth advantage to brain metastatic breast cancer cells, allowing them to thrive in the microenvironment of the brain tissue.

Brain metastatic breast cancer cells have a growth advantage in the brain. Our experimental metastasis data and proteomic analyses indicate that the brain metastatic cells, selected in vivo for their ability to establish brain metastases, possess a phenotype distinct from the parental circulating tumor cells and their bone metastatic counterparts. The protein expression profile of the brain metastatic cells and its functional validation imply a predisposition or bioenergetic adaptation of the tumor cells to the energy metabolism of the brain, conferring an advantage for tumor cell survival and proliferation in the brain microenvironment. We found that i.v. injection of the brain lesion–derived breast cancer cells leads to a high incidence of brain metastasis significantly surpassing the parental cells and the bone metastatic variant (Fig. 1). However, this approach includes the possibility that the formation of brain metastases is a result of enhanced tumor cell survival in the bloodstream or an ability to cross the blood-brain barrier efficiently. To eliminate these factors and directly assess tumor cell survival and growth in the brain, we implanted the F-luc–tagged BCM2 BrainG2 versus BCM2 Bone cells into the forebrain of experimental mice by stereotactic injection. Tumor cell growth was followed by repeated noninvasive bioluminescence imaging over a period of 21 days. Based on the percent increase in photon flux over the signal of the original implant, we measured a significant enhancement in tumor cell growth for the brain metastatic cells compared with their bone-derived counterparts (Fig. 6A). After 21 days, the overall tumor burden in the brain caused by the brain metastatic cells was cumulatively ∼100-fold larger than that measured for the bone metastatic cells (Fig. 6B). Furthermore, we found that the brain metastatic cells spread to other areas of the central nervous system and frequently extended to the spine (Fig. 6C), similar to the clinical situation often seen in breast cancer patients with brain metastases. Thus, breast cancer cells from brain lesions can acquire a stable change in functional phenotype that confers a growth advantage in the central nervous system. Our proteomic data and cell function analyses suggest that this phenotype involves an adaptation in energy metabolism that promotes tumor cell growth in the brain.

Figure 6.

Growth advantage of brain metastatic breast cancer cells in the brain. For direct measurements of tumor cell growth rates within the brain tissue, 1 × 104 F-luc–tagged BCM2 BrainG2 versus BCM2 Bone cells were implanted into the forebrain of 6-wk-old female SCID mice by stereotactic injection at 2 mm lateral, 1 mm anterior to bregma, and 3-mm depth from dura. A, growth rate of brain implanted tumor cells measured over a period of 21 d by noninvasive bioluminescence imaging. The percent increase in tumor signal was significantly higher in BCM2 BrainG2 cells than in BCM2 Bone cells. P = 0.0005, t = 4.26 (two-sample t test comparing mean increase in photon flux with group sizes of n = 10). Each symbol represents an individual animal. Red bar, median value for each group. B, ex vivo imaging of excised brains 21 d after tumor cell implantation illustrates enhanced overall tumor burden and intracranial spreading of BCM2 BrainG2 cells compared with BCM2 Bone cells. C, metastatic extension from the site of implantation in the brain to the spine was seen only in mice injected with BCM2 BrainG2 cells. Two representative mice are shown from each group comparing BCM2 BrainG2 to BCM2 Bone.

Figure 6.

Growth advantage of brain metastatic breast cancer cells in the brain. For direct measurements of tumor cell growth rates within the brain tissue, 1 × 104 F-luc–tagged BCM2 BrainG2 versus BCM2 Bone cells were implanted into the forebrain of 6-wk-old female SCID mice by stereotactic injection at 2 mm lateral, 1 mm anterior to bregma, and 3-mm depth from dura. A, growth rate of brain implanted tumor cells measured over a period of 21 d by noninvasive bioluminescence imaging. The percent increase in tumor signal was significantly higher in BCM2 BrainG2 cells than in BCM2 Bone cells. P = 0.0005, t = 4.26 (two-sample t test comparing mean increase in photon flux with group sizes of n = 10). Each symbol represents an individual animal. Red bar, median value for each group. B, ex vivo imaging of excised brains 21 d after tumor cell implantation illustrates enhanced overall tumor burden and intracranial spreading of BCM2 BrainG2 cells compared with BCM2 Bone cells. C, metastatic extension from the site of implantation in the brain to the spine was seen only in mice injected with BCM2 BrainG2 cells. Two representative mice are shown from each group comparing BCM2 BrainG2 to BCM2 Bone.

Close modal

An important question arising from this and other studies is whether tumor cell clones with a potential for seeding brain metastases already exist within certain primary tumors, or if the development of brain metastases is primarily based on the evolution of a tumor cell phenotype that supports brain colonization and expansion within the unique microenvironment of the brain. A number of studies suggest that gene expression signatures, indicating a metastatic potential or poor prognosis, can exist within primary tumors and be preserved throughout metastatic progression (4549). Many of those genes are directly involved in cell proliferation, indicate the epithelial cell type from which the primary tumor derived, or contribute to overall cell motility and invasion. In addition, expression profiles associated with organ-specific breast cancer metastasis to bone or lung have been reported and comprise genes that are likely critical for tumor cell entry into those target organ microenvironments (50, 51). Notably, these and other studies also show that gene expression pattern, indicative of metastatic potential and target organ tropism, can be preserved after in vivo selected tumor cells are expanded in tissue culture (52).

Our study is based on originally circulating breast cancer cells that represent an advanced state of disease progression, as the cells were derived from a patient with active metastatic disease, years after the primary tumor had been removed. Strikingly, the majority of proteins, which we found differentially expressed in the brain metastatic variant of the originally circulating tumor cells, is involved in cell metabolism and may reflect either a predisposition or an adaptation of the tumor cells to the specific conditions in the brain tissue. In this context, it is important to emphasize that brain metastases have an exceptional long latency, with a median latent interval of 2 to 3 years after initial breast cancer diagnosis (53). Metastases to bone and other target organs generally appear much earlier and also occur with a significantly higher frequency (54). Our data are compatible with the concept that a subset of tumor cells evolves during malignant progression, perhaps from a preexisting cancer cell population, which is capable of gaining entry to the protected environment of the central nervous system. Importantly, these tumor cells can apparently differentiate along pathways that allow them to adapt and ultimately thrive in the brain microenvironment in response to signals from the brain tissue.

Grant support: NIH training grant T32 HL 07695 and later by the NIAID sub-contract grant UCSD/MCB0237059 (E.I. Chen); grant U19 AI063603-02 (J. Hewel); Susan G. Komen grant PDF0403205 (J.S. Krueger); the Deutsche Forschungsgemeinschaft (BE 1540-9/1; to K. Becker). Additional support came from NIH grant P41RR11823 (J.R. Yates III); NCI grants CA095458 and CA112287, and CBCRP grants 10YB-020 and 11IB-0077 (B. Felding-Habermann); NIH grant DK064951 (A. Kralli); and the Sam and Rose Stein Endowment Fund.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

The lentiviral expression system for breast cancer cell tagging with Firefly luciferase was developed and provided by Drs. Bruce Torbett and Mario Tschan of TSRI and the statistical analyses were supported by Dr. Jim Koziol of TSRI.

1
Kirsch DG, Loeffler JS. Brain metastases in patients with breast cancer: new horizons.
Clin Breast Cancer
2005
;
6
:
115
–24.
2
Weil RJ, Palmieri DC, Bronder JL, Stark AM, Steeg PS. Breast cancer metastasis to the central nervous system.
Am J Pathol
2005
;
167
:
913
–20.
3
Klos KJ, O'Neill BP. Brain metastases.
Neurologist
2004
;
10
:
31
–46.
4
Lassman AB, DeAngelis LM. Brain metastases.
Neurol Clin
2003
;
21
:
1
–23, vii.
5
Chang EL, Lo S. Diagnosis and management of central nervous system metastases from breast cancer.
Oncologist
2003
;
8
:
398
–410.
6
Fidler IJ. The organ microenvironment and cancer metastasis.
Differentiation
2002
;
70
:
498
–505.
7
Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease.
Nat Med
1995
;
1
:
27
–31.
8
Hanahan D, Weinberg RA. The hallmarks of cancer.
Cell
2000
;
100
:
57
–70.
9
Ohno S, Ishida M, Kataoka A, Murakami S. Brain metastasis of breast cancer.
Breast Cancer
2004
;
11
:
27
–9.
10
Washburn MP, Wolters D, Yates JR III. Large-scale analysis of the yeast proteome by multidimensional protein identification technology.
Nat Biotechnol
2001
;
19
:
242
–7.
11
Raichle ME, Mintun MA. Brain work and brain imaging.
Annu Rev Neurosci
2006
;
29
:
449
–76.
12
Magistretti PJ, Pellerin L, Rothman DL, Shulman RG. Energy on demand.
Science
1999
;
283
:
496
–7.
13
Garcia-Espinosa MA, Rodrigues TB, Sierra A, et al. Cerebral glucose metabolism and the glutamine cycle as detected by in vivo and in vitro13C NMR spectroscopy.
Neurochem Int
2004
;
45
:
297
–303.
14
Rao J, Oz G, Seaquist ER. Regulation of cerebral glucose metabolism.
Minerva Endocrinol
2006
;
31
:
149
–58.
15
Lubman DM, Kachman MT, Wang H, et al. Two-dimensional liquid separations-mass mapping of proteins from human cancer cell lysates.
J Chromatogr B Analyt Technol Biomed Life Sci
2002
;
782
:
183
–96.
16
Chen EI, Florens L, Axelrod FT, et al. Maspin alters the carcinoma proteome.
FASEB J
2005
;
19
:
1123
–4.
17
Yates JR III, Eng JK, McCormack AL, Schieltz D. Method to correlate tandem mass spectra of modified peptides to amino acid sequences in the protein database.
Anal Chem
1995
;
67
:
1426
–36.
18
Tabb DL, McDonald WH, Yates JR III. DTASelect and Contrast: tools for assembling and comparing protein identifications from shotgun proteomics.
J Proteome Res
2002
;
1
:
21
–6.
19
Liu H, Sadygov RG, Yates JR III. A model for random sampling and estimation of relative protein abundance in shotgun proteomics.
Anal Chem
2004
;
76
:
4193
–201.
20
Becker K, Gui M, Traxler A, Kirsten C, Schirmer RH. Redox processes in malaria and other parasitic diseases. Determination of intracellular glutathione.
Histochemistry
1994
;
102
:
389
–95.
21
Nordhoff A, Bucheler US, Werner D, Schirmer RH. Folding of the four domains and dimerization are impaired by the Gly446→Glu exchange in human glutathione reductase. Implications for the design of antiparasitic drugs.
Biochemistry
1993
;
32
:
4060
–6.
22
Rolli M, Fransvea E, Pilch J, Saven A, Felding-Habermann B. Activated integrin αvβ3 cooperates with metalloproteinase MMP-9 in regulating migration of metastatic breast cancer cells.
Proc Natl Acad Sci U S A
2003
;
100
:
9482
–7.
23
Lin J, Handschin C, Spiegelman BM. Metabolic control through the PGC-1 family of transcription coactivators.
Cell Metab
2005
;
1
:
361
–70.
24
Schreiber SN, Emter R, Hock MB, et al. The estrogen-related receptor α (ERRα) functions in PPARγ coactivator 1α (PGC-1α)-induced mitochondrial biogenesis.
Proc Natl Acad Sci U S A
2004
;
101
:
6472
–7.
25
Vega RB, Huss JM, Kelly DP. The coactivator PGC-1 cooperates with peroxisome proliferator-activated receptor α in transcriptional control of nuclear genes encoding mitochondrial fatty acid oxidation enzymes.
Mol Cell Biol
2000
;
20
:
1868
–76.
26
Huss JM, Torra IP, Staels B, Giguere V, Kelly DP. Estrogen-related receptor α directs peroxisome proliferator-activated receptor α signaling in the transcriptional control of energy metabolism in cardiac and skeletal muscle.
Mol Cell Biol
2004
;
24
:
9079
–91.
27
Zong H, Ren JM, Young LH, et al. AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivation.
Proc Natl Acad Sci U S A
2002
;
99
:
15983
–7.
28
Carling D. The AMP-activated protein kinase cascade-a unifying system for energy control.
Trends Biochem Sci
2004
;
29
:
18
–24.
29
Xue B, Kahn BB. AMPK integrates nutrient and hormonal signals to regulate food intake and energy balance through effects in the hypothalamus and peripheral tissues.
J Physiol
2006
;
574
:
73
–83.
30
Fryer LG, Carling D. AMP-activated protein kinase and the metabolic syndrome.
Biochem Soc Trans
2005
;
33
:
362
–6.
31
Luo Z, Saha AK, Xiang X, Ruderman NB. AMPK, the metabolic syndrome and cancer.
Trends Pharmacol Sci
2005
;
26
:
69
–76.
32
Neumann D, Schlattner U, Wallimann T. A molecular approach to the concerted action of kinases involved in energy homoeostasis.
Biochem Soc Trans
2003
;
31
:
169
–74.
33
Horecker BL. Alternative pathways of carbohydrate metabolism in relation to evolutionary development.
Comp Biochem Physiol
1962
;
4
:
363
–9.
34
Holmgren A. Thioredoxin and glutaredoxin systems.
J Biol Chem
1989
;
264
:
13963
–6.
35
Warburg O. On the origin of cancer cells.
Science
1956
;
123
:
309
–14.
36
Weber G. Enzymology of cancer cells (first of two parts).
N Engl J Med
1977
;
296
:
486
–92.
37
Weber G. Enzymology of cancer cells (second of two parts).
N Engl J Med
1977
;
296
:
541
–51.
38
Lee YJ, Galoforo SS, Berns CM, et al. Glucose deprivation-induced cytotoxicity in drug resistant human breast carcinoma MCF-7/ADR cells: role of c-myc and bcl-2 in apoptotic cell death.
J Cell Sci
1997
;
110
:
681
–6.
39
Laszlo J, Humphreys SR, Goldin A. Effects of glucose analogues (2-deoxy-d-glucose, 2-deoxy-d-galactose) on experimental tumors.
J Natl Cancer Inst
1960
;
24
:
267
–81.
40
Lin X, Zhang F, Bradbury CM, et al. 2-Deoxy-d-glucose-induced cytotoxicity and radiosensitization in tumor cells is mediated via disruptions in thiol metabolism.
Cancer Res
2003
;
63
:
3413
–7.
41
Blackburn RV, Spitz DR, Liu X, et al. Metabolic oxidative stress activates signal transduction and gene expression during glucose deprivation in human tumor cells.
Free Radic Biol Med
1999
;
26
:
419
–30.
42
Spitz DR, Sim JE, Ridnour LA, Galoforo SS, Lee YJ. Glucose deprivation-induced oxidative stress in human tumor cells. A fundamental defect in metabolism?
Ann N Y Acad Sci
2000
;
899
:
349
–62.
43
Miller J, Gordon C. The regulation of proteasome degradation by multi-ubiquitin chain binding proteins.
FEBS Lett
2005
;
579
:
3224
–30.
44
Ding Q, Dimayuga E, Keller JN. Proteasome regulation of oxidative stress in aging and age-related diseases of the CNS.
Antioxid Redox Signal
2006
;
8
:
163
–72.
45
Weigelt B, Glas AM, Wessels LF, et al. Gene expression profiles of primary breast tumors maintained in distant metastases.
Proc Natl Acad Sci U S A
2003
;
100
:
15901
–5.
46
Weigelt B, Hu Z, He X, et al. Molecular portraits and 70-gene prognosis signature are preserved throughout the metastatic process of breast cancer.
Cancer Res
2005
;
65
:
9155
–8.
47
Sorlie T, Tibshirani R, Parker J, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets.
Proc Natl Acad Sci U S A
2003
;
100
:
8418
–23.
48
Sorlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications.
Proc Natl Acad Sci U S A
2001
;
98
:
10869
–74.
49
Van't Veer LJ, Dai H, van de Vijver MJ, et al. Gene expression profiling predicts clinical outcome of breast cancer.
Nature
2002
;
415
:
530
–6.
50
Minn AJ, Kang Y, Serganova I, et al. Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors.
J Clin Invest
2005
;
115
:
44
–55.
51
Montel V, Huang TY, Mose E, Pestonjamasp K, Tarin D. Expression profiling of primary tumors and matched lymphatic and lung metastases in a xenogeneic breast cancer model.
Am J Pathol
2005
;
166
:
1565
–79.
52
Ross DT, Scherf U, Eisen MB, et al. Systematic variation in gene expression patterns in human cancer cell lines.
Nat Genet
2000
;
24
:
227
–35.
53
Bendell JC, Domchek SM, Burstein HJ, et al. Central nervous system metastases in women who receive trastuzumab-based therapy for metastatic breast carcinoma.
Cancer
2003
;
97
:
2972
–7.
54
Pantel K, Muller V, Auer M, et al. Detection and clinical implications of early systemic tumor cell dissemination in breast cancer.
Clin Cancer Res
2003
;
9
:
6326
–34.