[5] Zhang et all Cell

download [5] Zhang et all  Cell

of 14

Transcript of [5] Zhang et all Cell

  • 8/9/2019 [5] Zhang et all Cell

    1/14

    Glycine Decarboxylase Activity Drives

    Non-Small Cell Lung CancerTumor-Initiating Cells and TumorigenesisWen Cai Zhang,1,3 Ng Shyh-Chang,1 He Yang,5 Amit Rai,6 Shivshankar Umashankar,6,7 Siming Ma,1 Boon Seng Soh,1

    Li Li Sun,1 Bee Choo Tai,11 Min En Nga,9 Kishore Kumar Bhakoo,12 Senthil Raja Jayapal,13 Massimo Nichane,1 Qiang Yu,2

    Dokeu A. Ahmed,4 Christie Tan,4 Wong Poo Sing,10 John Tam,10Agasthian Thirugananam,14 Monireh Soroush Noghabi,1

    Yin Huei Pang,9 Haw Siang Ang,5 Wayne Mitchell,16,17 Paul Robson,1 Philipp Kaldis,13 Ross Andrew Soo,5,8

    Sanjay Swarup,6,7 Elaine Hsuen Lim,3,8,15,*and Bing Lim1,18,*1Stem Cell and Developmental Biology2Cancer Biology and Pharmacology

    Genome Institute of Singapore, 60 Biopolis Street, Singapore 1386723Department of Respiratory Medicine4Department of Thoracic Surgery

    Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore 3084335Cancer Science Institute of Singapore, National University of Singapore, 28 Medical Drive, Singapore 1174566Metabolites Biology Lab, Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 1175437NUS Environmental Research Institute (NERI), T-Lab Building (TL), National University of Singapore, 5A Engineering Drive 1, Singapore 1174118Department of Haematology-Oncology9Department of Pathology10Department of Thoracic Surgery

    National University Hospital, 5 Lower Kent Ridge Road, Singapore 11907411Department of Community, Occupational and Family Medicine, Yong Loo Lin School of Medicine, National University of Singapore,

    Singapore 11759712Translational Molecular Imaging Group (TMIG), Laboratory of Molecular Imaging (LMI), Singapore BioImaging Consortium (SBIC),

    11 Biopolis Way, Singapore 13866713Cell Division and Cancer Laboratory (PRK), Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 13867314Department of Thoracic Surgery15Department of Medical Oncology

    National Cancer Centre, 11 Hospital Drive, Singapore 169610

    16Bioinformatics, Experimental Therapeutics Centre, 31 Biopolis Way, Singapore 13866917Divisionof InformationSciences, Schoolof Computer Engineering, NanyangTechnological University, 50 NanyangAvenue, Singapore 63979818Beth-Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA

    *Correspondence: [email protected](E.H.L.),[email protected](B.L.)

    DOI10.1016/j.cell.2011.11.050

    SUMMARY

    Identification of the factors critical to the tumor-initi-

    ating cell (TIC) state mayopen new avenues in cancer

    therapy. Here we show that the metabolic enzyme

    glycine decarboxylase (GLDC) is critical for TICs in

    non-small cell lung cancer (NSCLC). TICs from pri-mary NSCLC tumors express high levels of the onco-

    genic stem cell factor LIN28B and GLDC, which are

    both required for TIC growth and tumorigenesis.

    Overexpression of GLDC and other glycine/serine

    enzymes, but not catalytically inactive GLDC, pro-

    motes cellular transformation and tumorigenesis.

    We found that GLDC induces dramatic changes in

    glycolysis and glycine/serine metabolism, leading

    to changes in pyrimidine metabolism to regulate

    cancer cell proliferation. In the clinic, aberrant activa-

    tion of GLDC correlates with poorer survival in lung

    cancer patients, and aberrant GLDC expression is

    observed in multiple cancer types. This link between

    glycine metabolism and tumorigenesis may provide

    novel targets for advancing anticancer therapy.

    INTRODUCTION

    Despite numerous advances in our knowledge of cancer (Vogel-

    stein and Kinzler, 2004; Hanahan and Weinberg, 2011), our

    ability to develop clinically effective therapies based on this

    understanding has met with limited success. Current therapies

    can control tumor growth initially, but most patients ultimately

    relapse. One prominent example is lung cancer, the leading

    cause of cancer-related mortality with over 1 million deaths

    each year (Jemal et al., 2011). Non-small cell lung cancer

    (NSCLC) accounts for approximately 85% of all lung cancers.

    Although NSCLC patients with epidermal growth factor receptor

    (EGFR) mutations respond to EGFR inhibitors initially, most

    patients experience a relapse within 1 year (Sequist et al.,

    Cell148, 259272, January 20, 2012 2012 Elsevier Inc. 259

    mailto:[email protected]:[email protected]://dx.doi.org/10.1016/j.cell.2011.11.050http://dx.doi.org/10.1016/j.cell.2011.11.050mailto:[email protected]:[email protected]
  • 8/9/2019 [5] Zhang et all Cell

    2/14

    2007). These findings underscore the urgent need for both com-

    bination therapies and also new approaches to treat cancerous

    tumors. One such approach may be to target tumor-initiating

    cells (TICs).

    Data from leukemias, germ cell tumors, and a number of solidtumors support the notion that cancers are maintained by

    a subpopulation of self-renewing and evolving TICs. This is

    also popularly known as the cancer stem cell (CSC) model

    (Reya et al., 2001; Rosenand Jordan, 2009). Although the validity

    of the CSC model is an issue of controversy in melanoma (Quin-

    tana et al., 2008, 2010; Boiko et al., 2010; Civenni et al., 2011),

    many other solid tumors appear to follow the CSC model (Ishi-

    zawa et al., 2010). Recently it was proposed that at earlier stages

    of tumorigenesis, rare TIC clones differentiate into nonmalignant

    progeny to form the bulk of the tumor, whereas at advanced

    stages, TIC clones constitute the bulk of the tumor (Boiko

    et al., 2010). Studies with mouse models of lung cancer have

    also begun to reconcile the connection between the evolving

    genotype of TIC clones and the surface phenotype of TICs (Cur-tis et al., 2010). Thus accumulated findings suggest that target-

    ing TICs may be a promising approach for eradicating tumors

    early. However, progress in the targeting of TICs to improve

    cancer therapy has been hindered by a lack of understanding

    of the molecular pathways that are critical to TICs.

    Recent studies have led to an emerging appreciation of the

    importance of metabolic reprogramming in cancer (Hsu and Sa-

    batini, 2008; Vander Heiden et al., 2009; Hanahan andWeinberg,

    2011). Most recently, the embryonic isoform of pyruvate kinase

    PKM2, in collaboration with phosphoglycerate mutase, was

    found to regulate the shift from oxidative phosphoryla-

    tion to glycolysis in cancer cells (Christofk et al., 2008; Vander

    Heiden et al., 2010). These findings have led to a resurgence of

    interest in the Warburg effectthe phenomenon whereby cancer

    cells, like embryonic cells, preferentially use glycolysis even

    under aerobic conditions (Warburg, 1956). Besides glycolysis,

    an arm of metabolism that results in sarcosine production has

    also been implicated in prostate cancer (Sreekumar et al.,

    2009). These data suggest that metabolic reprogramming is

    crucial for tumorigenesis, and much remains to be uncovered.

    Here we show that glycine metabolism and the metabolic

    enzyme glycine decarboxylase (GLDC) drive TICs and tumori-

    genesis in NSCLC. Using CD166 as a surface marker and

    NOD/SCID Il2rg/ mice as xenotransplantation recipients, we

    isolated lung TICs from a broad range of primary NSCLC tumors

    (stages IIII). Primary lung TICs express high levels of LIN28B,

    GLDC, and many other glycine/serine metabolism enzymes.Both LIN28B and GLDC were required for lung TIC proliferation

    and tumor growth. Overexpression of GLDC alone, and other

    glycine/serine enzymes, promotes cellular transformation both

    in vitro andin vivo. Metabolomic analysis shows that GLDC over-

    expression induces dramatic changes in glycolysis and glycine

    metabolism, leading to changes in pyrimidine metabolism for

    cancer cell proliferation. In human patients, aberrant upregula-

    tion of GLDC is significantly associated with higher mortality

    from lung cancer, and aberrant GLDC expression is observed

    in multiple cancer types. Our findings establish a link between

    glycine metabolism and tumorigenesis and may provide novel

    targets for advancing anticancer therapy.

    RESULTS

    TICs in Lung Cancer

    To assess the cellular heterogeneity within NSCLC, we obtained

    freshly resected lung tumors from 36 human patients with abroad range of stage IIII primary NSCLC (Table S1 available

    online). Patient lung cancer cells were directly transplanted

    subcutaneously into NOD/SCID Il2rg/ mice with Matrigel

    (Quintana et al., 2008). Using this maximally sensitive assay,

    we estimated by limiting dilution analysis that lung TICs exist

    with a low frequency of 1 in 4 3 105 cells in unsorted NSCLC

    tumor cells(n = 36 patients; Figure1A), consistent with published

    findings (Ishizawa et al., 2010).

    To profile the surface phenotype of this subpopulation of lung

    TICs, we fractionated the NSCLC tumors by fluorescence-

    activated cell sorting (FACS; Figure S1A). After excluding

    hematopoietic and endothelial cells (Lin), we tested a panel of

    cell-surface markers, including CD166, CD44, CD133, and

    EpCAM (Figure 1B). We found that CD166 was the most robustmarker for enriching the lung TIC subpopulation, compared to

    CD133, CD44, or EpCAM, allowing us to reliably enrich lung

    TICs by nearly 100-fold (Figures 1A and 1B). In 12 out of 12

    NSCLC patient tumors (lung adenocarcinoma), the CD166+

    Lin fraction contained cells that consistently initiated lung tumor

    formation in vivo. In contrast, CD166Lin tumor cells generally

    failed to initiate lung tumor formation even after 8 months of

    observation, although they also expressed carcinoembryonic

    antigen (CEA), a tumor-specific marker not expressed in normal

    adult lung cells (Figures 1A, 1B, andS1B). Similar results were

    observed in lung squamous cell carcinoma and large cell carci-

    noma (Figure S1C). Although CD166 expression varied across

    the NSCLC tumors we examined, CD166 was consistently

    higher in lung tumors than in normal adjacent lung tissues

    (n = 25 patients;Figures S1D and S1E).

    CD166+ lung TICs demonstrate a capacity for self-renewal

    and differentiation in vivo. Serial transplantations showed that

    only the CD166+ fraction was able to self-renew and initiate pri-

    mary and secondary xenograft tumors (Figures 1A and S1F).

    Upon transplantation, CD166+ lung TICs differentiated to form

    xenograft tumors that phenocopy the complex cytoarchitec-

    ture of their parental patient tumors, sharing similar histolog-

    ical morphology by hematoxylin-eosin (H&E) staining and

    similar tissue distributions of CD166, cytokeratin, E-cadherin,

    vimentin, smooth muscle actin, and synaptophysin (Figures

    1C, S1G, and S1H). Furthermore, we found that transplants

    with more TICs grow more rapidly, suggesting that lung TICfrequency is correlated with tumor growth rate (Figures 1D

    andS1I).

    The self-renewal capacity of CD166+ lung TICs is further

    corroborated by in vitro assays. We tested the CD166+ fraction

    for the ability to form tumor spheres, a widely used in vitro tech-

    nique for assessing self-renewal capacity (Dontu et al., 2003).

    Although both primary CD166+ and CD166 cells remained

    viable in vitro, only primary CD166+ but not CD166 cells were

    able to form compact self-renewing spheres (n = 9 patients;

    Figures 1E, 1F, andS1J). Using immunofluorescence and flow

    cytometry, we found that the lung tumor spheres retained high

    levels of CD166 expression but undetectable CD133 expression

    260 Cell148, 259272, January 20, 2012 2012 Elsevier Inc.

  • 8/9/2019 [5] Zhang et all Cell

    3/14

    in contrast (Figures S1K and S1L). When primary lung tumor

    spheres were dissociated into single cells and transplanted

    into NOD/SCID Il2rg/ mice in vivo, we found that as few as

    15 single cells consistently initiated tumorigenesis (Figures 1G

    andS1M).

    The increased tumor-initiating frequency of lung tumor sphere

    cells suggests that they are even more highly enriched for lung

    TICs than the patient tumor CD166+ fraction, and that lung

    TICs expanded during in vitro culture to form tumor spheres.

    To test whether CD166 drives tumorigenicity in lung TICs, we

    A

    CellsNo. cells per

    injection

    No. tumors

    / No.

    injections

    LTICs

    frequency

    (95% CI)

    Enrichment

    factor (x)Pvalue

    150,000 2/4

    C

    H&E

    Patient tumor Xenograft tumor

    Unsorted

    1/403,971

    (1/255,949 -

    1/637,598)

    1

    200,000 1/10

    300,000 4/5

    500,000 3/5

    1,000,000 9/9

    2,000,000 2/2

    CD166+Lin-1/5,175

    (1/2,837 -

    1/9,441)

    78 1.37E-23

    1,000 2/8

    5,000 2/3

    10,000 7/9

    23,000 2/2

    25 000 4/4

    CD166

    PopulationNo. tumors/

    No. injections

    CD166+ 12/12

    B

    ,

    50,000 1/1

    CD166-Lin-1/3,297,091

    (1/460,464 -

    1/23,608,384)

    0.1 4.82E-03210,000 0/1

    260,000 1/3

    500,000 0/3

    400

    mm3)

    CD166+Lin- (1st xenograft; 5,000)

    CD166+Lin- (patient; 5,000)

    Lin- (patient; 150,000)

    D

    pan-CK

    CD166- 0/10

    CD44+ 6/8

    CD44- 6/8

    CD133+ 6/11

    CD133- 7/13

    EpCAM+ 10/13

    EpCAM- 8/14 0

    100

    200

    300

    14 15 16 17 18 19 20 21

    Tumorvolume

    Weeks

    CD166-L n- (pat ent; 100,000)

    AdC

    F G

    0

    30

    60

    90

    120

    No.ofspheres/10,0

    00cells

    AdC SCC LCC

    E

    D166-Lin-

    SCCAdC LCC

    CD166+Lin-

    No.

    Cells

    No. tumors/ No. injections

    1st passage 10th passage

    2500 12/12 N.D.

    500 18/18 18/18

    50 24/24 24/24

    10 12/12 12/12

    5 16/18 17/18

    1 5/18 5/18

    GTumor sphere assay

    CD

    Figure 1. CD166+ Fraction Contains TICs from NSCLC Patients

    (A)Frequency of TICs in unsorted,CD166+, andCD166 subpopulationsof cells from36 NSCLCpatients. CI, confidence interval. Lin cells, CD45 CD31 cells.

    (B) Tumor initiation frequency by various FACS-purified fractions of Lin lung cancer cells isolated from primary xenograft tumors shown in (A). 5 3 104

    CD166,

    CD44, CD133, and EpCAM cells were tested for tumor initiation in NOD/SCID Il2rg/ mice (n = 12).

    (C) Histological analysis of patient tumors and primary xenografts derived from patient tumor CD166+

    Lin lung cancer cells. Tumors were stained for H&E,

    CD166, and pan-CK (cytokeratin). Scale bar, 50 mm.

    (D) Representative tumor-growth curves of xenografts derived from different cell fractions in a lung adenocarcinoma (AdC) patient tumor and the primaryxenograft.

    (E) Phase-contrast images of tumor spheres seeded with CD166+ Lin (top) and CD166 Lin (bottom) cells in lung adenocarcinoma (AdC), squamous cell

    carcinoma (SCC), and large cell carcinoma (LCC). Scale bar, 50 mm.

    (F) Quantification of tumor spheres formed by cells from NSCLC patient (AdC, SCC, or LCC) CD166+Lin, CD166Lin, and Lin populations.

    (G) Frequency of tumorigenesis by single patient-derived tumor sphere cells (n = 3). N.D., not determined.

    In all panels, error bars represent the standard error of the mean (SEM). See alsoFigure S1andTable S1.

    Cell148, 259272, January 20, 2012 2012 Elsevier Inc. 261

  • 8/9/2019 [5] Zhang et all Cell

    4/14

    knocked down CD166 in two lines of NSCLC patient-derived

    tumor spheres by retroviral shRNA (Figure S1N). We found that

    the tumorigenicity of lung TICs in the tumor spheres was not

    significantly affected by CD166 shRNA, demonstrating that

    CD166 is an inert cell-surface marker that enriches for lung

    TICs (Figures S1OS1Q).

    Lung TICs Express High levels of Glycine/Serine

    Metabolism Enzymes

    To gain a deeper understanding of the molecular basis for the

    TIC state and its tumorigenic capacity, we sought to obtain

    a molecular signature for lung TICs. To do this, we performed

    genome-wide transcriptome analysis on CD166 Lin tumor

    cells, CD166+ Lin tumor cells, and lung tumor spheres, in

    increasing order of lung TIC frequency (Figure 2A). As a negative

    control, we also profiled CD166+ versus CD166 cells from

    normal adjacent lung tissues (n = 3 patients; Table S1). This

    led us to a profile of genes that are upregulated and down-

    regulated in lung TICs, compared to non-TICs (Figure 2B).

    Lung TIC-associated genes include the oncogenic stem cell

    factor LIN28B, embryonic lung transcription factors like PEA3

    and thetrachealesshomologNPAS1 (Viswanathan et al., 2009;

    4

    o

    4

    BA

    Enrichment

    S/X-

    X+/X-

    P+/P-

    N+/N-

    Log2ratio

    0

    -4N+/X- P+/X- S/X-X+/X-

    Log2rati

    0

    -4N+/X- P+/X- S/X-X+/X_

    X+ S

    Candidate

    genes

    S: sphere; X, xenograft tumor; P, primary tumor;

    N, normal tissue; +, CD166+; -, CD166-.

    1

    2

    3

    4

    onlevel(log2ratio)

    S/X+ X+/X-

    P+/P- N+/N-

    HSPA1A

    IGLL1

    RENB

    P

    PPP1R

    15A

    RASD

    1

    MMP9

    RRAD

    CXCL

    2

    GADD

    45G

    TNFAIP3

    C D

    0% 2% 4% 6% 8% 10%

    Cell cycle

    DNA replication

    Percentage of genes in pathway

    -5

    -4

    -3

    -2

    -1

    RelativemRNAexpressio

    GLDC

    NPAS1

    LIN28B

    RG9MTD2

    CCNB1

    TMEM118

    SMC6

    PEA3

    REEP1

    FOXK2

    Glycine, serine and threonine

    metabolism

    Pyrimidine metabolism

    MAPK signaling pathway

    p53 signaling pathwayExperimental

    Predicted

    E

    PSPHGlycine CO2 + NH3 + CH2Phosphoserine Serine

    GLDC

    L-2-Amino-acetoacetate

    GCAT

    SHMT13P-Hydroxy pyruvate

    PSAT1

    Figure 2. Lung TICs Express High Levels of GLDC and LIN28B

    (A) Venndiagramshowingstrategy for enriching tumorigenic geneexpressionprofile by genome-wide transcriptomeanalysis.A listof thedifferentially expressed

    genes(cutoff thresholdof 1.5-fold,p < 0.05)common between P+ versusP, X+ versus X, and S versusXwasderived, withthe differentially expressed genesof

    N+

    versus N excluded. Thegenelistwas further filtered to select only genesfurtherupregulatedor downregulated in S versus X+

    . N,normal lungtissue (n= 3); P,

    patient tumor (n = 1); S, tumor sphere (n = 4); X, xenograft tumor (n = 3); +, CD166+

    ; , CD166. Total, n = 11.

    (B) Graphs of relativeexpression of candidatelung TIC-associated genes in increasing (left, n = 194) or decreasing (right, n = 295) trends across differentCD166+

    fraction cells from normal lung tissue (N), primary tumor (P), xenograft tumor (X), and tumor spheres (S) versus nontumorigenic CD166 (X) cells.

    (C) Top-ranked genes differentially expressed in lung TICs.

    (D) Enrichment of KEGG pathways by genes differentially expressed in lung TICs. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

    (E) Schematic diagram of glycine, serine, and threonine metabolism genes significantly (p < 0.05) upregulated in lung TICs. [, upregulation.

    See alsoFigure S2.

    262 Cell148, 259272, January 20, 2012 2012 Elsevier Inc.

  • 8/9/2019 [5] Zhang et all Cell

    5/14

    Liu et al., 2003; Levesque et al., 2007), as well as cell-cycle regu-

    lators like CCNB1 and GADD45G (Figure 2C). The highest-

    ranking genes were validated by qRT-PCR (Figure S2A). KEGG

    pathway analysis of the lung TIC-gene profile showed that the

    top enriched pathways were cell cycle, DNA replication,glycine, serine, and threonine metabolism, pyrimidine metab-

    olism, MAPK signaling pathway, and p53 signaling path-

    way (Figure 2D). Within the glycine, serine, and threonine

    metabolism pathway, we found that glycine/serine metabolism

    enzymes likeGLDC, glycine C-acetyltransferase (GCAT), serine

    hydroxymethyltransferase (SHMT1), phosphoserine phospha-

    tase (PSPH), and phosphoserine aminotransferase (PSAT1)

    were all upregulated in lung TICs (Figures 2E andS2BS2D). In

    particular,GLDCwas one of the most highly upregulated genes

    in multiple analyses of lung TIC-enriched populations, at both the

    mRNA and protein levels (Figures 2C andS2C). GLDC is a key

    component of the highly conserved glycine cleavage system

    in amino acid metabolism that catalyzes the breakdown of

    glycine to form CO2, NH3, and 5,10-methylene-tetrahydrofolate(CH2-THF) to fuel one-carbon metabolism (Kume et al., 1991).

    GLDC Is an Oncogene that Promotes Tumorigenesis

    and Cellular Transformation

    High expression ofGLDCandLIN28Bin lung TIC-enriched pop-

    ulations,but not in CD166 lung cancer cells and CD166+ normal

    lung cells, suggests that these two genes drive tumorigenicity in

    lung TICs. To test this hypothesis, we knocked down GLDCand

    LIN28B in lung tumor spheres with shRNAs (Figure S3A) and

    compared their growth both in vitro and in vivo. We found that

    both GLDC and LIN28B were necessary for cell proliferation in

    sphere culture, as well as anchorage-independent colony forma-

    tionin softagar (Figures 3A and S3B). Importantly, tumorigenicity

    was also significantly reduced upon knockdown of either GLDC

    or LIN28B (Figures 3B and S3C). A549 lung adenocarcinoma

    cells showed similar results (Figures S3DS3G). Our results

    suggest that lung TICs and lung tumorigenesis are dependent

    on GLDC. This led us to ask what oncogenes upregulate

    GLDC. Because the E2F pathway upregulates many metabolic

    genes during cell proliferation, we examined the expression of

    GLDC over the course of the cell cycle in both normal human

    lung fibroblasts (HLFs) and the transformed A549 cells after

    synchronization by serum starvation. Our results showed that

    GLDC is insensitive to cell-cycle progression in both normal

    HLFs and transformed A549 cells, suggesting that GLDC is not

    regulated by cell-cycle or E2F signals (Figure 3C). We then

    examinedGLDClevels in MCF10A cells after transformation byoncogenic KRASG12D, PIK3CAE545K, and MYCT58A. Our results

    show that all three oncogenes induceGLDC by 20-fold, sug-

    gesting that oncogene-induced GLDC transcription is common

    to the cellular transformation process mediated by oncogenic

    Ras, PI3K, and Myc (Figure 3D).

    To test whether aberrant GLDC upregulation is sufficient

    to drive cellular transformation, as has been shown for LIN28B

    (Viswanathan et al., 2009), we overexpressed GLDC in NIH/

    3T3 cells (Figure S3H). We found that GLDC overexpression

    significantly increased colony formation by 3T3 cells under nor-

    mal culture conditions (Figures 3E and 3F). To test for cellular

    transformation in vitro, we cultured the 3T3 cells overexpressing

    GLDC under anchorage-independent conditions and found that

    GLDCtransforms 3T3 cells readily with a rate exceeding that of

    LIN28B (Figures 3G and S3I). Upon transplantation into NOD/

    SCID Il2rg/ mice, 3T3 cells overexpressingGLDC consistently

    formed tumors in 6/6 transplants, and 3T3 cells overexpressingLIN28B formed tumors in 3/6 transplants, whereas 3T3 cells

    overexpressing the empty control vector never formed tumors

    (Figures S3JS3L).

    To test whetherGLDCcan also transform normal primary HLF

    and normal primary human bronchial epithelial (NHBE) cells, we

    overexpressed GLDC in HLF and NHBE cells (Figures S3M

    andS3O). Both HLFand NHBE cells showeda dramatic increase

    in cell proliferation upon overexpression of GLDC alone (Fig-

    ures 3H3J and S3P). Surprisingly we found that GLDC also

    transforms HLF and NHBE cells readily in vitro (Figures 3K

    and S3Q). However, perhaps because primary adult HLF and

    NHBE cells are not immortalized, GLDC-overexpressing HLF

    and NHBE cells do not form tumors upon transplantation (Fig-

    ures S3N and S3R). In contrast, CD166 lung tumor cells, whichalso could not form tumors in vivo,could now initiate tumorigen-

    esis ata lowfrequency upon overexpression ofGLDC (Figure3L).

    Collectively, our results show thatGLDC is an oncogene that is

    both necessary and sufficient to promote tumorigenesis.

    GLDC Promotes Tumorigenesis through Its Metabolic

    Activity

    Although GLDC is a metabolic enzyme, it remained unclear

    whether GLDC promotes tumorigenesis through a metabolism-

    dependent or -independent mechanism. To address this ques-

    tion, we engineered a series of four point mutations within or

    near the evolutionarily conserved catalytic active site of the

    GLDC enzyme to disrupt its metabolic activity (Figure 4A). These

    point mutations comprised three nonlethal GLDC mutations

    found in human patients with nonketotic hyperglycinemia

    (H753P, P769L, G771R; Figures S4A and S4B) and one mutation

    K754A that is predicted to abrogate the covalent bond with the

    critical pyridoxal-50-phosphate cofactor (Nakai et al., 2005;

    Kure et al., 2006). When we overexpressed these four GLDC

    mutants in 3T3 cells, none of them could lead to tumorigenesis

    in vivo, whereas wild-type GLDC could, even though all of

    them were expressed at high levels similar to that in transformed

    A549 cells (Figure 4B). Thus the metabolic activity of GLDC is

    required for its tumorigenic function.

    In addition, the upregulation of many other upstream enzymes

    in the glycine/serine pathway in lung TICs further supports the

    idea that metabolic activity in the glycine/serine pathway is re-sponsible for promoting tumorigenesis (Figure 2E). To test this

    idea, we also overexpressed PSAT1, PSPH, SHMT1, SHMT2,

    and GCAT in 3T3 cells and transplanted them in vivo to test

    for cellular transformation and tumorigenesis (Figure 4C). By

    2 months, we found that three other glycine/serine enzymes

    PSAT1, PSPH, and SHMT2could also transform 3T3 cells to

    form tumors in vivo (Figure 4D). Interestingly, we noted that

    PSAT1, PSPH, and SHMT2 overexpression only led to a slight

    upregulation of GLDC protein (Figure 4E), suggesting that their

    tumorigenic activity is due to increased glycine/serine metabo-

    lism, rather than indirect upregulation ofGLDC. These findings

    indicate that increasedmetabolism in the glycine/serinepathway

    Cell148, 259272, January 20, 2012 2012 Elsevier Inc. 263

  • 8/9/2019 [5] Zhang et all Cell

    6/14

    Tumor spheres

    120,000 TS-Ctrl-sh

    A B C

    h h h h

    HLF

    Time after release in serum

    hh

    8hc h 4

    h

    8h

    A549

    Time after release in serum

    h0.8

    1

    s(g)

    Ctrl-sh

    GD-sh

    0

    20,000

    40,000

    60,000

    80,000

    100,000

    0 2 4

    Numberofcells

    TS-28B-shTS-GD-sh

    1

    2

    4

    1

    FOS

    CDK1

    GLDC

    0

    2 4c

    HSP90

    CDK1

    HSP90

    GLDC

    8

    244

    0

    0.2

    0.4

    0.6

    Massofxenografttumou

    r 28B-sh

    *

    **

    F

    100

    125

    nies/

    3T3

    3T3-GD3T3-28B

    E

    3T3

    3T3

    D

    MCF10A

    20

    30

    express

    ion

    Days

    G

    60

    70

    80

    5,0

    00

    3T3

    3T3-GD3T3-28B

    0

    25

    50

    75

    No.ofadherentcolon

    5003T3

    ** **3T3-GD

    3T3-28B

    0

    10

    RelativeGLDCe

    0

    10

    20

    30

    40

    50

    60

    No.ofagarcolonies/5

    3T3

    ** **

    60,000

    80,000

    100,000

    erofcells

    HLF-GD

    HLF-28B

    HLF

    H

    25

    30

    35

    nies/5,0

    00

    HLF

    HLF-GD

    HLF-28B

    ** **

    KJ

    100

    120

    140

    colonies/

    LF

    HLF

    HLF-GD

    HLF-28B****

    HLF

    HLF

    HL

    I

    0

    20,000

    40,000

    0 3 6

    Numbe

    Days 0

    5

    10

    15

    20

    No.ofagarcolon

    HLF

    0

    20

    40

    60

    80

    No.ofadherent

    1,0

    00HL

    Xenograft

    LF-GD

    HLF-28B

    Group No. cells /

    injection

    No. tumors/

    No. injections

    CD166-200,000 0/12

    100,000 0/12

    CD166- GD200,000 4/12

    100,000 1/12

    L

    g

    ,

    Figure 3. GLDC and LIN28B Are Necessary and Sufficient for Malignant Growth

    (A) Proliferation curve of tumor sphere (TS) cells with shRNA knockdown of either GLDC (GD-sh) or LIN28B (28B-sh).

    (B) Quantitative mass analysis of xenograft tumors formed 30 days after transplanting 100,000 tumor sphere cells with either GLDC knockdown (GD-sh) or

    LIN28B knockdown (28B-sh).

    (C) Western blot analysis of endogenous GLDC during the cell cycle in synchronized normal human HLFs and transformed A549 cells. HLF or A549 cells were

    serum-starved for 72 hr followed by release into serum-containing medium with samples collected at indicated time points. Expression of GLDC, FOS (early

    264 Cell148, 259272, January 20, 2012 2012 Elsevier Inc.

  • 8/9/2019 [5] Zhang et all Cell

    7/14

    due to GLDC or other glycine/serine enzymes can exert a potent

    tumorigenic effect.

    GLDC Regulates Glycine Metabolism, with Effects on

    Glycolysis and Pyrimidines

    Given that GLDC promotes tumorigenesis through a metabo-

    lism-dependent mechanism, we performed metabolomic anal-

    ysis to gain deeper mechanistic insights into the GLDC-driven

    metabolism changes that lead to tumorigenesis. We used liquid

    chromatography-mass spectrometry (LC-MS) to perform me-

    tabolomics profiling of HLF cells and 3T3 cells overexpressing

    GLDC, as well as A549 lung adenocarcinoma cells with retroviral

    knockdown of GLDC, relative to empty vector controls. We

    found that glycine-related metabolites, glycolysis intermediates,

    serum response), andCDK1 (E2Ftarget) weretested. Normal growing, unsynchronized cells (Cyc)were usedas a control. HSP90was used as a loading control.

    CDK1, cyclin-dependent kinase 1; HSP90, heat shock protein 90.

    (D) Expression of endogenous GLDC in MCF10A cells transformed by oncogenic KRASG12D

    , PIK3CAE545K

    , MYC, or MYCT58A

    , by qRT-PCR.

    (E) Colony formation assay in adherent conditions by seeding 500 3T3 cells overexpressing either GLDC (3T3-GD) or LIN28B (3T3-28B).

    (F) Quantitative analysis of colony formation efficiency under adherent conditions as shown in (E).

    (G) Quantitative analysis of soft agar colony formation by 5,000 3T3 cells overexpressing either GLDC (3T3-GD) or LIN28B (3T3-28B). Colonies were stained with

    INT on day 28.

    (H) Proliferation curve of HLF cells overexpressing GLDC (HLF-GD), LIN28B (HLF-28B), or the empty vector (HLF).

    (I) Colony formation assay in adherent conditions seeding 1,000 HLF cells overexpressing GLDC (HLF-GD), LIN28B (HLF-28B), or the empty vector (HLF).

    (J) Quantitative analysis of colony formation efficiency under adherent conditions as shown in (I).

    (K) Quantitative analysis of soft agar colony formation by 5,000 HLF cells overexpressing either GLDC (HLF-GD), LIN28B (HLF-28B), or the empty vector (HLF).

    (L)Tumorformationby CD166 lung tumor cells 3 months after overexpression of GLDC. CD166 tumor cells from xenografts were sortedby FACS andinfected

    with retrovirus expressing either the empty vector (CD166) or GLDC (CD166 GD), followed by transplantation into mice 24 hr after infection (n = 12 for each

    group).

    In all panels, error bars represent SEM. *p < 0.05, **p < 0.01. See also Figure S3.

    BA

    WT

    H

    753P

    K

    754A

    P

    769L

    G

    771R

    V

    ector

    A

    549

    8/

    8

    0/

    8

    0/

    8

    0/

    8

    0/

    8

    0/

    8

    8/

    8

    No. tumors/

    No. injections

    D E

    Group No. tumors/

    No. injections

    3T3-GLDC 6/6

    3T3-PSAT1 3/6

    3T3-PSPH 2/6

    3T3-SHMT2

    1/63T3-SHMT1 0/6

    3T3-GCAT 0/6

    3T3 0/6

    1

    10

    100

    1,000

    10,000

    100,000

    RelativeRN

    Aexpressionlevel

    C

    -Actin

    GLDC

    GLDC-GFP

    R

    Figure 4. GLDC Promotes Tumorigenesis through Its Metabolic Activity

    (A) Crystal structure of T. thermophilus GLDC near the catalytic active site. The labeled bacterial residues H703b, K704b (purple), P729b, and G731b are

    homologous to H753, K754, P769, and G711 of human GLDC. Residues implicated in human nonketotic hyperglycinemia are shown (red). PLP, pyridoxal-5 0-

    phosphate cofactor (green).

    (B) GLDC protein expression and tumor formation efficiency of 3T3 cells overexpressing wild-type or mutant GLDC. Four point mutations were tested: H753P,

    K754A, P769L, and G771R. Incidence of tumor formation was determined 2 months after injection with 1.5 3 106 cells per mouse (n = 8). A549 cells served as

    a positive control. WT, wild-type.

    (C) Geneexpression in 3T3 cells overexpressing GLDC, PSPH, PSAT1, GCAT, SHMT1, and SHMT2, relativeto 3T3cells with theempty vector,as determined by

    qRT-PCR.

    (D)Tumor formationefficiency of 3T3 cells overexpressing GLDC, PSPH,PSAT1, GCAT, SHMT1, SHMT2, or theempty vector. Incidenceof tumor formation was

    determined 2 months after injection with 1.5 3 106 cells per mouse (n = 6).

    (E) GLDC protein expression of 3T3 cells overexpressingGLDC, PSPH, PSAT1, GCAT, SHMT1, andSHMT2.A549 cells served as a positive control.b-actin was

    used as a loading control.

    In all panels, error bars represent SEM. See also Figure S4.

    Cell148, 259272, January 20, 2012 2012 Elsevier Inc. 265

  • 8/9/2019 [5] Zhang et all Cell

    8/14

    and many pyrimidines were significantly perturbed by both

    GLDC overexpression and knockdown (p < 0.05; Figures

    5A5D). For glycine-related metabolites, we found that sarcosine

    (N-methylglycine) levels increased significantly upon GLDC

    overexpression and dropped significantly upon GLDC knock-down, indicating that GLDC promotes sarcosine synthesis or

    accumulation (Figure 5A). Consistent with this observation,

    betaine aldehyde in the betaine-sarcosine-glycine pathway for

    glycine synthesis also showed the same pattern of changes (Fig-

    ure 5A). Glycine levels decrease withGLDCoverexpression and

    increase withGLDCknockdown, in agreement with the fact that

    GLDC breaks down glycine irreversibly. In contrast, serine levels

    increase with GLDC overexpression and decrease with GLDC

    knockdown, suggesting that GLDC promotes serine synthesis

    or uptake (Figure 5A).

    Surprisingly,GLDCperturbation also led to dramatic changes

    in glycolysis and other amino acids (Figures 5B, 5C, andS5A).

    Our data suggest that GLDC promotes glycolysis, leading

    to the increased synthesis or accumulation of glucose-1-phosphate, phosphoenolpyruvate, pyruvate, and lactate (Fig-

    ures 5B and 5C). In fact many of the upstream glycine/serine

    metabolism enzymes that we found upregulated in lung TICs,

    such as PSAT1, PSPH, and SHMT1/2, channel glycolytic inter-

    mediates into de novo serine and glycine biosynthesis (Fig-

    ure 2E), suggesting that GLDC is working in a concerted fashion

    with these enzymes to promote the glycolysis-serine-glycine

    flux. This is supported by our finding that GLDC does not signif-

    icantly promote glycine uptake (Figures S5B and S5C) but

    promotes glycolysis instead (Figures 5B and 5C).

    Finally, our metabolomics analysis also revealed that GLDC

    promotes the synthesis or accumulation of pyrimidines, in-

    cluding thymidine, deoxyuridine, thymine, uracil, and cytosine

    (Figure 5D). The GLDC-catalyzed reaction converts glycine into

    CH2-THF (Kume et al., 1991). CH2-THF contains the methylene

    group that fuels de novo thymidine synthesis from deoxyuridine

    in concert with pyrimidine biosynthesis and hence nucleotide

    synthesis during cell proliferation (Tibbetts and Appling, 2010).

    Recent studies suggest that early oncogenesis involves aberrant

    activation of cell proliferation, which then leads to a crisis of

    nucleotide deficiency and replication stress (Bester et al., 2011).

    Our observations on pyrimidine synthesis suggest that upregula-

    tion of GLDC could promote cellular transformation by over-

    coming this deficiency to progress onward in early oncogenesis.

    To test whether any of the metabolite changes induced by

    GLDC can mimic GLDCs effects on cancer cells, we analyzed

    whether an increased exogenous supply of specific metabolitescould rescue GLDC retroviral knockdown in A549 cells. We

    found that 10 mM of sarcosine could significantly rescue the

    proliferation defect uponGLDCknockdown, with little effect on

    control A549 cells (Figure 5F), indicating that increased sarco-

    sine-glycine metabolite flux can rescue the effects of reduced

    GLDC enzyme. To further test whether the production of

    CH2-THF is necessary for GLDCs effects on proliferation, we

    tested whether the antifolate drug methotrexate could specifi-

    cally abrogate GLDC-induced proliferation by reducing the tetra-

    hydrofolate (THF) cofactor needed to produce CH2-THF for

    pyrimidine synthesis. Our results show that low doses of metho-

    trexate specifically abrogated GLDC-induced proliferation in 3T3

    and HLF cells, with little effect on control 3T3 and HLF cells

    (Figure 5E). Furthermore, methotrexate in combination with

    GLDC shRNA killed transformed A549 cells much more effec-

    tivelythan eitheralone(Figure5E), suggesting that a combination

    of antifolates with a GLDC inhibitor could completely shut offglycine catabolism to treat cancer cells more effectively. Using

    these metabolic data, we constructed a model of how aberrant

    GLDC expression might reprogram glycolysis and glycine

    metabolism fluxes in cancer cells to promote cancer cell prolifer-

    ation and tumorigenesis (Figure 5G).

    Prognostic Significance of Aberrant GLDC Expression

    in NSCLC Patients

    To assess whether our experimental findings on GLDC are rele-

    vant to human lung cancer patients in the clinic, we used tissue

    microarray immunohistochemistry to examine the prognostic

    significance of GLDC expression, tumor size, tumor grade, and

    cancer stage in clinical tumor samples from cohorts of NSCLC

    patients (n = 143) (Figures 6A andS6;Table S2). Subdistributionhazard ratio (SHR) analysis showed that patients with high or

    grade 3+ GLDC expression have a 3-fold higher risk of lung

    cancer mortality compared to patients with low or grade 0

    GLDC expression, even when adjusted for cancer stage (SHR =

    3.01, 95% confidence interval [CI]: 1.486.10, p = 0.002) (Fig-

    ure 6B). Cumulative mortality analysis also showed that high

    GLDC expression (grade 3+) is significantly associated with

    higher cumulative incidence of mortality across 143 NSCLC

    lung cancer patients, even when adjusted for cancer stage

    (p = 0.005) (Figure 6C). CD166 expression was not significantly

    associated with higher mortality in lung cancer patientswhich

    was not unexpected given that only 1 in 5 3 103 CD166+ cells

    are tumorigenic (Figures S6AS6C). Indeed, coimmunostaining

    of lung tumors revealed that GLDC+ cells mostly form a subset

    of CD166+ cells, andthat notall CD166+ cells areGLDC+ (Figures

    6D and S6D). LIN28B immunohistochemistry was also not signif-

    icantly correlated with lung cancer patient mortality (Figures

    S6AS6C), although western blots revealed that lung TICs

    express a second LIN28B isoform that is indiscernible from

    immunohistochemistry staining,thus renderingour LIN28B stain-

    ing results inconclusive (Figure S6E). Our immunohistochemistry

    results on clinical tumor samples areconsistent with theidea that

    lung TICs constitute the bulk of the tumors in late stages of

    malignancy and demonstrate that aberrant activation of GLDC

    is significantly associated with human mortality in NSCLC

    patientsfurther supporting its role as a metabolic oncogene in

    human NSCLC.

    Aberrant GLDC Expression in Other Cancers

    To check whether aberrant GLDC expression is specific to

    NSCLC, we examined a variety of other cancers. Surprisingly

    GLDC is also aberrantly upregulated in subsets of primary

    tumors from other cancers, especially ovarian and germ cell

    tumors (Figure 7A; Table S3). Further analysis of 606 human

    cancer cell lines showed that 158 (26.1%) cancer cell lines over-

    express GLDC, including lines from ovarian, germ cell, cervical,

    lung, lymphoma, prostate, bladder, and colon cancer (Figure 7B;

    Table S4). To test whether GLDC is also required for growth by

    one of these GLDC-overexpressing cell lines, we knocked

    266 Cell148, 259272, January 20, 2012 2012 Elsevier Inc.

  • 8/9/2019 [5] Zhang et all Cell

    9/14

    A B

    Glycine-related metabolites Glycolysis

    4.00

    le)

    Cells

    2.00

    4.00 Cells

    arcos ne

    2 Betaine aldehyde3 Serine

    4 Glycine

    -

    2 Phosphoenolpyruvate

    3 Pyruvate

    0.25

    0.50

    1.00

    .

    1 2 3

    Foldchange(Log2scal

    3T3-GD/Ctrl

    HLF-GD/Ctrl

    A549-GD-sh/Ctrl0.03

    0.06

    0.13

    0.25

    0.50

    1.00

    1 2 3 4

    Foldchange(Log2sca

    le)

    3T3-GD/Ctrl

    HLF-GD/Ctrl

    A549-GD-sh/Ctrl

    2.00

    4.00

    (Log2scale)

    1.5

    2.0

    2.5

    ateOD

    3T3-GD

    3T3 Pyrimidines

    1 Thymidine

    2 Deoxyuridine

    3 Thymine

    4 Cytosine

    DC

    1.5

    2.0

    2.5A549

    A549-GD-sh

    0.02

    F

    0.25

    0.50

    .1 2 3 4 5

    Foldchang

    3T3-GD/Ctrl

    HLF-GD/Ctrl

    A549-GD-sh/Ctrl0.0

    0.5

    1.0

    0 1 2 4 8 24 Hr

    Lacta 5 Uracil

    0.0

    0.5

    1.0

    0 1 2 4 8 24 Hr

    E

    0

    20

    40

    60

    80

    100

    120

    Agarcolonynumber 0 uM Sarcosine

    10 uM Sarcosine

    0

    20

    40

    0 0 .1 0. 2 0. 5 1 2 5 1 0 2 0 0 0 .1 0 .2 0 .5 1 2 5 1 0 20

    No.ofadherentcolonies

    /200cells

    Methotrexate ( M)

    3T3

    Control GLDC

    G

    Serine

    Sarcosine

    Glycolysis

    Betaine Pyrimidine

    t b li

    Phosphoserine

    Gl cine CH -THF

    Tetrahydrofolate (THF)

    Methotrexate0

    5

    10

    15

    20

    0 0.1 0.2 0.5 1 2 5 10 0 0.1 0.2 0.5 1 2 5 10

    No.ofadherentcolonies

    /200cells

    Methotrexate ( M)

    HLF

    Control GLDC

    GLDCAldehyde metabolism

    CO2NH3NADH

    0

    50

    100

    0 10 20 50 0 10 20 50 0 10 20 50

    No.ofsoftagarcolonies

    /1000cells

    Methotrexate ( M)

    A549

    Control GLDC shRNA1 GLDC shRNA2

    Figure 5. Metabolomics of Cells upon GLDC Overexpression and Knockdown

    (AD) Relative fold change in levels of (A) glycine-related metabolites, (B) glycolysis intermediates, and (D) pyrimidines in 3T3 cells with GLDC overexpression

    (3T3-GD/Ctrl), HLF cells with GLDC overexpression (HLF-GD/Ctrl), and A549 cells with GLDC knockdown (A549-GD-sh/Ctrl), as determined by LC-MS

    metabolomics. (C) Lactate production by 3T3 cells with GLDC overexpression or A549 cells with GLDC knockdown.

    (E) Effects of the antifolate drug methotrexate on colony formation after GLDC overexpression or knockdown. 3T3 and HLF cells overexpressing GLDC were

    plated at clonal density and exposed to varying concentrations of methotrexate for 8 days. A549 cells with GLDC knockdown were plated in soft agar at clonal

    density and exposed to varying concentrations of methotrexate for 14 days.

    (F) Effects of sarcosine on soft agar colony formation after GLDC knockdown in A549 cells. One thousand cells were seeded in soft agar at clonal density and

    exposed to 10mM sarcosine for 14 days.

    (G) Model of metabolic flux changes induced by GLDC.

    In all panels, error bars represent SEM. See also Figure S5.

    Cell148, 259272, January 20, 2012 2012 Elsevier Inc. 267

  • 8/9/2019 [5] Zhang et all Cell

    10/14

    A GLDC

    1+ 3+2+0

    Ad

    B

    GLDC Intensity SHR (95% CI) P-value

    0.005

    1 1.28 (0.61 2.71) 0.514

    0.8

    0.6

    1.0C GLDC3+

    1+2+

    0

    tiveincidence

    2 1.19 (0.50 2.84) 0.700

    3 3.01 (1.48 6.10) 0.002

    D

    0.2

    .

    Time to lung cancer death (years)

    P = 0.005

    n=143

    Cumul

    0 2 4 6 8 10 12 14

    0

    CD166

    CD166 GLDC DAPIDAPI CD166 GLDC DAPIDAPI

    GLDC GLDCCD166

    Figure 6. GLDC Is a Prognostic Indicator for Mortality in NSCLC Patients

    (A) GLDCimmunohistochemistrystaining in a NSCLCtumor microarray (n = 143).Representative images shown for humanprimarylung adenocarcinomas (AdC)

    immunostained with GLDC. Staining intensity grade is indicated in the upper right corner. The boxed regions in the upper images are shown at higher magni-

    fication in the lower images. Scale bar, 100 mm.

    (B) Subdistribution hazard ratios for each GLDC staining intensity grade, adjusted for American Joint Committee on Cancer (AJCC) staging. CI, confidence

    interval.

    (C) Cumulative incidence of lung cancer mortality adjusted for AJCC staging, for patients with each GLDC staining intensity grade.

    (D) Coimmunofluorescence staining of CD166 (red) and GLDC (green) on primary lung cancer patient tumors, counterstained with DAPI (blue). Representative

    cases with coexpression of high levels of CD166 and high levels of GLDC (left panel) and low levels of CD166 and low levels of GLDC (right panel) are shown.

    Higher magnification inset is shown in bottom left corner. Scale bar, 50mm.

    See alsoFigure S6andTable S2.

    268 Cell148, 259272, January 20, 2012 2012 Elsevier Inc.

  • 8/9/2019 [5] Zhang et all Cell

    11/14

    down GLDC in CACO2 cells. Indeed we found that GLDC knock-

    down reduced their proliferation and tumorigenic potential upon

    transplantation, suggesting thatGLDCmay act as an oncogene

    in other cancer cells as well (Figures 7C7E). To examine the

    possibility that GLDC is a housekeeping gene for cell prolifera-

    tion, we also knocked down GLDC in normal HLFs (Figure S7A).

    We found that HLF proliferation was unaffected by retroviral

    knockdown of GLDC (Figures S7B and S7C). Furthermore we

    observed that GLDC is highly expressed only in a few normal

    tissues, including postmitotic liver cells, kidney cells, placenta

    cells, and olfactory bulb neurons (Figure S7D). Altogether our

    observations in both experimental and clinical settings suggest

    that human GLDC is not a housekeeping gene required for cell

    proliferation but rather an oncogenic metabolic enzyme aber-

    rantly upregulated in NSCLC and possibly several other human

    cancers.

    DISCUSSION

    TIC State in Lung Cancer

    Our work sheds new light on the nature of the TIC state and the

    role of metabolic reprogramming in tumorigenesis. In this study,

    we isolated a subpopulation of TICs from NSCLC patients using

    themarker CD166 andshowed that both theoncogenic stem cell

    2

    3

    4

    LDCin

    sues

    A GLDC expression across primary tissue cancers

    -4

    -3

    -2

    -1

    0

    1

    Ovary

    Germc

    ell

    Cervix

    Brain

    Lung

    Lymphoma

    Prostate

    Head&N

    eck

    Esophagus

    Bladder

    Colon

    Log2

    foldchangeof

    tumor/norma

    ltis

    10000B

    1

    10

    100

    1000

    NormalizedGLDCintensity

    250

    0.5

    1.0

    RelativeGLDC

    expression

    Ctrl-shGD-sh

    C

    CACO2

    600,000

    800,000

    fcells

    Ctrl-sh

    GD-sh

    28B-sh

    D E

    Ctrl-sh GD-sh 28B-sh

    .

    0

    200,000

    400,000

    0 2 4 6

    Numbero

    Days0.0

    0.5

    1.0

    R

    elativeLIN28B

    expression

    Ctrl-sh28B-sh

    Figure 7. GLDC Is Aberrantly Expressed in Other Cancers

    (A) Log2-transformed fold changes in GLDC expression of patient tumors versus normal adjacent tissues across different cancers. Data are normalized and

    aggregated from 51 GEO datasets containing 84 sets of tumor expression data (tumor versus normal) with 2,020 tumor samples and 671 normal samples. Fold

    change cutoff was set at 1.5 by yellow line.

    (B) NormalizedGLDCexpression across various cancer cell lines (n = 606). Yellow line indicates the mean value (250) ofGLDCexpression intensities between

    patient lung tumors and normal lung tissues.

    (C) qRT-PCR forGLDCand LIN28Bin CACO2 colon cancer cells expressing shRNA against GLDC(GD-sh) orLIN28B(28B-sh).

    (D) Proliferation curve of CACO2 cells expressing shRNA againstGLDC(GD-sh) orLIN28Bgene (28B-sh) described in (C). Cell numbers were measured on days

    2, 4, and 6.

    (E) Tumorformation in mice upon injecting2.53 104

    CACO2 colon cancer cells withGLDC knockdown (CACO2-GD-sh)or LIN28B knockdown (CACO2-28B-sh).

    Mice were assessed by week 13 (n = 6).

    In all panels, error bars represent SEM. See also Figure S7andTable S3 and S4.

    Cell148, 259272, January 20, 2012 2012 Elsevier Inc. 269

  • 8/9/2019 [5] Zhang et all Cell

    12/14

    factor LIN28B and the glycine metabolism enzyme GLDC drive

    the tumorigenicity of lung cancer TICs.

    Our data showed that CD166 enriched for TICs in primary

    NSCLC, and that CD166 served as an inert surface marker. In

    contrast, our results on CD133 are different from the results re-ported by Eramo et al. (2008) even though both studies used

    the same CD133 antibody. This is most likely due to differences

    in the xenotransplantation assays, which tend to underestimate

    the true frequency of TICs. We employed a more sensitive

    mouse xenotransplantation assay using NOD/SCID Il2rg/

    mice instead of SCID mice, and we directly transplanted primary

    tumor cells with Matrigel instead of expanding the tumor cells

    in vitro. Previous studies have demonstrated that using a more

    sensitive mouse xenotransplantation assay dramatically im-

    proves our understanding of TICs (Quintana et al., 2008). Our

    present study supports this notion, leading us to CD166 as a

    new marker for the lung TIC-containing fraction. In normal phys-

    iology, CD166 is expressed predominantly during embryonic

    development, including the embryonic upper airway, primitivecardiac cells, and mesenchymal stem cells (Avril-Delplanque

    et al., 2005; Murakami et al., 2007; Hennrick et al., 2007; Sabatini

    et al., 2005). Expression of CD166 in the embryonic lung is

    consistent with our observation that CD166+ lung TICs express

    high levels of embryonic lung transcription factors like PEA3

    and thetrachealess homologNPAS1, as well as the oncogenic

    stem cell factor LIN28B(Liu et al., 2003; Levesque et al., 2007;

    Viswanathan et al., 2009). Interestingly, mouse Lin28 is also

    expressed in the embryonic lung during normal development

    (Yang and Moss, 2003). These observations suggest that the

    TIC state in lung cancer is similar to the embryonic lung progen-

    itor state in many aspects.

    GLDC Is a Metabolic Oncogene

    Our results demonstrate that multiple components in the glycine/

    serine pathway are also oncogenes. In addition to embryonic

    lung factors, lung TICs also express high levels of GLDC,

    GCAT, SHMT1/2, PSPH,and PSAT1, suggesting that TICs rely

    on glycine/serine metabolism for tumorigenesis. Overexpression

    of catalytically active GLDC, as well as PSAT1, PSPH, and

    SHMT2, could induce cellular transformation in 3T3 cells to

    form tumors, whereas retroviral knockdown of GLDC signifi-

    cantly reduces the tumorigenicity of lung cancer cells. We further

    observed that GLDC+ cells mostly form a subset of CD166+ cells

    in lung tumors.

    PSAT1, PSPH, and SHMT1/2 lie upstream of GLDC in

    the glycine/serine pathway, diverting glycolytic flux from3-phosphoglycerate through serine to glycine. GLDC is an

    oxidoreductase that catalyzes the irreversible rate-limiting step

    of glycine catabolism, by breaking down each glycine molecule

    in the glycine cleavage system to produce NADH, CO2, NH3, and

    CH2-THF (Kume et al., 1991). CH2-THF fuels the one-carbon/

    folate metabolism pool, which in turn supplies methylene groups

    for biosynthesis (Tibbetts and Appling, 2010). Consistent with

    these facts, we found that GLDC regulates many metabolites

    in glycolysis and the glycine/serine pathway, leading to specific

    changes in pyrimidine synthesis. Pyrimidine derivatives like

    thymidine, in turn, are required for nucleotide synthesis in cell

    proliferation. Recent studies suggest that early oncogenesis

    involves aberrant activation of cell proliferation, which then leads

    to a crisis of nucleotide deficiency and replication stress (Bester

    et al., 2011)a crisis thatGLDC upregulation could overcome

    for continued progression in tumorigenesis. Interestingly, we

    found that GLDC also increases the levels of N-methylglycineor sarcosine, an oncometabolite implicated in prostate cancer

    (Sreekumar et al., 2009). Furthermore, we observed that GLDC

    promotes glycolysis. Combined with our findings on LIN28,

    which has been shown to promote glucose uptake and glycol-

    ysis (Zhu et al., 2010, 2011), GLDC might be cooperating with

    LIN28 as well as PSAT1, PSPH, and SHMT1/2 to divert the

    glycolytic flux to glycine and produce CH2-THF. These observa-

    tions support the notion that the Warburg effect promotes

    biosynthesis for tumorigenesis (Hsu and Sabatini, 2008; Vander

    Heiden et al., 2009).

    GLDC and Glycine Metabolism Are Relevant to Human

    Cancer Patients

    From the prognostic perspective, aberrant GLDC expression issignificantly correlated with the survival rates of NSCLC patients.

    This is consistent with the model that TIC clones expand to

    constitute the bulk of the tumor in advanced stages of malig-

    nancy (Boiko et al., 2010). Aberrantly increased GLDC is also

    widespread in many other human cancers, including lymphoma,

    ovarian, germ cell, cervical, prostate, bladder, and colon cancer,

    whereas most normal adult human tissues express very low

    levels of GLDC. Our experimental data further suggest that in

    cancers that rely on GLDC and glycine metabolism, the highly

    toxic antifolate drug methotrexate might be initially effective

    because it targets TICs, although our data suggest an even

    more effective chemotherapy could be potentially achieved by

    combining an antifolate drug with a GLDC inhibitor or by search-

    ing for a glycine cleavage complex-specific antifolate drug

    much like the search for kinase-specific inhibitors in targeted

    cancer therapy.

    Our study links a glycine metabolism enzyme to lung cancer

    and tumorigenesis. Recently several metabolic enzymes have

    been linked to cancer in patients, supporting the status of meta-

    bolic reprogramming as a new hallmark of cancer (Hanahan and

    Weinberg, 2011). In particular, the pyruvate kinase M2 isoform

    PKM2, isocitrate dehydrogenase IDH1/2, and phosphoglycerate

    dehydrogenase PHGDH have been implicated in multiple

    cancers (Christofk et al., 2008; Parsons et al., 2008; Dang et al.,

    2009; Locasale et al., 2011; Possemato et al., 2011). Regardless

    of the controversy over the frequency of TICs at different stages

    of malignancy, our approach shows that characterizing theunique molecular basis that defines cancer cells with tumori-

    genic capacity may nevertheless provide novel drug targets for

    advancing cancer therapy.

    EXPERIMENTAL PROCEDURES

    Tumor Cell Preparation

    NSCLC tumors were collected from patients according to protocols approved

    by the Ethics Committee of the National University of Singapore. Samples

    were washed, dissociated, and incubated in DNase and collagenase/dispase.

    After incubation, cell clusters and red blood cells were removed. Then single

    cells were resuspended and ready for transplantation. See the Extended

    Experimental Proceduresfor more details.

    270 Cell148, 259272, January 20, 2012 2012 Elsevier Inc.

  • 8/9/2019 [5] Zhang et all Cell

    13/14

  • 8/9/2019 [5] Zhang et all Cell

    14/14

    Hsu, P.P., and Sabatini, D.M. (2008). Cancer cell metabolism: Warburg and

    beyond. Cell134, 703707.

    Hu, Y.F., and Smyth, G.K. (2009). ELDA: extreme limiting dilution analysis for

    comparing depleted and enriched populations in stem cell and other assays.

    J. Immunol. Methods347, 7078.

    Huang, W., Sherman, B.T., and Lempicki, R.A. (2009). Systematic and integra-

    tive analysis of large gene lists using DAVID bioinformatics resources. Nat.

    Protoc.4, 4457.

    Ishizawa, K., Rasheed, Z.A., Karisch, R., Wang, Q.J., Kowalski, J., Susky, E.,

    Pereira, K., Karamboulas, C., Moghal, N., Rajeshkumar, N.V., et al. (2010).

    Tumor-initiating cells are rare in many human tumors. Cell Stem Cell 7,

    279282.

    Jemal, A., Bray, F., Center, M.M., Ferlay, J., Ward, E., and Forman, D. (2011).

    Global cancer statistics. CA Cancer J. Clin. 61, 6990.

    Kume, A., Koyata, H., Sakakibara, T., Ishiguro, Y., Kure, S., and Hiraga, K.

    (1991). The glycine cleavage system. Molecular cloning of the chicken and

    human glycine decarboxylase cDNAs and some characteristics involved in

    the deduced protein structures. J. Biol. Chem.266, 33233329.

    Kure, S., Kato, K., Dinopoulos, A., Gail, C., DeGrauw, T.J., Christodoulou, J.,

    Bzduch, V., Kalmanchey, R., Fekete, G., Trojovsky, A., et al. (2006). Compre-

    hensive mutation analysisof GLDC, AMT,and GCSHin nonketotic hyperglyci-

    nemia. Hum. Mutat. 27, 343352.

    Levesque, B.M., Zhou, S.T., Shan, L.,Johnston, P.,Kong,Y.P., Degan, S.,and

    Sunday, M.E. (2007). NPAS1 regulates branching morphogenesis in embry-

    onic lung. Am. J. Respir. Cell Mol. Biol. 36, 427434.

    Liu, Y.R., Jiang, H.Y., Crawford, H.C., and Hogan, B.L.M. (2003). Role for ETS

    domain transcription factors Pea3/Erm in mouse lung development. Dev. Biol.

    261, 1024.

    Locasale, J.W., Grassian, A.R., Melman, T., Lyssiotis, C.A., Mattaini, K.R.,

    Bass, A.J., Heffron, G., Metallo, C.M., Muranen, T., Sharfi, H., et al. (2011).

    Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to

    oncogenesis. Nat. Genet. 43, 869874.

    Murakami, Y., Hirata,H., Miyamoto, Y., Nagahashi, A., Sawa, Y., Jakt, M.,Asa-

    hara, T., and Kawamata, S. (2007). Isolation of cardiac cells from E8.5yolk sac

    by ALCAM (CD166) expression. Mech. Dev.124, 830839.

    Nakai, T., Nakagawa, N., Maoka, N., Masui, R., Kuramitsu, S., and Kamiya, N.

    (2005). Structure of P-protein of the glycine cleavage system: implications for

    nonketotic hyperglycinemia. EMBO J.24, 15231536.

    Parsons, D.W., Jones, S., Zhang, X.S., Lin, J.C.H., Leary, R.J., Angenendt, P.,

    Mankoo, P., Carter, H., Siu, I.M., Gallia, G.L., et al. (2008). An integrated

    genomic analysis of human glioblastoma multiforme. Science 321, 1807

    1812.

    Possemato, R., Marks, K.M., Shaul, Y.D., Pacold, M.E., Kim, D., Birsoy, K.,

    Sethumadhavan, S., Woo, H.K., Jang, H.G., Jha, A.K., et al. (2011). Functional

    genomics reveal that the serine synthesis pathway is essential in breast

    cancer. Nature476, 346350.

    Quintana, E., Shackleton, M., Sabel, M.S., Fullen, D.R., Johnson, T.M., and

    Morrison, S.J. (2008). Efficient tumour formation by single human melanoma

    cells. Nature456, 593598.

    Quintana, E., Shackleton, M., Foster, H.R., Fullen, D.R., Sabel, M.S., Johnson,

    T.M., and Morrison, S.J. (2010). Phenotypic heterogeneity among tumorigenic

    melanoma cells from patients that is reversible and not hierarchically orga-

    nized. Cancer Cell18, 510523.

    Reya, T., Morrison, S.J., Clarke, M.F., and Weissman, I.L. (2001). Stem cells,

    cancer, and cancer stem cells. Nature414, 105111.

    Rosen, J.M., and Jordan, C.T. (2009). The increasing complexity of the cancer

    stem cell paradigm. Science324, 16701673.

    Sabatini, F., Petecchia, L., Tavian, M.,Jodon de Villeroche, V.,Rossi,G.A., and

    Brouty-Boye, D. (2005). Human bronchial fibroblasts exhibit a mesenchymal

    stem cell phenotype and multilineage differentiating potentialities. Lab. Invest.

    85, 962971.

    Sequist, L.V., Joshi, V.A., Janne, P.A., Muzikansky, A., Fidias, P., Meyerson,

    M., Haber, D.A., Kucherlapati, R., Johnson, B.E., and Lynch, T.J. (2007).

    Response to treatment and survival of patients with non-small cell lung cancer

    undergoing somatic EGFR mutation testing. Oncologist 12, 9098.

    Sreekumar, A., Poisson, L.M., Rajendiran, T.M., Khan, A.P., Cao, Q., Yu, J.D.,

    Laxman, B., Mehra, R., Lonigro, R.J., Li, Y., et al. (2009). Metabolomic profiles

    delineate potential role for sarcosine in prostate cancer progression. Nature

    457, 910914.

    Tibbetts, A.S., and Appling, D.R. (2010). Compartmentalization of mammalian

    folate-mediated one-carbon metabolism. Annu. Rev. Nutr.30, 5781.

    Vander Heiden, M.G., Cantley, L.C., and Thompson, C.B. (2009). Under-

    standing the Warburg effect: the metabolic requirements of cell proliferation.

    Science 324, 10291033.

    Vander Heiden, M.G., Locasale, J.W., Swanson, K.D., Sharfi, H., Heffron, G.J.,

    Amador-Noguez, D., Christofk, H.R., Wagner, G., Rabinowitz, J.D., Asara,

    J.M., and Cantley, L.C. (2010). Evidence for an alternative glycolytic pathway

    in rapidly proliferating cells. Science329, 14921499.

    Viswanathan, S.R., Powers, J.T., Einhorn, W., Hoshida, Y., Ng, T.L., Toffanin,

    S., OSullivan, M., Lu, J., Phillips, L.A., Lockhart, V.L., et al. (2009). Lin28

    promotes transformation and is associated with advanced human malignan-

    cies. Nat. Genet.41, 843848.

    Vogelstein, B., and Kinzler, K.W. (2004). Cancer genes and the pathways they

    control. Nat. Med.10, 789799.

    Warburg, O. (1956). On the origin of cancer cells. Science 123, 309314.

    Yang, D.H., and Moss, E.G. (2003). Temporally regulated expression of Lin-28

    in diverse tissues of the developing mouse. Gene Expr. Patterns 3, 719726.

    Zhu, H., Shah, S., Shyh-Chang, N., Shinoda, G., Einhorn, W.S., Viswanathan,

    S.R., Takeuchi, A., Grasemann, C., Rinn, J.L., Lopez, M.F., et al. (2010). Lin28a

    transgenic mice manifest size and puberty phenotypes identified in human

    genetic association studies. Nat. Genet.42, 626630.

    Zhu, H., Shyh-Chang, N., Segre` , A.V., Shinoda, G., Shah, S.P., Einhorn, W.S.,

    Takeuchi, A., Engreitz, J.M., Hagan, J.P., Kharas, M.G., et al; DIAGRAM

    Consortium; MAGIC Investigators. (2011). The Lin28/let-7 axis regulates

    glucose metabolism. Cell147, 8194.