Argininosuccinate lyase is a potential therapeutic target in breast cancer

  • Authors:
    • Hau-Lun Huang
    • Wei-Ching Chen
    • Hui-Ping Hsu
    • Chien-Yu Cho
    • Yu‑Hsuan Hung
    • Chih-Yang Wang
    • Ming-Derg Lai
  • View Affiliations

  • Published online on: September 16, 2015     https://doi.org/10.3892/or.2015.4280
  • Pages: 3131-3139
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Arginine is a non-essential amino acid that modulates nitric oxide production and cancer homeostasis. In our previous study, we observed that blocking argininosuccinate lyase (ASL) attenuates tumor progression in liver cancer. However, the role of ASL in human breast cancer has been studied to a lesser degree. In the present study, we investigated the effect of targeting ASL in breast cancer. We found that ASL was induced by ER stress and was significantly upregulated in breast cancer tissues compared to that in the corresponding normal tissues. Downregulation of ASL inhibited the growth of breast cancer in vitro and in vivo. The level of cell cycle-related gene, cyclin A2, was reduced and was accompanied by a delay in G2/M transition. ASL shRNA-induced cell inhibition was rescued by exogenous cyclin A2. Furthermore, autophagy was observed in the cells expressing ASL shRNA, and inhibition of autophagy reduced cell growth, indicating that autophagy played a cell survival role in the ASL knockdown cells. Moreover, inhibition of ASL reduced NO content. Introduction of the NO donor partially restored the growth inhibition by ASL shRNA. Thus, the mechanism induced by ASL shRNA which occurred in human breast cancer may be attributed to a decrease in cyclin A2 and NO.

Introduction

Breast cancer is a common cancer in women worldwide. Many physiological conditions, including hormone secretion and metabolic homeostasis, influence breast cancer progression (1). Breast cancer patients with metabolic dysregulation are associated with poor response to current chemotherapy (2). The growth of tumor cells is coupled by metabolic reprogramming (3,4). The metabolic shift is observed during carcinogenesis and has been considered to be a reliable marker for tumors. The intermediary metabolism can also fuel cell growth. For example, cancer cells are addicted to glutamine due to its usage as a supplement. The glucose metabolites, serine and glycine, mediate one-carbon metabolism which is important in tumorigenesis (5,6). The semi-essential amino acid arginine plays an important role in nitric oxide production and the urea cycle, and is a precursor for glutamate, proline, polyamones and agmatine (7). The concentration of plasma arginine is lower in breast, colon and pancreatic cancer patients (810). A dietary supplement with arginine increases colonic carcinogenesis (11). Compared to breast cancer patients fed a standard diet, patients with dietary L-arginine supplementation have higher tumor protein synthesis. Furthermore, the protein synthesis rate was found to be highly correlated with Ki67 expression (12). In contrast, deprivation of dietary arginine inhibits cancer metastasis (13,14). Arginine depletion by ADI is used as an approach in cancer therapy (15,16).

Arginine is synthesized from citrulline by argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL). ASS catalyzes the conversion of citrulline and aspartate into argininosuccinate. Argininosuccinate is then converted into arginine and fumarate by ASL. A high level of ASS is observed in malignant lung, ovarian, gastric and colonic epithelium compared to corresponding normal epithelium (1719). In contrast, tumors usually express reduced ASS including breast cancer, hepatocellular carcinoma, melanoma, renal cell carcinoma and pancreatic cancer, and the ASS level is inversely correlated with survival (2023). Tumors with loss of ASS are dependent on extracellular arginine for growth, characteristic of arginine auxotrophy. Breast cancer and melanoma with deficiency of ASS are sensitive to arginine deprivation via arginine deiminase (4,24). The complex of ASS and ASL with NOS contributes to the efficient channeling for NO production (25,26). ASL-deficient mice and argininosuccinic aciduria patients have a deficiency in the production of NO (25). ASL is highly expressed in hepatocellular carcinoma and downregulation of ASL by shRNA attenuates tumor growth in vivo (27).

Endoplasmic reticulum (ER), which plays a major role in membrane and secretory protein synthesis, has been associated with metabolic disease (2830). Endoplasmic reticulum stress, which emanates from the accumulation of unfolded protein, has a profound impact on the pathogenesis of many diseases, including liver disease, diabetes and cancer (3134). In response to ER stress, the unfolded protein response (UPR) initiates signaling cascades and restores the protein-folding homeostasis. The amino acid metabolism genes are also activated under ER stress condition (35,36). Given the relationship between ER stress, metabolism and cancer progression, it is predictable that ER stress may affect metabolic enzymes in cancer cells. ASL upregulation by an ER stress inducer was observed in liver cancer cells (27). Arginine metabolic enzyme is usually expressed in liver cells. It is uncertain whether the arginine metabolic enzyme, ASL, exerts functions in other types of tissues. The present study aimed to determine the relationship between ER stress and ASL in breast cancer cells and to ascertain whether the arginine-NO complex mediates breast cancer growth.

Our results revealed that ASL is elevated by ER stress and is highly expressed in breast tumor tissues. Downregulation of ASL by ASL shRNA decreased tumor growth in vivo and in vitro. ASL knockdown induced cyclin A2 degradation and the cell growth was rescued by exogenous cyclin A2. Furthermore, ASL downregulation inhibited NO expression and induced autophagy.

Materials and methods

Reagents, chemicals and antibodies

The anti-ASL antibody was purchased from Abnova (Taipei, Taiwan). The anti-GRP78 antibody was purchased from BD (Erembodegem, Belgium). The anti-β-actin antibody was purchased from Chemicon (Pittsburgh, PA, USA). The anti-GAPDH antibody was purchased from GeneTex (Irvine, CA, USA). The anti-cyclin A2, anti-CDK4 and anti-CDK2 antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). The anti-cyclin B1 and anti-cyclin E1 antibodies were purchased from Epitomics (Burlingame, CA, USA). The anti-cyclin D1 antibody was purchased from Cell Signaling (Beverly, MA, USA). The anti-LC3B antibody was purchased from Sigma (St. Louis, MO, USA). The anti-HA antibody was purchased from Roche Applied Science (Mannheim, Germany). The powder of G418, thiazolyl blue tetrazolium bromide (MTT), 3-methyladenine (3-MA), bafilomycin A1, sodium nitrite, 2-amino purine and arginine were purchased from Sigma. The plasmid containing ASL was purchased from OriGene Technologies, Inc. (Rockville, MD, USA). The plasmid containing cyclin A2 was provided by Dr Ih-Jen Su (Division of Clinical Research, National Health Research Institute, Taiwan).

Cell culture

MCF-7 and MDA MB-231 cells and the stable transfectants were cultured in Dulbecco's modified Eagle's medium (DMEM) (HyClone, Logan, UT, USA) containing 10% fetal bovine serum (FBS) (Biological Industries, Kibbutz Beit-Haemek, Israel), 100 U/ml penicillin and 100 µg/ml streptomycin (Invitrogen Corporation, Carlsbad, CA, USA) at 37°C in 5% CO2.

Reverse transcription-polymerase chain reaction (RT-PCR) analysis

Total RNA was extracted with TRIzol (MDBio, Taiwan). cDNA was synthesized using M-MLV transcriptase (Promega, Madison, MI, USA). PCR was performed using Pro Taq polymerase (PROtech Technology Enterprise Co., Ltd., Taipei, Taiwan) on a thermocycler (ABI, Foster City, CA, USA). The 5′ and 3′ human ASL gene-specific primers were: 5′-TGA TGC CCC AGA AGA AAA AC-3′ (sense) and 5′-TTT GCG GAC CAG GTA ATA GG-3′ (antisense); the 5′ and 3′ human GRP78 gene-specific primers were: 5′-CGC CTC ATC GGA CGC ACT TG-3′ (sense) and 5′-AGG TTC CAC CGC CCA GGT CA-3′ (antisense); the 5′ and 3′ human CCND1 gene-specific primers were: 5′-AAC TAC CTG GAC CGC TTC CT-3′ (sense) and 5′-TGA GGC GGT AGT AGG ACA GG-3′ (antisense); the 5′ and 3′ human CCNE1 gene-specific primers were: 5′-ATC CCC ACA CCT GAC AAA GA-3′ (sense) and 5′-AGG GGA CTT AAA CGC CAC TT-3′ (antisense); the 5′ and 3′ human CCNA2 gene-specific primers were: 5′-GCA CCC CTT AAG GAT CTT CC-3′ (sense) and 5′-CCT CTC AGC ACT GAC ATG GA-3′ (antisense); the 5′ and 3′ human CCNB1 gene-specific primers were: 5′-GGC CAA AAT GCC TAT GAA GA-3′ (sense) and 5′-AA CAT GGC AGT GAC ACC AA-3′ (antisense).

Western blotting

The cell lysates were prepared in RIPA lysis buffer. The protein concentration was determined with the Micro BCA™ protein assay kit (Millipore, Billerica, MA, USA). Cell lysates were loaded onto acrylamide gels and were then transferred onto polyvinylidene fluoride membranes (Amersham Biosciences, Piscataway, NJ, USA) after electrophoresis. The membranes were incubated with the indicated antibody for the specific protein, probed with ECL Western Blotting Detection system (Millipore) and visualized using the BioSpectrum AC imaging system.

Tissue samples

The specimens of breast cancer and corresponding normal liver were obtained from the Human Biobank within the Research Center of Clinical Medicine of the National Cheng Kung University Hospital (Tainan, Taiwan) following the approval of the Institutional Review Board.

Oncomine database analysis

The expression of ASL in clinical specimens of cancer vs. normal patients was analyzed using Oncomine database (https://www.oncomine.org/resource/login.html). We analyzed the results of fold-change, cancer subtypes and p-values with a threshold of p<0.05.

Kaplan-Meier plotter analysis

The overall survival of the patients with high and low ASL expression was analyzed using Kaplan-Meier plotter (http://www.kmplot.com/). We analyzed the relapse-free survival and ASL expression with probe 204608 in upper tertile patients. The number-at-risk, the hazard ratio and the log-rank p were indicated.

RNA interference and lentiviral production

The shRNA targeting ASL was obtained from the National RNAi Core Facility (Academia Sinica, Taipei, Taiwan). The target sequence of shRNA was 5′-AGGAGGCTGCTGTGTGTTT-3′ (shASL1669). The lentiviral production was managed according to the protocol provided by the National RNAi Core Facility.

Colony formation assay

Colony formation was performed by seeding cells into 6-well plates. The colonies were stained with 2% methylene blue and counted after incubation for 10 days in 5% CO2 and 37°C.

Anchorage-independent growth ability

Agar (0.6%) in DMEM was prepared as an under layer in a plastic Petri dish. Five thousand cells were suspended in 0.3% agar in DMEM containing 10% FBS and added over the upper layer. The plates were placed in a 5% CO2 atmosphere humidified incubator at 37°C for 14 days and the colonies were quantified.

Tumorigenicity in NOD/SCID mice

NOD/SCID mice were obtained from the Animal Center of the National Cheng Kung University. All study protocols were approved by the Animal Welfare Committee of the National Cheng Kung University. MDA MB-231 cells (5×105) were subcutaneously implanted into the NOD/SCID mice. For our model of inhibiting tumor growth by lentiviral ASL shRNA, the NOD/SCID mice implanted with MDA MB-231 cells for 10 days were intratumorally injected with lentiviral particles.

Monodansylcadaverine (MDC) staining of autophagy

The monodansylcadaverine (MDC) staining was analyzed by Cayman autophagy/cytotoxicity dual staining kit (Cayman Chemical Company, Ann Arbor, MI, USA) and detected by fluorescence microscopy.

Measurement of intracellular arginine content

The intracellular arginine concentration was analyzed by HPLC analysis using Agilent ZORBAX Eclipse AAA column (Agilent PN 993400-902) (Agilent Technologies, Inc., Santa Clara, CA, USA).

Statistical analysis

All statistical analyses were performed using the Student's t-test. The error bars in the graphs represent the SEM.

Results

ASL expression is induced by ER stress and is highly expressed in breast cancer

To analyze whether ASL is induced by ER stress, the breast cancer cell lines, MCF-7 and MDA MB-231, were incubated with tunicamycin. ASL expression was increased after tunicamycin treatment as demonstrated by western blotting (Fig. 1A). We further examined whether ASL expression was increased in human breast cancer. Western blot analysis was used to detect ASL and GRP78 expression in 14 paired breast cancer and adjacent normal breast tissues. The results indicated that ASL was overexpressed in the breast cancer tissues (Fig. 1B). From the Oncomine database, ASL expression was upregulated in different subtypes of breast cancer (Fig. 1C, Tables I and II). Kaplan-Meier plotter analysis in breast cancer showed a correlation between overexpression of ASL and lower overall survival rates (Fig. 1D).

Table I

ASL expression of the normal and breast cancer tissues from the Oncomine database.

Table I

ASL expression of the normal and breast cancer tissues from the Oncomine database.

Tissue (no.)P-valueFold-changeRef.
Normal breast (61)
Mucinous breast carcinoma (4)
0.0041.601TCGA
Normal breast (61)
Invasive breast carcinoma (76)
2.56E-081.316
Normal breast (61)
Invasive ductal and lobular carcinoma (3)
2.20E-021.298
Normal breast (61)
Invasive lobular breast carcinoma (36)
3.62E-051.359
Normal breast (61)
Invasive ductal breast carcinoma (389)
9.21E-101.287

[i] The number in the parenthesis indicates the patient number. ASL, argininosuccinate lyase; TCGA, The Cancer Genome Atlas.

Table II

ASL expression of the normal and breast cancer tissues from the Oncomine database.

Table II

ASL expression of the normal and breast cancer tissues from the Oncomine database.

Tissue (no.)P-valueFold-changeRef.
Normal breast (144)
Invasive lobular breast carcinoma (148)
2.36E-121.108Curtis et al(41)
Normal breast (144)
Invasive ductal breast carcinoma (1,556)
1.85E-281.111
Normal breast (144)
Breast carcinoma (14)
8.00E-031.056
Normal breast (144)
Medullary breast carcinoma (32)
9.13E-041.137
Normal breast (144)
Invasive ductal and invasive lobular breast carcinoma (90)
3.45E-061.07
Normal breast (144)
Mucinous breast carcinoma (46)
7.00E-031.049
Normal breast (144)
Ductal breast carcinoma in situ (10)
7.70E-021.067

[i] The number in the parenthesis indicates the patient number. ASL, argininosuccinate lyase.

ASL shRNA inhibits breast cancer cell growth

To study the role of ASL in breast cancer, MDA MB-231 cells were transfected with ASL shRNA and stable transfectants were selected by puromycin. The mRNA and protein levels of the ASL knockdown stable transfectants were determined by RT-PCR and western blotting (Fig. 2A). The proliferation of the ASL knockdown stable transfectants was determined by colony formation assay. ASL knockdown stable transfectants exhibited a significantly reduced proliferation in vitro (Fig. 2B). To investigate the effect of ASL on tumorigenic ability, the anchorage-independent growth ability of the ASL knockdown stable transfectants was analyzed. Decreased anchorage-independent growth ability was observed in the ASL knockdown stable transfectants of the MDA MB-231 cells (Fig. 2C). We further investigated the tumorigenicity of ASL knockdown stable transfectants in vivo and found that tumor growth was decreased by ASL shRNA (Fig. 2D). Moreover, there was a significant increase in the survival rate of the ASL knockdown stable transfectants when compare to that of the parental MDA MB-231 cells (Fig. 2E). The results indicated that ASL shRNA decreased cell growth in vivo and in vitro.

ASL shRNA inhibits cyclin A2 expression and causes G2/M cell cycle delay

To further study ASL shRNA-induced growth inhibition, we examined the changes in cell cycle progression using flow cytometric methods. A delay in G2/M phase was observed in the ASL knockdown stable transfectants (Fig. 3A). We then determined whether ASL shRNA affects the changes in cell cycle-associated molecules. Cyclin A2 was significantly reduced in the ASL knockdown transfectants, while cyclin D1 E1 and B1, CDK2 and CDK4 were not decreased (Fig. 3B). The mRNA expression of cyclins was not altered (Fig. 3C). Since the cyclins are frequently regulated by protein degradation, we ascertained whether the downregulation of cyclins by ASL shRNA could be restored by a proteasome inhibitor. Addition of MG132, a proteasome inhibitor, restored cyclin A2 protein expression (Fig. 3D).

Ectopic expression of cyclin A2 restores ASL shRNA-induced growth inhibition

Since ASL shRNA reduced cyclin A2 expression and cell growth, we next examined the role of cyclin A2 in the proliferation by ectopically expressing cyclin A2 in the ASL knockdown transfectants. Ectopic cyclin A2 reversed the cell growth inhibition by ASL shRNA, indicating that cyclin A2 plays an important role in the inhibition of cell growth by ASL shRNA (Fig. 4).

ASL shRNA induces autophagy in breast cancer cells

Autophagy is required for amino acid maintenance and responds to nitrogen deprivation in breast cancer (37). We next examined whether ASL shRNA induces autophagy. LC3B expression and autophagic vacuoles stained by MDC were increased in the ASL knockdown transfectants (Fig. 5A and B). To study the correlation between autophagy and cell growth, the autophagic inhibitors, 3-MA and bafilomycin A1, were incubated with the parental and ASL-knockdown MDA MB-231 cells.

The cell growth of the ASL knockdown transfectants was higher than that of the parental MDA MB-231 cells after treatment with the autophagic inhibitors, indicating the autophagy-induced pro-survival role by ASL shRNA (Fig. 5C). The cellular arginine level was analyzed by HPLC analysis. There was no significant difference in the arginine level between parental cells and the ASL knockdown transfectants (Fig. 5D). These data indicate that autophagy is induced independent of total cellular arginine content.

ASL shRNA attenuates NO content in breast cancer cells

Nitric oxide synthase (NOS) and ASL constitute the citrulline-argininosuccinate-arginine cycle and permit nitric oxide production. Therefore, we examined the NO content in ASL knockdown transfectants. The NO content was significantly decreased by ASL shRNA (Fig. 6A). The NO inhibitor, L-NMMA, also attenuated the NO level in the MDA MB-231 cells (Fig. 6B). Since NO production is implicated in cancer progression, we analyzed whether an NOS inhibitor attenuates breast cancer growth. The proliferation was inhibited by L-NMMA in MDA-MB-231 cells as demonstrated by colony formation (Fig. 6C) and MTT assays (Fig. 6D). The NO donor, sodium nitrite (NaNO2), partially restored the ASL shRNA-induced growth inhibition as demonstrated by colony formation (Fig. 6E) and MTT assays (Fig. 6F), indicating that reduction in the NO level may in part be responsible for the growth inhibition by ASL shRNA in breast cancer.

Discussion

In the present study, we demonstrated that ASL expression was induced by ER stress and was overexpressed in breast cancer. ASL shRNA attenuated cell proliferation and anchorage-independent growth. The breast cancer cells with low ASL expression had lower ability to form tumors in NOD/SCID mice. Furthermore, ASL downregulation induced autophagy. The cyclin A2 and NO levels in the ASL knockdown transfectants were decreased and ectopic cyclin A2 and NO donor restored the inhibition of cell growth by ASL shRNA. A similar effect of NO on cell proliferation was further observed using an NOS inhibitor, L-NMMA, suggesting that NO played an important role in the ASL knockdown transfectants.

ASL plays an important role in liver cancer progression (27). This finding demonstrating the effect of ASL on cancer cell growth in vivo and in vitro is consistent with the present study. Kaplan-Meier plotter database indicated that the breast cancer patients with high ASL expression were associated with a poor clinical outcome. ASL expression was also overexpressed in the breast cancer tissues in the Cancer Genome Anatomy Project (CGAP) database. Downregulation of ASL was found to contribute to tumor regression in both liver and breast cancer. These data indicate that ASL may play a tumorigenic role in human cancer, suggesting that ASL may serve as a therapeutic target. The role of ASL in other types of cancers warrants further study.

The mouse model with ASL deficiency has an effect on NO synthesis (25). The complex of ASL, ASS and NOS is responsible for NO recycling (38). Our data support the notion that the metabolic enzyme, ASL, contributes to NO production. Excessive NO production has been implicated in cancer progression (39). The NO donor restored the cell inhibition by ASL shRNA and the NOS inhibitor attenuated cell growth, indicating the oncogenic role of NO in cancer development. However, a previous study found that macrophage and natural killer cell-derived NO exerts an antitumor effect (40). The mechanism by which NO mediates cancer growth warrants further investigation.

In conclusion, ASL is overexpressed in breast cancer and ASL downregulation decreases tumor growth by inhibiting cyclin A2 and NO. Administration of ASL shRNA may be a novel treatment to prevent cancer cell proliferation and induce cancer cell death.

Acknowledgments

The present study was supported by a grant (to M.D. Lai) NSC-100-2325-B-006-008 from the National Science Council, Taiwan, and NHRI-EX100-9927B1 from the National Health Research Institute, Taiwan; to Establish Centers of Excellence for Cancer Research in Taiwan, DOH101-TD-C-111-003 Department of Health, Executive Yuan, Taiwan.

References

1 

McDonnell DP, Park S, Goulet MT, Jasper J, Wardell SE, Chang CY, Norris JD, Guyton JR and Nelson ER: Obesity, cholesterol metabolism, and breast cancer pathogenesis. Cancer Res. 74:4976–4982. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Stebbing J, Sharma A, North B, Athersuch TJ, Zebrowski A, Pchejetski D, Coombes RC, Nicholson JK and Keun HC: A metabolic phenotyping approach to understanding relationships between metabolic syndrome and breast tumour responses to chemotherapy. Ann Oncol. 23:860–866. 2012. View Article : Google Scholar

3 

Hirayama A, Kami K, Sugimoto M, Sugawara M, Toki N, Onozuka H, Kinoshita T, Saito N, Ochiai A, Tomita M, et al: Quantitative metabolome profiling of colon and stomach cancer microenvironment by capillary electrophoresis time-of-flight mass spectrometry. Cancer Res. 69:4918–4925. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Qiu F, Chen YR, Liu X, Chu CY, Shen LJ, Xu J, Gaur S, Forman HJ, Zhang H, Zheng S, et al: Arginine starvation impairs mitochondrial respiratory function in ASS1-deficient breast cancer cells. Sci Signal. 7:ra312014. View Article : Google Scholar : PubMed/NCBI

5 

Possemato R, Marks KM, Shaul YD, Pacold ME, Kim D, Birsoy K, Sethumadhavan S, Woo HK, Jang HG, Jha AK, et al: Functional genomics reveal that the serine synthesis pathway is essential in breast cancer. Nature. 476:346–350. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Amelio I, Cutruzzolá F, Antonov A, Agostini M and Melino G: Serine and glycine metabolism in cancer. Trends Biochem Sci. 39:191–198. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Wu G and Morris SM Jr: Arginine metabolism: Nitric oxide and beyond. Biochem J. 336:1–17. 1998. View Article : Google Scholar : PubMed/NCBI

8 

Vissers YL, Dejong CH, Luiking YC, Fearon KC, von Meyenfeldt MF and Deutz NE: Plasma arginine concentrations are reduced in cancer patients: Evidence for arginine deficiency? Am J Clin Nutr. 81:1142–1146. 2005.PubMed/NCBI

9 

Daly JM, Reynolds J, Thom A, Kinsley L, Dietrick-Gallagher M, Shou J and Ruggieri B: Immune and metabolic effects of arginine in the surgical patient. Ann Surg. 208:512–523. 1988. View Article : Google Scholar : PubMed/NCBI

10 

Talamonti MS, Kim SP, Yao KA, Wayne JD, Feinglass J, Bennett CL and Rao S: Surgical outcomes of patients with gastric carcinoma: The importance of primary tumor location and microvessel invasion. Surgery. 134:720–729. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Yerushalmi HF, Besselsen DG, Ignatenko NA, Blohm-Mangone KA, Padilla-Torres JL, Stringer DE, Guillen JM, Holubec H, Payne CM and Gerner EW: Role of polyamines in arginine-dependent colon carcinogenesis in ApcMin/+ mice. Mol Carcinog. 45:764–773. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Park KG, Heys SD, Blessing K, Kelly P, McNurlan MA, Eremin O and Garlick PJ: Stimulation of human breast cancers by dietary L-arginine. Clin Sci. 82:413–417. 1992. View Article : Google Scholar : PubMed/NCBI

13 

Yeatman TJ, Risley GL and Brunson ME: Depletion of dietary arginine inhibits growth of metastatic tumor. Arch Surg. 126:1376–1382. 1991. View Article : Google Scholar : PubMed/NCBI

14 

Ma Q, Wang Y, Gao X, Ma Z and Song Z: L-arginine reduces cell proliferation and ornithine decarboxylase activity in patients with colorectal adenoma and adenocarcinoma. Clin Cancer Res. 13:7407–7412. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Feun L, You M, Wu CJ, Kuo MT, Wangpaichitr M, Spector S and Savaraj N: Arginine deprivation as a targeted therapy for cancer. Curr Pharm Des. 14:1049–1057. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Phillips MM, Sheaff MT and Szlosarek PW: Targeting arginine-dependent cancers with arginine-degrading enzymes: Opportunities and challenges. Cancer Res Treat. 45:251–262. 2013. View Article : Google Scholar

17 

Delage B, Fennell DA, Nicholson L, McNeish I, Lemoine NR, Crook T and Szlosarek PW: Arginine deprivation and arginino-succinate synthetase expression in the treatment of cancer. Int J Cancer. 126:2762–2772. 2010.PubMed/NCBI

18 

Shen LJ and Shen WC: Drug evaluation: ADI-PEG-20 - a PEGylated arginine deiminase for arginine-auxotrophic cancers. Curr Opin Mol Ther. 8:240–248. 2006.PubMed/NCBI

19 

Shen LJ, Beloussow K and Shen WC: Modulation of arginine metabolic pathways as the potential anti-tumor mechanism of recombinant arginine deiminase. Cancer Lett. 231:30–35. 2006. View Article : Google Scholar

20 

Dillon BJ, Prieto VG, Curley SA, Ensor CM, Holtsberg FW, Bomalaski JS and Clark MA: Incidence and distribution of argininosuccinate synthetase deficiency in human cancers: A method for identifying cancers sensitive to arginine deprivation. Cancer. 100:826–833. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Ensor CM, Holtsberg FW, Bomalaski JS and Clark MA: Pegylated arginine deiminase (ADI-SS PEG20,000 mw) inhibits human melanomas and hepatocellular carcinomas in vitro and in vivo. Cancer Res. 62:5443–5450. 2002.PubMed/NCBI

22 

Bowles TL, Kim R, Galante J, Parsons CM, Virudachalam S, Kung HJ and Bold RJ: Pancreatic cancer cell lines deficient in argininosuccinate synthetase are sensitive to arginine deprivation by arginine deiminase. Int J Cancer. 123:1950–1955. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Yoon CY, Shim YJ, Kim EH, Lee JH, Won NH, Kim JH, Park IS, Yoon DK and Min BH: Renal cell carcinoma does not express argininosuccinate synthetase and is highly sensitive to arginine deprivation via arginine deiminase. Int J Cancer. 120:897–905. 2007. View Article : Google Scholar

24 

Feun LG, Marini A, Walker G, Elgart G, Moffat F, Rodgers SE, Wu CJ, You M, Wangpaichitr M, Kuo MT, et al: Negative argininosuccinate synthetase expression in melanoma tumours may predict clinical benefit from arginine-depleting therapy with pegylated arginine deiminase. Br J Cancer. 106:1481–1485. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Erez A, Nagamani SC, Shchelochkov OA, Premkumar MH, Campeau PM, Chen Y, Garg HK, Li L, Mian A, Bertin TK, et al: Requirement of argininosuccinate lyase for systemic nitric oxide production. Nat Med. 17:1619–1626. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Turner MA, Simpson A, McInnes RR and Howell PL: Human argininosuccinate lyase: A structural basis for intragenic complementation. Proc Natl Acad Sci USA. 94:9063–9068. 1997. View Article : Google Scholar : PubMed/NCBI

27 

Huang HL, Hsu HP, Shieh SC, Chang YS, Chen WC, Cho CY, Teng CF, Su IJ, Hung WC and Lai MD: Attenuation of argininosuccinate lyase inhibits cancer growth via cyclin A2 and nitric oxide. Mol Cancer Ther. 12:2505–2516. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Cunard R and Sharma K: The endoplasmic reticulum stress response and diabetic kidney disease. Am J Physiol Renal Physiol. 300:F1054–F1061. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Lee AH, Scapa EF, Cohen DE and Glimcher LH: Regulation of hepatic lipogenesis by the transcription factor XBP1. Science. 320:1492–1496. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Wang X, Eno CO, Altman BJ, Zhu Y, Zhao G, Olberding KE, Rathmell JC and Li C: ER stress modulates cellular metabolism. Biochem J. 435:285–296. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Clarke HJ, Chambers JE, Liniker E and Marciniak SJ: Endoplasmic reticulum stress in malignancy. Cancer Cell. 25:563–573. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Wang G, Yang ZQ and Zhang K: Endoplasmic reticulum stress response in cancer: Molecular mechanism and therapeutic potential. Am J Transl Res. 2:65–74. 2010.PubMed/NCBI

33 

Malhi H and Kaufman RJ: Endoplasmic reticulum stress in liver disease. J Hepatol. 54:795–809. 2011. View Article : Google Scholar

34 

Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, Tuncman G, Görgün C, Glimcher LH and Hotamisligil GS: Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 306:457–461. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Barbosa-Tessmann IP, Chen C, Zhong C, Schuster SM, Nick HS and Kilberg MS: Activation of the unfolded protein response pathway induces human asparagine synthetase gene expression. J Biol Chem. 274:31139–31144. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Okada T, Yoshida H, Akazawa R, Negishi M and Mori K: Distinct roles of activating transcription factor 6 (ATF6) and double-stranded RNA-activated protein kinase-like endoplasmic reticulum kinase (PERK) in transcription during the mammalian unfolded protein response. Biochem J. 366:585–594. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H and Levine B: Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature. 402:672–676. 1999. View Article : Google Scholar : PubMed/NCBI

38 

Oyadomari S, Gotoh T, Aoyagi K, Araki E, Shichiri M and Mori M: Coinduction of endothelial nitric oxide synthase and arginine recycling enzymes in aorta of diabetic rats. Nitric Oxide. 5:252–260. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Korde Choudhari S, Sridharan G, Gadbail A and Poornima V: Nitric oxide and oral cancer: A review. Oral Oncol. 48:475–483. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Lechner M, Lirk P and Rieder J: Inducible nitric oxide synthase (iNOS) in tumor biology: The two sides of the same coin. Semin Cancer Biol. 15:277–289. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, et al: The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 486:346–352. 2012.PubMed/NCBI

Related Articles

Journal Cover

December-2015
Volume 34 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang H, Chen W, Hsu H, Cho C, Hung YH, Wang C and Lai M: Argininosuccinate lyase is a potential therapeutic target in breast cancer. Oncol Rep 34: 3131-3139, 2015
APA
Huang, H., Chen, W., Hsu, H., Cho, C., Hung, Y., Wang, C., & Lai, M. (2015). Argininosuccinate lyase is a potential therapeutic target in breast cancer. Oncology Reports, 34, 3131-3139. https://doi.org/10.3892/or.2015.4280
MLA
Huang, H., Chen, W., Hsu, H., Cho, C., Hung, Y., Wang, C., Lai, M."Argininosuccinate lyase is a potential therapeutic target in breast cancer". Oncology Reports 34.6 (2015): 3131-3139.
Chicago
Huang, H., Chen, W., Hsu, H., Cho, C., Hung, Y., Wang, C., Lai, M."Argininosuccinate lyase is a potential therapeutic target in breast cancer". Oncology Reports 34, no. 6 (2015): 3131-3139. https://doi.org/10.3892/or.2015.4280