1
|
Piecyk M, Triki M, Laval P, Duret C, Fauvre J, Cussonneau L, Machon C, Guitton J, Rama N, Gibert B, Ichim G, Catez F, Bourdelais F, Durand S, Diaz J, Coste I, Renno T, Manié SN, Aznar N, Ansieau S, Ferraro‐Peyret C, Chaveroux C. The stress sensor GCN2 differentially controls ribosome biogenesis in colon cancer according to the nutritional context. Mol Oncol 2024; 18:2111-2135. [PMID: 37452637 PMCID: PMC11467793 DOI: 10.1002/1878-0261.13491] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/25/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023] Open
Abstract
Nutrient availability is a key determinant of tumor cell behavior. While nutrient-rich conditions favor proliferation and tumor growth, scarcity, and particularly glutamine starvation, promotes cell dedifferentiation and chemoresistance. Here, linking ribosome biogenesis plasticity with tumor cell fate, we uncover that the amino acid sensor general control non-derepressible 2 (GCN2; also known as eIF-2-alpha kinase 4) represses the expression of the precursor of ribosomal RNA (rRNA), 47S, under metabolic stress. We show that blockade of GCN2 triggers cell death by an irremediable nucleolar stress and subsequent TP53-mediated apoptosis in patient-derived models of colon adenocarcinoma (COAD). In nutrient-rich conditions, a cell-autonomous GCN2 activity supports cell proliferation by stimulating 47S rRNA transcription, independently of the canonical integrated stress response (ISR) axis. Impairment of GCN2 activity prevents nuclear translocation of methionyl-tRNA synthetase (MetRS), resulting in nucleolar stress, mTORC1 inhibition and, ultimately, autophagy induction. Inhibition of the GCN2-MetRS axis drastically improves the cytotoxicity of RNA polymerase I (RNA pol I) inhibitors, including the first-line chemotherapy oxaliplatin, on patient-derived COAD tumoroids. Our data thus reveal that GCN2 differentially controls ribosome biogenesis according to the nutritional context. Furthermore, pharmacological co-inhibition of the two GCN2 branches and RNA pol I activity may represent a valuable strategy for elimination of proliferative and metabolically stressed COAD cells.
Collapse
Affiliation(s)
- Marie Piecyk
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Mouna Triki
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Pierre‐Alexandre Laval
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Cedric Duret
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Joelle Fauvre
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Laura Cussonneau
- INRAE, Unité de Nutrition Humaine, UMR1019Université Clermont AuvergneClermont‐FerrandFrance
| | - Christelle Machon
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
- Biochemistry and Pharmaco‐Toxicology Laboratory, Lyon Sud HospitalHospices Civils de Lyon Pierre‐Bénite, University Hospital of LyonFrance
| | - Jerôme Guitton
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
- Biochemistry and Pharmaco‐Toxicology Laboratory, Lyon Sud HospitalHospices Civils de Lyon Pierre‐Bénite, University Hospital of LyonFrance
| | - Nicolas Rama
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Benjamin Gibert
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Gabriel Ichim
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Frederic Catez
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Fleur Bourdelais
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Sebastien Durand
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Jean‐Jacques Diaz
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Isabelle Coste
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Toufic Renno
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Serge N. Manié
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Nicolas Aznar
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Stephane Ansieau
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| | - Carole Ferraro‐Peyret
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
- Hospices Civils de Lyon, Plateforme AURAGENFrance
| | - Cedric Chaveroux
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon BérardUniversité de Lyon, Université Claude Bernard Lyon 1France
| |
Collapse
|
2
|
miR-34a and IRE1A/XBP-1(S) Form a Double-Negative Feedback Loop to Regulate Hypoxia-Induced EMT, Metastasis, Chemo-Resistance and Autophagy. Cancers (Basel) 2023; 15:cancers15041143. [PMID: 36831485 PMCID: PMC9954576 DOI: 10.3390/cancers15041143] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/01/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Tumor-associated hypoxia, i.e., decreased availability of oxygen, results in a poor clinical outcome since it promotes EMT, metastasis, and chemotherapy-resistance. We have previously identified p53 and its target miR-34a, as critical determinants of the effect of hypoxia on colorectal cancer (CRC). Here, we aimed to characterize mechanisms that contribute to the selective advantage of cells with loss of p53/miR-34a function in a hypoxic environment. Using in silico prediction, we identified XBP-1 and IRE1A as potential miR-34a targets. IRE1A and XBP-1 are central components of the unfolded protein response that is activated by ER stress, which is also induced in tumor cells as a response to harsh conditions surrounding tumors such as hypoxia and a limited supply of nutrients. Here we characterized the XBP-1(S) transcription factor and its regulator IRE1A as direct, conserved miR-34a targets in CRC cells. After hypoxia and DNA damage, IRE1A and XBP-1 were repressed by p53 in a miR-34a-dependent manner, whereas p53-deficient cells showed induction of IRE1A and XBP-1(S). Furthermore, miR-34a expression was directly suppressed by XBP-1(S). In p53-deficient CRC cells, hypoxia-induced EMT, migration, invasion, metastases formation, and resistance to 5-FU were dependent on IRE1A/XBP-1(S) activation. Hypoxia-induced autophagy was identified as an XBP-1(S)-dependent mediator of 5-FU resistance and was reversed by ectopic miR-34a expression. The HIF1A/IRE1A/XBP-1(S)/p53/miR-34a feedback loop described here represents a central regulator of the response to hypoxia and ER stress that maintains cellular homeostasis. In tumors, the inactivation of p53 and miR-34a may result in IRE1A/XPB-1(S)-mediated EMT and autophagy, which ultimately promotes metastasis and chemoresistance.
Collapse
|
3
|
Fletcher T, Thompson AJ, Ashrafian H, Darzi A. The measurement and modification of hypoxia in colorectal cancer: overlooked but not forgotten. Gastroenterol Rep (Oxf) 2022; 10:goac042. [PMID: 36032656 PMCID: PMC9406947 DOI: 10.1093/gastro/goac042] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/18/2022] [Accepted: 07/21/2022] [Indexed: 11/14/2022] Open
Abstract
Tumour hypoxia is the inevitable consequence of a tumour's rapid growth and disorganized, inefficient vasculature. The compensatory mechanisms employed by tumours, and indeed the absence of oxygen itself, hinder the ability of all treatment modalities. The clinical consequence is poorer overall survival, disease-free survival, and locoregional control. Recognizing this, clinicians have been attenuating the effect of hypoxia, primarily with hypoxic modification or with hypoxia-activated pro-drugs, and notable success has been demonstrated. However, in the case of colorectal cancer (CRC), there is a general paucity of knowledge and evidence surrounding the measurement and modification of hypoxia, and this is possibly due to the comparative inaccessibility of such tumours. We specifically review the role of hypoxia in CRC and focus on the current evidence for the existence of hypoxia in CRC, the majority of which originates from indirect positron emission topography imaging with hypoxia selective radiotracers; the evidence correlating CRC hypoxia with poorer oncological outcome, which is largely based on the measurement of hypoxia inducible factor in correlation with clinical outcome; the evidence of hypoxic modification in CRC, of which no direct evidence exists, but is reflected in a number of indirect markers; the prognostic and monitoring implications of accurate CRC hypoxia quantification and its potential in the field of precision oncology; and the present and future imaging tools and technologies being developed for the measurement of CRC hypoxia, including the use of blood-oxygen-level-dependent magnetic resonance imaging and diffuse reflectance spectroscopy.
Collapse
Affiliation(s)
- Teddy Fletcher
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| | - Alex J Thompson
- The Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK
| | - Hutan Ashrafian
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| | - Ara Darzi
- Department of Surgery and Cancer, Queen Elizabeth the Queen Mother Wing, St Mary’s Hospital, Imperial College London, London, UK
| |
Collapse
|
4
|
Liu S, Washio J, Sato S, Abiko Y, Shinohara Y, Kobayashi Y, Otani H, Sasaki S, Wang X, Takahashi N. Rewired Cellular Metabolic Profiles in Response to Metformin under Different Oxygen and Nutrient Conditions. Int J Mol Sci 2022; 23:ijms23020989. [PMID: 35055173 PMCID: PMC8781974 DOI: 10.3390/ijms23020989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Metformin is a metabolic disruptor, and its efficacy and effects on metabolic profiles under different oxygen and nutrient conditions remain unclear. Therefore, the present study examined the effects of metformin on cell growth, the metabolic activities and consumption of glucose, glutamine, and pyruvate, and the intracellular ratio of nicotinamide adenine dinucleotide (NAD+) and reduced nicotinamide adenine dinucleotide (NADH) under normoxic (21% O2) and hypoxic (1% O2) conditions. The efficacy of metformin with nutrient removal from culture media was also investigated. The results obtained show that the efficacy of metformin was closely associated with cell types and environmental factors. Acute exposure to metformin had no effect on lactate production from glucose, glutamine, or pyruvate, whereas long-term exposure to metformin increased the consumption of glucose and pyruvate and the production of lactate in the culture media of HeLa and HaCaT cells as well as the metabolic activity of glucose. The NAD+/NADH ratio decreased during growth with metformin regardless of its efficacy. Furthermore, the inhibitory effects of metformin were enhanced in all cell lines following the removal of glucose or pyruvate from culture media. Collectively, the present results reveal that metformin efficacy may be regulated by oxygen conditions and nutrient availability, and indicate the potential of the metabolic switch induced by metformin as combinational therapy.
Collapse
Affiliation(s)
- Shan Liu
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai 9808575, Japan; (S.L.); (S.S.); (Y.A.); (Y.S.); (Y.K.); (H.O.); (S.S.); (N.T.)
- Department of Head and Neck Oncology, Sichuan University West China School of Stomatology, Chengdu 610041, China;
| | - Jumpei Washio
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai 9808575, Japan; (S.L.); (S.S.); (Y.A.); (Y.S.); (Y.K.); (H.O.); (S.S.); (N.T.)
- Correspondence: ; Tel.: +81-22-717-8295
| | - Satoko Sato
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai 9808575, Japan; (S.L.); (S.S.); (Y.A.); (Y.S.); (Y.K.); (H.O.); (S.S.); (N.T.)
| | - Yuki Abiko
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai 9808575, Japan; (S.L.); (S.S.); (Y.A.); (Y.S.); (Y.K.); (H.O.); (S.S.); (N.T.)
| | - Yuta Shinohara
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai 9808575, Japan; (S.L.); (S.S.); (Y.A.); (Y.S.); (Y.K.); (H.O.); (S.S.); (N.T.)
| | - Yuri Kobayashi
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai 9808575, Japan; (S.L.); (S.S.); (Y.A.); (Y.S.); (Y.K.); (H.O.); (S.S.); (N.T.)
| | - Haruki Otani
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai 9808575, Japan; (S.L.); (S.S.); (Y.A.); (Y.S.); (Y.K.); (H.O.); (S.S.); (N.T.)
| | - Shiori Sasaki
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai 9808575, Japan; (S.L.); (S.S.); (Y.A.); (Y.S.); (Y.K.); (H.O.); (S.S.); (N.T.)
| | - Xiaoyi Wang
- Department of Head and Neck Oncology, Sichuan University West China School of Stomatology, Chengdu 610041, China;
| | - Nobuhiro Takahashi
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai 9808575, Japan; (S.L.); (S.S.); (Y.A.); (Y.S.); (Y.K.); (H.O.); (S.S.); (N.T.)
| |
Collapse
|
5
|
Zhao S, Zhou L, Dicker DT, Lev A, Zhang S, Ross E, El-Deiry WS. Anti-cancer efficacy including Rb-deficient tumors and VHL-independent HIF1α proteasomal destabilization by dual targeting of CDK1 or CDK4/6 and HSP90. Sci Rep 2021; 11:20871. [PMID: 34686682 PMCID: PMC8536770 DOI: 10.1038/s41598-021-00150-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 09/27/2021] [Indexed: 12/23/2022] Open
Abstract
A prevalent characteristic of solid tumors is intra-tumoral hypoxia. Hypoxia-inducible factor 1α (HIF1α) predominantly mediates the adaptive response to O2 oscillation and is linked to multiple malignant hallmarks. Here we describe a strategy to robustly target HIF1α by dual inhibition of CDK(s) and heat shock protein 90 (HSP90). We show that CDK1 may contribute to HSP90-mediated HIF1α stabilization. CDK1 knockdown enhances the decrease of HIF1α by HSP90 inhibition. Dual inhibition of CDK1 and HSP90 significantly increases apoptosis and synergistically inhibits cancer cell viability. Similarly, targeting CDK4/6 using FDA-approved inhibitors in combination with HSP90 inhibition shows a class effect on HIF1α inhibition and cancer cell viability suppression not only in colorectal but also in various other cancer types, including Rb-deficient cancer cells. Dual inhibition of CDK4/6 and HSP90 suppresses tumor growth in vivo. In summary, combined targeting of CDK(s) (CDK1 or CDK4/6) and HSP90 remarkably inhibits the expression level of HIF1α and shows promising anti-cancer efficacy with therapeutic potential.
Collapse
Affiliation(s)
- Shuai Zhao
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Pathobiology Graduate Program, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - David T Dicker
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA
| | - Avital Lev
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA.,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Eric Ross
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, USA. .,Pathobiology Graduate Program, Brown University, Providence, RI, USA. .,Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA. .,Joint Program in Cancer Biology, Brown University and Lifespan Cancer Institute, Providence, RI, USA. .,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, USA. .,Fox Chase Cancer Center, Philadelphia, PA, USA. .,Hematology/Oncology Division, Lifespan Cancer Institute, Providence, RI, USA.
| |
Collapse
|
6
|
Schito L, Rey S, Xu P, Man S, Cruz‐Muñoz W, Kerbel RS. Metronomic chemotherapy offsets HIFα induction upon maximum-tolerated dose in metastatic cancers. EMBO Mol Med 2020; 12:e11416. [PMID: 32686360 PMCID: PMC7507002 DOI: 10.15252/emmm.201911416] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 06/21/2020] [Accepted: 06/23/2020] [Indexed: 12/22/2022] Open
Abstract
Conventional maximum-tolerated dose (MTD) chemotherapy relies on periodic, massive cancer cell ablation events followed by treatment-free intermissions, stereotypically resulting in resistance, relapse, and mortality. Furthermore, MTD chemotherapy can promote metastatic dissemination via activation of a transcriptional program dependent on hypoxia-inducible factor (HIF)-1α and (HIF)-2α (hereafter referred to as HIFα). Instead, frequent low-dose metronomic (LDM) chemotherapy displays less adverse effects while preserving significant pre-clinical anticancer activity. Consequently, we hereby compared the effect of MTD or LDM chemotherapy upon HIFα in models of advanced, metastatic colon and breast cancer. Our results revealed that LDM chemotherapy could offset paralog-specific, MTD-dependent HIFα induction in colon cancers disseminating to the liver and lungs, while limiting HIFα and hypoxia in breast cancer lung metastases. Moreover, we assessed the translational significance of HIFα activity in colorectal and breast TCGA/microarray data, by developing two compact, 11-gene transcriptomic signatures allowing the stratification/identification of patients likely to benefit from LDM and/or HIFα-targeting therapies. Altogether, these results suggest LDM chemotherapy as a potential maintenance strategy to stave off HIFα induction within the intra-metastatic tumor microenvironment.
Collapse
Affiliation(s)
- Luana Schito
- UCD School of MedicineUniversity College DublinDublin 4Ireland
- UCD Conway Institute of Biomolecular & Biomedical ResearchUniversity College DublinBelfield, Dublin 4Ireland
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - Sergio Rey
- UCD School of MedicineUniversity College DublinDublin 4Ireland
- UCD Conway Institute of Biomolecular & Biomedical ResearchUniversity College DublinBelfield, Dublin 4Ireland
| | - Ping Xu
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - Shan Man
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - William Cruz‐Muñoz
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - Robert S Kerbel
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
| |
Collapse
|
7
|
Casciello F, Al-Ejeh F, Miranda M, Kelly G, Baxter E, Windloch K, Gannon F, Lee JS. G9a-mediated repression of CDH10 in hypoxia enhances breast tumour cell motility and associates with poor survival outcome. Am J Cancer Res 2020; 10:4515-4529. [PMID: 32292512 PMCID: PMC7150496 DOI: 10.7150/thno.41453] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: Epigenetic mechanisms are fundamental processes that can modulate gene expression, allowing cellular adaptation to environmental conditions. Hypoxia is an important factor known to initiate the metastatic cascade in cancer, activating cell motility and invasion by silencing cell adhesion genes. G9a is a histone methyltransferase previously shown to accumulate in hypoxic conditions. While its oncogenic activity has been previously reported, not much is known about the role G9a plays in the hypoxia-mediated metastatic cascade. Methods: The role of G9a in cell motility in hypoxic condition was determined by inhibiting G9a either by short-hairpin mediated knock down or pharmacologically using a small molecule inhibitor. Through gene expression profiling, we identified CDH10 to be an important G9a target that regulates breast cancer cell motility. Lung metastasis assay in mice was used to determine the physiological significance of G9a. Results: We demonstrate that, while hypoxia enhances breast cancer migratory capacity, blocking G9a severely reduces cellular motility under both normoxic and hypoxic conditions and prevents the hypoxia-mediated induction of cellular movement. Moreover, inhibition of G9a histone methyltransferase activity in mice using a specific small molecule inhibitor significantly reduced growth and colonisation of breast cancer cells in the lung. We identify the type-II cadherin CDH10 as being a novel hypoxia-dependent gene, directly repressed by G9a through histone methylation. CDH10 overexpression significantly reduces cellular movements in breast cancer cell lines and prevents the hypoxia-mediated increase in cell motility. In addition, we show that CDH10 expression is prognostic in breast cancer and that it is inversely correlated to EHMT2 (G9a) transcript levels in many tumor-types, including breast cancer. Conclusion: We propose that G9a promotes cellular motility during hypoxic stress through the silencing of the cell adhesion molecule CDH10 and we describe CDH10 as a novel prognostic biomarker for breast cancer.
Collapse
|
8
|
Yang Y, Qu A, Wu Q, Zhang X, Wang L, Li C, Dong Z, Du L, Wang C. Prognostic value of a hypoxia-related microRNA signature in patients with colorectal cancer. Aging (Albany NY) 2020; 12:35-52. [PMID: 31926112 PMCID: PMC6977676 DOI: 10.18632/aging.102228] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/21/2019] [Indexed: 12/24/2022]
Abstract
Hypoxia has been particularly associated with poor prognosis in cancer patients. Recent studies have suggested that hypoxia-related miRNAs play a critical role in various cancers, including colorectal cancer (CRC). In the present study, we found 52 differentially expressed miRNAs in HT-29 cells under hypoxic conditions versus normoxic conditions by analyzing the profiles of miRNAs. Using Cox model, we developed a hypoxia-related miRNA signature consisting of four miRNAs, which could successfully discriminate high-risk patients in the Cancer Genome Atlas (TCGA) training cohort (n=381). The prognostic value of this signature was further confirmed in the TCGA testing cohort (n=190) and an independent validation cohort composed of formalin-fixed paraffin-embedded clinical CRC samples (n=220), respectively. Multivariable Cox regression and stratified survival analysis revealed this signature was an independent prognostic factor for CRC patients. Time-dependent receiver operating characteristic (ROC) analysis showed that the area under the curve (AUC) of this signature was significantly larger than that of any other clinical risk factors or single miRNA alone. A nomogram was constructed for clinical use, which incorporated both the miRNA signature and clinical risk factors and performed well in the calibration plots. Collectively, this novel hypoxia-related miRNA signature was an independent prognostic factor, and it possessed a stronger predictive power in identifying high-risk CRC patients than currently used clinicopathological features.
Collapse
Affiliation(s)
- Yongmei Yang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China.,Key Laboratory of Tumor Marker Translational Medicine, Shandong Provincial Medicine and Health, Jinan 250012, Shandong Province, China
| | - Ailin Qu
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China.,Key Laboratory of Tumor Marker Translational Medicine, Shandong Provincial Medicine and Health, Jinan 250012, Shandong Province, China
| | - Qi Wu
- Department of Blood Transfusion, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China.,Key Laboratory of Tumor Marker Translational Medicine, Shandong Provincial Medicine and Health, Jinan 250012, Shandong Province, China
| | - Lili Wang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China.,Key Laboratory of Tumor Marker Translational Medicine, Shandong Provincial Medicine and Health, Jinan 250012, Shandong Province, China
| | - Chen Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong Province, China
| | - Zhaogang Dong
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China.,Key Laboratory of Tumor Marker Translational Medicine, Shandong Provincial Medicine and Health, Jinan 250012, Shandong Province, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong Province, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong Province, China
| |
Collapse
|
9
|
Olivares-Marin IK, González-Hernández JC, Madrigal-Perez LA. Resveratrol cytotoxicity is energy-dependent. J Food Biochem 2019; 43:e13008. [PMID: 31385323 DOI: 10.1111/jfbc.13008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/03/2019] [Accepted: 07/21/2019] [Indexed: 12/16/2022]
Abstract
Resveratrol is a phytochemical that may promote health. However, it has also been reported to be a toxic compound. The molecular mechanism by which resveratrol acts remains unclear. The inhibition of the oxidative phosphorylation (OXPHOS) pathway appears to be the molecular mechanism of resveratrol. Taking this into account, we propose that the cytotoxic properties of resveratrol depend on the energy (e.g., carbohydrates, lipids, and proteins) availability in the cells. In this regard, in a condition with low energy accessibility, resveratrol could enhance ATP starvation to lethal levels. In contrast, when cells are supplemented with high quantities of energy and resveratrol, the inhibition of OXPHOS might produce a low-energy environment, mimicking the beneficial effects of caloric restriction. This review suggests that investigating a possible complex relationship between caloric intake and the differential effects of resveratrol on OXPHOS may be justified. PRACTICAL APPLICATIONS: A low-calorie diet accompanied by significant levels of resveratrol might modify cellular bioenergetics, which could impact cellular viability and enhance the anti-cancer properties of resveratrol.
Collapse
Affiliation(s)
| | | | - Luis Alberto Madrigal-Perez
- Laboratorio de Biotecnología Microbiana, Instituto Tecnológico Superior de Ciudad Hidalgo, Ciudad Hidalgo, Mexico
| |
Collapse
|
10
|
Abstract
PURPOSE DLG7 (disc large homolog 7) is a microtubule-associated protein encoded by DLGAP5 (DLG associated protein 5) gene and has an important role during spindle assembly. Spindle assembly deregulation is a well-known cause of genomic instability. The aim of this study was to investigate the influence of DLGAP5 expression on survival and to evaluate its potential use as a biomarker in colorectal cancer (CRC). METHODS DLGAP5 expression was measured in the primary tumor and corresponding normal mucosa samples from 109 patients with CRC and correlated to clinical and pathological data. The results were validated in a second, publically available patient cohort. Molecular effects of DLG7/DLGAP5 in CRC were analyzed via functional assays in knockdown cell lines. RESULTS DLGAP5 downregulation led to a significant reduction of the invasion and migration potential in CRC. In addition, DLGAP5 expression correlates with nodal status and advanced UICC stage (III-IV).Subgroup analyses revealed a correlation between DLGAP5 overexpression and poor survival in patients with non-metastatic disease (M0). Furthermore, overexpression of DLGAP5 is associated with worse overall survival in distinct molecular CRC subtypes. CONCLUSIONS The results of this study suggest the importance of DLGAP5 in defining a more aggressive CRC phenotype. DLG7/DLGAP5 represents a potential biomarker for CRC in molecular subgroups of CRC.
Collapse
|
11
|
Vyas M, Patel N, Celli R, Wajapeyee N, Jain D, Zhang X. Glucose Metabolic Reprogramming and Cell Proliferation Arrest in Colorectal Micropapillary Carcinoma. Gastroenterology Res 2019; 12:128-134. [PMID: 31236153 PMCID: PMC6575135 DOI: 10.14740/gr1145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/30/2019] [Indexed: 12/24/2022] Open
Abstract
Background Micropapillary carcinoma (MPC) has been reported as an aggressive variant of colorectal carcinoma (CRC) associated with frequent lymphovascular invasion and poor outcome. Altered glycogen metabolism by metabolic reprogramming plays a critical role for cancer cell growth and survival. We aimed to investigate glucose metabolic reprogramming in colorectal MPC. Methods Immmunostains for Ki-67 and glucose transporter 1 (GLUT1) were performed on 10 colorectal MPCs. Real-time PCR analysis of expressions of GLUT1 and glycogen metabolizing enzymes: glycogen synthase (GYS1) and glycogen phosphorylase (PYGL) was performed on cultured monolayer and three-dimensional (3D) spheroid HCT116 colon cancer cells. Results GLUT1 was strongly expressed in MPC as compared to adjacent conventional glandular component, and was also significantly increased expression in 3D spheroids. Upregulation of GYS1 and PYGL was markedly increased in 3D spheroids. The proliferation rate (Ki-67) of MPC was significantly lower compared to conventional glandular component. The 3D spheroids showed increased cell cycle arrest. Our results demonstrate altered glycogen metabolism in colorectal MPC. Conclusion The reprogramming of glycogen metabolism in MPC provides a source of energy contributing to tumor cell survival in a low proliferation state. Targeting glucose-regulated metabolism may warrant consideration as possible MPC therapies.
Collapse
Affiliation(s)
- Monika Vyas
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natalie Patel
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Romulo Celli
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Narendra Wajapeyee
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Dhanpat Jain
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
12
|
Feng Y, Li Y, Li L, Wang X, Chen Z. Identification of specific modules and significant genes associated with colon cancer by weighted gene co‑expression network analysis. Mol Med Rep 2019; 20:693-700. [PMID: 31180534 DOI: 10.3892/mmr.2019.10295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/12/2019] [Indexed: 11/06/2022] Open
Abstract
Colon cancer is one of the most commonly diagnosed malignancies and is a leading cause of cancer‑associated mortality. The aim of the present study was to investigate the molecular mechanisms underlying colon cancer and identify potentially significant genes associated with the disease using weighted gene co‑expression network analysis (WGCNA). The test datasets used were downloaded from The Cancer Genome Atlas (TCGA) database. WGCNA was applied to analyze microarray data obtained from colon adenocarcinoma samples to identify significant modules and highly associated genes. A gene co‑expression network was constructed and different gene modules were selected. Functional and pathway enrichment analyses were performed to investigate the molecular mechanisms of colon cancer. In addition, highly connected hub genes associated with the most significant module were selected for further analysis. Nine specific modules associated with colon cancer were identified, of which the turquoise module was observed to exhibit the greatest association with the disease. Pathway enrichment analysis of the turquoise module suggested that genes in the turquoise module were associated with 'RNA polymerase' and 'purine metabolism'. Furthermore, gene ontology enrichment analysis revealed the top 30 hub genes with a higher degree in the turquoise module, such as σ‑non‑opioid intracellular receptor 1, transmembrane protein 147 TMEM147) and carbamoyl‑phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase, were predominantly enriched in the biological processes 'translation' and 'gene expression'. Experimental verification demonstrated that the expression of TMEM147 in colon cancer was significantly increased compared with the control. Therefore, the results suggested that genes associated with RNA polymerase and the purine metabolic pathways may be substantially involved in the pathogenesis of colon cancer. Furthermore, TMEM147 may represent a biomarker for colon cancer.
Collapse
Affiliation(s)
- Ye Feng
- Department of Gastrointestinal Colorectal and Anal Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yanbo Li
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lin Li
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xuefeng Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhi Chen
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
13
|
Han Y, Jo H, Cho JH, Dhanasekaran DN, Song YS. Resveratrol as a Tumor-Suppressive Nutraceutical Modulating Tumor Microenvironment and Malignant Behaviors of Cancer. Int J Mol Sci 2019; 20:925. [PMID: 30791624 PMCID: PMC6412705 DOI: 10.3390/ijms20040925] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor-suppressive effects of resveratrol have been shown in various types of cancer. However, regulation of tumor microenvironment by resveratrol is still unclear. Recent findings suggest resveratrol can potentiate its tumor-suppressive effect through modulation of the signaling pathways of cellular components (fibroblasts, macrophages and T cells). Also, studies have shown that resveratrol can suppress malignant phenotypes of cancer cells acquired in response to stresses of the tumor microenvironment, such as hypoxia, oxidative stress and inflammation. We discuss the effects of resveratrol on cancer cells in stress environment of tumors as well as interactions between cancer cells and non-cancer cells in this review.
Collapse
Affiliation(s)
- Youngjin Han
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - HyunA Jo
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - Jae Hyun Cho
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma city, OK 73012, USA.
| | - Yong Sang Song
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea.
| |
Collapse
|
14
|
Liu D. Concomitant dysregulation of the estrogen receptor and BRAF/MEK signaling pathways is common in colorectal cancer and predicts a worse prognosis. Cell Oncol (Dordr) 2019; 42:197-209. [PMID: 30645729 DOI: 10.1007/s13402-018-00420-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2018] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Recurrence is a major cause of colorectal cancer (CRC)-related death. As yet, the accurate identification of CRC patients at high risk of recurrence is still a major clinical challenge. Previously, we found that an estrogen receptor (ER) pathway gene signature may predict disease recurrence in CRC patients. The aim of this study is to evaluate the potential application of additional pathway-specific gene signatures in the prediction of CRC recurrence. METHODS The activities of 26 cancer-related pathways in CRC were semi-quantified using gene signature-based Bayesian binary regression analysis, and putative associations of the pathways with cancer recurrence risk were assessed using survival analysis. RESULTS Among the 26 pathways tested, inactivation of the estrogen receptor (ER) pathway was found to be one of the most common events in CRC. Inactivation of this pathway was found to be frequently accompanied by over-activation of the BRAF/MEK pathway, and these two pathways were found to be associated with opposite effects on several clinicopathological CRC features, including microsatellite instability, subsite location, advanced stage and recurrence. Survival analysis of four independent CRC patient cohorts revealed that while the BRAF/MEK pathway was more strongly associated with recurrence than the ER pathway in mixed-stage CRCs, the ER pathway was a better predictor of recurrence than the BRAF/MEK pathway in stage II CRC. A combined use of these two pathways improved the prediction of CRC recurrence in both mixed stage CRC (n = 1122; overall HR: 2.518, 95% CI: 1.570-4.038, p < 0.001) and stage II CRC (n = 535; overall HR: 1.976, 95% CI: 1.306-2.989, p = 0.001). CONCLUSIONS Combined activity of the ER and BRAF/MEK pathways may represent a novel biomarker for CRC prognosis and clinical management.
Collapse
Affiliation(s)
- Dingxie Liu
- Bluewater Biotech LLC, PO Box 1010, New Providence, NJ, 07974, USA.
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
15
|
Greenhough A, Bagley C, Heesom KJ, Gurevich DB, Gay D, Bond M, Collard TJ, Paraskeva C, Martin P, Sansom OJ, Malik K, Williams AC. Cancer cell adaptation to hypoxia involves a HIF-GPRC5A-YAP axis. EMBO Mol Med 2018; 10:e8699. [PMID: 30143543 PMCID: PMC6220329 DOI: 10.15252/emmm.201708699] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 02/06/2023] Open
Abstract
Hypoxia is a hallmark of solid tumours and a key physiological feature distinguishing cancer from normal tissue. However, a major challenge remains in identifying tractable molecular targets that hypoxic cancer cells depend on for survival. Here, we used SILAC-based proteomics to identify the orphan G protein-coupled receptor GPRC5A as a novel hypoxia-induced protein that functions to protect cancer cells from apoptosis during oxygen deprivation. Using genetic approaches in vitro and in vivo, we reveal HIFs as direct activators of GPRC5A transcription. Furthermore, we find that GPRC5A is upregulated in the colonic epithelium of patients with mesenteric ischaemia, and in colorectal cancers high GPRC5A correlates with hypoxia gene signatures and poor clinical outcomes. Mechanistically, we show that GPRC5A enables hypoxic cell survival by activating the Hippo pathway effector YAP and its anti-apoptotic target gene BCL2L1 Importantly, we show that the apoptosis induced by GPRC5A depletion in hypoxia can be rescued by constitutively active YAP. Our study identifies a novel HIF-GPRC5A-YAP axis as a critical mediator of the hypoxia-induced adaptive response and a potential target for cancer therapy.
Collapse
Affiliation(s)
- Alexander Greenhough
- Cancer Research UK Colorectal Tumour Biology Group, School of Cellular & Molecular Medicine, Faculty of Life Sciences University of Bristol, Bristol, UK
- Cancer Epigenetics Laboratory, School of Cellular & Molecular Medicine, Faculty of Life Sciences University of Bristol, Bristol, UK
| | - Clare Bagley
- Cancer Research UK Colorectal Tumour Biology Group, School of Cellular & Molecular Medicine, Faculty of Life Sciences University of Bristol, Bristol, UK
| | - Kate J Heesom
- Proteomics Facility, Faculty of Life Sciences University of Bristol, Bristol, UK
| | - David B Gurevich
- School of Biochemistry, Faculty of Life Sciences University of Bristol, Bristol, UK
| | - David Gay
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Mark Bond
- School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Tracey J Collard
- Cancer Research UK Colorectal Tumour Biology Group, School of Cellular & Molecular Medicine, Faculty of Life Sciences University of Bristol, Bristol, UK
| | - Chris Paraskeva
- Cancer Research UK Colorectal Tumour Biology Group, School of Cellular & Molecular Medicine, Faculty of Life Sciences University of Bristol, Bristol, UK
| | - Paul Martin
- School of Biochemistry, Faculty of Life Sciences University of Bristol, Bristol, UK
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences University of Bristol, Bristol, UK
- School of Medicine, Cardiff University, Cardiff, UK
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Karim Malik
- Cancer Epigenetics Laboratory, School of Cellular & Molecular Medicine, Faculty of Life Sciences University of Bristol, Bristol, UK
| | - Ann C Williams
- Cancer Research UK Colorectal Tumour Biology Group, School of Cellular & Molecular Medicine, Faculty of Life Sciences University of Bristol, Bristol, UK
| |
Collapse
|
16
|
Haynes J, McKee TD, Haller A, Wang Y, Leung C, Gendoo DMA, Lima-Fernandes E, Kreso A, Wolman R, Szentgyorgyi E, Vines DC, Haibe-Kains B, Wouters BG, Metser U, Jaffray DA, Smith M, O'Brien CA. Administration of Hypoxia-Activated Prodrug Evofosfamide after Conventional Adjuvant Therapy Enhances Therapeutic Outcome and Targets Cancer-Initiating Cells in Preclinical Models of Colorectal Cancer. Clin Cancer Res 2018; 24:2116-2127. [PMID: 29476017 DOI: 10.1158/1078-0432.ccr-17-1715] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 12/21/2017] [Accepted: 02/19/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Cancer-initiating cells (C-IC) have been described in multiple cancer types, including colorectal cancer. C-ICs are defined by their capacity to self-renew, thereby driving tumor growth. C-ICs were initially thought to be static entities; however, recent studies have determined these cells to be dynamic and influenced by microenvironmental cues such as hypoxia. If hypoxia drives the formation of C-ICs, then therapeutic targeting of hypoxia could represent a novel means to target C-ICs.Experimental Design: Patient-derived colorectal cancer xenografts were treated with evofosfamide, a hypoxia-activated prodrug (HAP), in combination with 5-fluorouracil (5-FU) or chemoradiotherapy (5-FU and radiation; CRT). Treatment groups included both concurrent and sequential dosing regimens. Effects on the colorectal cancer-initiating cell (CC-IC) fraction were assessed by serial passage in vivo limiting dilution assays. FAZA-PET imaging was utilized as a noninvasive method to assess intratumoral hypoxia.Results: Hypoxia was sufficient to drive the formation of CC-ICs and colorectal cancer cells surviving conventional therapy were more hypoxic and C-IC-like. Using a novel approach to combination therapy, we show that sequential treatment with 5-FU or CRT followed by evofosfamide not only inhibits tumor growth of xenografts compared with 5-FU or CRT alone, but also significantly decreases the CC-IC fraction. Furthermore, noninvasive FAZA-PET hypoxia imaging was predictive of a tumor's response to evofosfamide.Conclusions: Our data demonstrate a novel means to target the CC-IC fraction by adding a HAP sequentially after conventional adjuvant therapy, as well as the use of FAZA-PET as a biomarker for hypoxia to identify tumors that will benefit most from this approach. Clin Cancer Res; 24(9); 2116-27. ©2018 AACR.
Collapse
Affiliation(s)
- Jennifer Haynes
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Trevor D McKee
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,STTARR Innovation Centre, University Health Network, Toronto, Ontario, Canada
| | - Andrew Haller
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yadong Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Cherry Leung
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Deena M A Gendoo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | - Antonija Kreso
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Robin Wolman
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Eva Szentgyorgyi
- Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - Douglass C Vines
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,STTARR Innovation Centre, University Health Network, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Bradly G Wouters
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Ur Metser
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada.,Techna Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - David A Jaffray
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,STTARR Innovation Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Techna Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - Myles Smith
- Department of Surgery, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Catherine A O'Brien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Ullmann P, Qureshi-Baig K, Rodriguez F, Ginolhac A, Nonnenmacher Y, Ternes D, Weiler J, Gäbler K, Bahlawane C, Hiller K, Haan S, Letellier E. Hypoxia-responsive miR-210 promotes self-renewal capacity of colon tumor-initiating cells by repressing ISCU and by inducing lactate production. Oncotarget 2018; 7:65454-65470. [PMID: 27589845 PMCID: PMC5323168 DOI: 10.18632/oncotarget.11772] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/25/2016] [Indexed: 01/01/2023] Open
Abstract
Low oxygen concentrations (hypoxia) are known to affect the cellular metabolism and have been suggested to regulate a subpopulation of cancer cells with tumorigenic properties, the so-called tumor-initiating cells (TICs). To better understand the mechanism of hypoxia-induced TIC activation, we set out to study the role of hypoxia-responsive miRNAs in recently established colon cancer patient-derived TICs. We were able to show that low oxygen concentrations consistently lead to the upregulation of miR-210 in different primary TIC-enriched cultures. Both stable overexpression of miR-210 and knockdown of its target gene ISCU resulted in enhanced TIC self-renewal. We could validate the tumorigenic properties of miR- 210 in in vivo experiments by showing that ectopic expression of miR-210 results in increased tumor incidence. Furthermore, enhanced miR-210 expression correlated with reduced TCA cycle activity and increased lactate levels. Importantly, by blocking lactate production via inhibition of LDHA, we could reverse the promoting effect of miR-210 on self-renewal capacity, thereby emphasizing the regulatory impact of the glycolytic phenotype on colon TIC properties. Finally, by assessing expression levels in patient tissue, we could demonstrate the clinical relevance of the miR-210/ISCU signaling axis for colorectal carcinoma. Taken together, our study highlights the importance of hypoxia-induced miR-210 in the regulation of colon cancer initiation.
Collapse
Affiliation(s)
- Pit Ullmann
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Komal Qureshi-Baig
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Fabien Rodriguez
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Aurélien Ginolhac
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | | | - Dominik Ternes
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Jil Weiler
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Karoline Gäbler
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Christelle Bahlawane
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Karsten Hiller
- Luxembourg Centre for Systems Biomedicine, L-4367 Belvaux, Luxembourg
| | - Serge Haan
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Elisabeth Letellier
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| |
Collapse
|
18
|
Zhang WJ, Chen C, Zhou ZH, Gao ST, Tee TJ, Yang LQ, Xu YY, Pang TH, Xu XY, Sun Q, Feng M, Wang H, Lu CL, Wu GZ, Wu S, Guan WX, Xu GF. Hypoxia-inducible factor-1 alpha Correlates with Tumor-Associated Macrophages Infiltration, Influences Survival of Gastric Cancer Patients. J Cancer 2017; 8:1818-1825. [PMID: 28819379 PMCID: PMC5556645 DOI: 10.7150/jca.19057] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Hypoxia was a common feature for accelerating tumor metastasis by both inducting epithelial-mesenchymal transition (EMT) of tumor cells and polarization of tumor-associated macrophages (TAMs). The association and roles between hypoxia, EMT and TAMs in the biological behavior of gastric cancer (GC) for the time being recurrence is unclear. Material and methods: hypoixa by expression of hypoxia-inducible factor-1 alpha (HIF-1α), polarized functional status of infiltrated TAMs by immunohistochemical staining of CD68 and CD163, and the expression of E-cadherin as EMT property had been evaluated in 236 patients consecutive with histologically confirmed GC. Clinical significance was assessed for all these patients. Results: High expression of HIF-1α was found in patients with aggressive features, especially for recurrent patients. High infiltration of TAMs and abnormal expression of EMT-marker were also related to aggressive characteristics and predicted poor prognosis in GC. Meanwwhile, there existed a significant correlation among expression of HIF-1α, infiltration of TAMs and EMT marker in GC tissues. Multivariate Cox analysis revealed that high expression of HIF-1α combined TAMs infiltration were independent prognostic factors for disease-specific survival rate. Conclusion: HIF-1α is an unfavorable indicator for prognosis, may promote tumor progression through the induction of EMT and establishment of a pro-tumor immunosuppressive microenvironment. Further investigation into the therapeutic effects of blocking hypoxia is possible a potential strategy for GC treatment.
Collapse
Affiliation(s)
- Wei-Jie Zhang
- Department of General surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Department of General surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Cheng Chen
- Department of Radiotherapy, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhi-Hua Zhou
- Department of Pathology, 101th Hospital of PLA, Wuxi, Jiangsu Providence, China
| | - Shan-Ting Gao
- Department of General surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Teong Jin Tee
- Department of Medical, Gastroenterology unit, Nilai Medical Center, Nilai, Negeri Sembilan, Malaysia
| | - Liu-Qing Yang
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Yuan-Yuan Xu
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Tao-Hong Pang
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Xin-Yun Xu
- Department of Pathology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Qi Sun
- Department of Pathology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Min Feng
- Department of General surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Department of General surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Hao Wang
- Department of General surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Department of General surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Chun-Lei Lu
- Department of General surgery, 101th Hospital of PLA, Wuxi, Jiangsu Providence, China
| | - Guo-Zhong Wu
- Department of General surgery, 101th Hospital of PLA, Wuxi, Jiangsu Providence, China
| | - Sheng Wu
- Department of General surgery, 101th Hospital of PLA, Wuxi, Jiangsu Providence, China
| | - Wen-Xian Guan
- Department of General surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.,Department of General surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Gui-Fang Xu
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| |
Collapse
|
19
|
Quantitative imaging outperforms molecular markers when predicting response to chemoradiotherapy for rectal cancer. Radiother Oncol 2017. [PMID: 28647399 DOI: 10.1016/j.radonc.2017.06.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE To explore the integration of imaging and molecular data for response prediction to chemoradiotherapy (CRT) for rectal cancer. MATERIAL AND METHODS Eighty-five rectal cancer patients underwent preoperative CRT. 18F-FDG PET/CT and diffusion-weighted imaging (DWI) were acquired before (TP1) and during CRT (TP2) and prior to surgery (TP3). Inflammatory cytokines and gene expression were analysed. Tumour response was defined as ypT0-1N0. Multivariate models were built combining the obtained parameters. Final models were calculated on the data combination with the highest AUC. RESULTS Twenty-two patients (26%) achieved ypT0-1N0 response. 18F-FDG PET/CT had worse predictive performance than DWI and T2-volumetry (AUC 0.61±0.04, 0.72±0.03, and 0.72±0.02, respectively). Combining all imaging parameters increased the AUC to 0.81±0.03. Adding cytokines or gene expression did not improve the AUC (AUC of 0.72±0.06 and 0.79±0.04 respectively). Final models combining 18F-FDG PET/CT, DWI, and T2-weighted volumetry at all TPs and using only TP1 and TP3, allowed ypT0-1N0 prediction with a 75% sensitivity, 94% specificity and PPV of 80%. CONCLUSIONS Combining 18F-FDG PET/CT, DWI, and T2-weighted MRI volumetry obtained before CRT and prior to surgery may help physicians in selecting rectal cancer patients for organ-preservation.
Collapse
|
20
|
Sztupinszki Z, Győrffy B. Colon cancer subtypes: concordance, effect on survival and selection of the most representative preclinical models. Sci Rep 2016; 6:37169. [PMID: 27849044 PMCID: PMC5111107 DOI: 10.1038/srep37169] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022] Open
Abstract
Multiple gene-expression-based subtypes have been proposed for the molecular subdivision of colon cancer in the last decade. We aimed to cross-validate these classifiers to explore their concordance and their power to predict survival. A gene-chip-based database comprising 2,166 samples from 12 independent datasets was set up. A total of 22 different molecular subtypes were re-trained including the CCHS, CIN25, CMS, ColoGuideEx, ColoGuidePro, CRCassigner, MDA114, Meta163, ODXcolon, Oncodefender, TCA19, and V7RHS classifiers as well as subtypes established by Budinska, Chang, DeSousa, Marisa, Merlos, Popovici, Schetter, Yuen, and Watanabe (first authors). Correlation with survival was assessed by Cox proportional hazards regression for each classifier using relapse-free survival data. The highest efficacy at predicting survival in stage 2-3 patients was achieved by Yuen (p = 3.9e-05, HR = 2.9), Marisa (p = 2.6e-05, HR = 2.6) and Chang (p = 9e-09, HR = 2.35). Finally, 61 colon cancer cell lines from four independent studies were assigned to the closest molecular subtype.
Collapse
Affiliation(s)
- Zsófia Sztupinszki
- MTA TTK Lendület Cancer Biomarker Research Group, 1117, Budapest, Hungary
- 2 Dept. of Pediatrics, Semmelweis University, 1094, Budapest, Hungary
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, 1117, Budapest, Hungary
- 2 Dept. of Pediatrics, Semmelweis University, 1094, Budapest, Hungary
| |
Collapse
|
21
|
Warfel NA, Sainz AG, Song JH, Kraft AS. PIM Kinase Inhibitors Kill Hypoxic Tumor Cells by Reducing Nrf2 Signaling and Increasing Reactive Oxygen Species. Mol Cancer Ther 2016; 15:1637-47. [PMID: 27196781 DOI: 10.1158/1535-7163.mct-15-1018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/02/2016] [Indexed: 12/26/2022]
Abstract
Intratumoral hypoxia is a significant obstacle to the successful treatment of solid tumors, and it is highly correlated with metastasis, therapeutic resistance, and disease recurrence in cancer patients. As a result, there is an urgent need to develop effective therapies that target hypoxic cells within the tumor microenvironment. The Proviral Integration site for Moloney murine leukemia virus (PIM) kinases represent a prosurvival pathway that is upregulated in response to hypoxia, in a HIF-1-independent manner. We demonstrate that pharmacologic or genetic inhibition of PIM kinases is significantly more toxic toward cancer cells in hypoxia as compared with normoxia. Xenograft studies confirm that PIM kinase inhibitors impede tumor growth and selectively kill hypoxic tumor cells in vivo Experiments show that PIM kinases enhance the ability of tumor cells to adapt to hypoxia-induced oxidative stress by increasing the nuclear localization and activity of nuclear factor-erythroid 2 p45-related factor 2 (Nrf2), which functions to increase the expression of antioxidant genes. Small molecule PIM kinase inhibitors prevent Nrf2 from accumulating in the nucleus, reducing the transcription of cytoprotective genes and leading to the build-up of intracellular reactive oxygen species (ROS) to toxic levels in hypoxic tumor cells. This toxic effect of PIM inhibitors can be successfully blocked by ROS scavengers, including N-acetyl cystine and superoxide dismutase. Thus, inhibition of PIM kinases has the potential to oppose hypoxia-mediated therapeutic resistance and induce cell death in the hypoxic tumor microenvironment. Mol Cancer Ther; 15(7); 1637-47. ©2016 AACR.
Collapse
Affiliation(s)
- Noel A Warfel
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona. University of Arizona Cancer Center, Tucson, Arizona.
| | - Alva G Sainz
- UROC-PREP program, University of Arizona College of Undergraduate Studies, Tucson, Arizona
| | - Jin H Song
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona. University of Arizona Cancer Center, Tucson, Arizona
| | - Andrew S Kraft
- University of Arizona Cancer Center, Tucson, Arizona. Department of Medicine, University of Arizona, Tucson, Arizona.
| |
Collapse
|
22
|
Märkl B, Märkl M, Schaller T, Mayr P, Schenkirsch G, Kriening B, Anthuber M. A new simple morphology-based risk score is prognostic in stage I/II colon cancers. Cancer Med 2016; 5:1492-501. [PMID: 27167601 PMCID: PMC4867555 DOI: 10.1002/cam4.737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/21/2016] [Accepted: 03/23/2016] [Indexed: 12/27/2022] Open
Abstract
A portion of stage I/II colon cancers (10-20%) exhibit an adverse clinical course. The administration of adjuvant chemotherapy is recommended only in certain high-risk situations. However, these risk factors recently failed to predict benefit from adjuvant therapy. We composed a new morphology-based risk score that includes pT1/2 versus 3/4 stage, vascular or lymphovascular invasion, invasion type according to Jass, tumor budding and paucity (less than two) of lymph nodes larger than 5 mm. The occurrence of each of these factors accounts for one point in the score (Range 0-5). This score was evaluated in a retrospective study that included 301 cases. The overall survival differed significantly between the three groups with median survival times of 103, 90, and 48 months, respectively. Multivariable analysis revealed morphology-based risk-high risk and low risk-as the sole independent factors for the prediction of death. Morphology-based risk scoring was superior to microsatellite status and NCCN risk stratification. This method identifies a group of patients that comprises 18% of the stage II cases with an adverse clinical course. Further studies are necessary to confirm its prognostic value and the possible therapeutic consequences.
Collapse
Affiliation(s)
- Bruno Märkl
- Institute of Pathology, Klinikum Augsburg, Augsburg, Germany
| | | | - Tina Schaller
- Institute of Pathology, Klinikum Augsburg, Augsburg, Germany
| | - Patrick Mayr
- Institute of Pathology, Klinikum Augsburg, Augsburg, Germany
| | - Gerhard Schenkirsch
- Clinical and Population-Based Cancer Registry Augsburg, Klinikum Augsburg, Augsburg, Germany
| | - Bernadette Kriening
- Department of Visceral- and Transplantation Surgery, Klinikum Augsburg, Augsburg, Germany
| | - Matthias Anthuber
- Department of Visceral- and Transplantation Surgery, Klinikum Augsburg, Augsburg, Germany
| |
Collapse
|
23
|
Dekervel J, Popovic D, van Malenstein H, Windmolders P, Heylen L, Libbrecht L, Bulle A, De Moor B, Van Cutsem E, Nevens F, Verslype C, van Pelt J. A Global Risk Score (GRS) to Simultaneously Predict Early and Late Tumor Recurrence Risk after Resection of Hepatocellular Carcinoma. Transl Oncol 2016; 9:139-146. [PMID: 27084430 PMCID: PMC4833966 DOI: 10.1016/j.tranon.2016.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/18/2016] [Accepted: 02/24/2016] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES: Recurrence of hepatocellular carcinoma can arise from the primary tumor (“early recurrence”) or de novo from tumor formation in a cirrhotic environment (“late recurrence”). We aimed to develop one simple gene expression score applicable in both the tumor and the surrounding liver that can predict the recurrence risk. METHODS: We determined differentially expressed genes in a cell model of cancer aggressiveness. These genes were first validated in three large published data sets of hepatocellular carcinoma from which we developed a seven-gene risk score. RESULTS: The gene score was applied on two independent large patient cohorts. In the first cohort, with only tumor data available, it could predict the recurrence risk at 3 years after resection (68 ± 10% vs 35 ± 7%, P = .03). In the second cohort, when applied on the tumor, this gene score predicted early recurrence (62 ± 5% vs 37 ± 4%, P < .001), and when applied on the surrounding liver tissue, the same genes also correlated with late recurrence. Four patient classes with each different time patterns and rates of recurrence could be identified based on combining tumor and liver scores. In a multivariate Cox regression analysis, our gene score remained significantly associated with recurrence, independent from other important cofactors such as disease stage (P = .007). CONCLUSIONS: We developed a Global Risk Score that is able to simultaneously predict the risk of early recurrence when applied on the tumor itself, as well as the risk of late recurrence when applied on the surrounding liver tissue.
Collapse
Affiliation(s)
- Jeroen Dekervel
- Laboratory of Hepatology, Department of Clinical and Experimental Medicine, University Hospitals Leuven & KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Dusan Popovic
- Department of Electrical Engineering (ESAT), STADIUS Center for Dynamical Systems, Signal Processing and Data Analytics/iMinds Medical IT, KU Leuven, Kasteelpark Arenberg 10, 3000, Leuven, Belgium
| | - Hannah van Malenstein
- Laboratory of Hepatology, Department of Clinical and Experimental Medicine, University Hospitals Leuven & KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Petra Windmolders
- Laboratory of Hepatology, Department of Clinical and Experimental Medicine, University Hospitals Leuven & KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Line Heylen
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven & Department of Microbiology and Immunology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Louis Libbrecht
- Laboratory of Hepatology, Department of Clinical and Experimental Medicine, University Hospitals Leuven & KU Leuven, Herestraat 49, 3000, Leuven, Belgium; Department of Pathology, University Hospital Ghent, De Pintelaan 185, 9000, Ghent, Belgium
| | - Ashenafi Bulle
- Laboratory of Hepatology, Department of Clinical and Experimental Medicine, University Hospitals Leuven & KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Bart De Moor
- Department of Electrical Engineering (ESAT), STADIUS Center for Dynamical Systems, Signal Processing and Data Analytics/iMinds Medical IT, KU Leuven, Kasteelpark Arenberg 10, 3000, Leuven, Belgium
| | - Eric Van Cutsem
- Department of Clinical Digestive Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Frederik Nevens
- Laboratory of Hepatology, Department of Clinical and Experimental Medicine, University Hospitals Leuven & KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Chris Verslype
- Laboratory of Hepatology, Department of Clinical and Experimental Medicine, University Hospitals Leuven & KU Leuven, Herestraat 49, 3000, Leuven, Belgium; Department of Clinical Digestive Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Jos van Pelt
- Laboratory of Hepatology, Department of Clinical and Experimental Medicine, University Hospitals Leuven & KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
24
|
Li Z, Wang J, Zhou T, Ye X. Establishment of a colorectal cancer nude mouse visualization model of HIF-1α overexpression. Oncol Lett 2016; 11:2725-2732. [PMID: 27073543 PMCID: PMC4812287 DOI: 10.3892/ol.2016.4287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 11/26/2015] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to establish a model of tumor cell growth and visualize HIF-1α overexpression in a nude mouse xenograft model of colorectal cancer (CRC). In the study, HIF-1α lentiviral vector and helper plasmid were co-transfected into 293T packaging cells using a liposome method, and the virus was collected following transfection and used to infect CRC SW480, SW620, LoVo and HCT116 cells. Puromycin was used for the selection and large-scale amplification of the stable HIF-1α expression of green fluorescent protein (GFP)-positive cells. HIF-1α-expressing cells were injected intraperitoneally into a nude mouse xenograft model, and resulting tumor nodules was separated and confirmed using an inverted fluorescence microscope. The results demonstrated that HIF-1α was not expressed in CRC cells in normoxic conditions. When treated with CoCl2, the expression of HIF-1α could be induced in all the cancer cell lines, except SW480. HIF-1α was highly expressed following infection with lentiviral particles. Stable expression of HIF-1α promoted migration in the SW480 cells. Following intraperitoneal injection of nude mice with SW480-HIF-1α, a significant number of tumor nodules formed in the intestinal wall compared with the controls (P<0.05). The successful construction of the dual expression HIF-1α and GFP visualization xenograft model provides a good foundation for the screening of HIF-1α-related functions and for investigating the therapeutic potential of drugs that target HIF-1α.
Collapse
Affiliation(s)
- Zhe Li
- Department of Thyroid and Breast Surgery, The Shanghai Tenth People's Hospital, Tong Ji University, Shanghai 200072, P.R. China
| | - Jiye Wang
- Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou, Zhejiang 310053, P.R. China
| | - Tianbao Zhou
- Hepatobiliary Surgery Centre, The Ningbo No. 2 Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Xiaolei Ye
- Division of Drugs and Pharamcology, Ningbo Institute of Medical Sciences, Ningbo, Zhejiang 315020, P.R. China
| |
Collapse
|
25
|
|
26
|
Williams CS, Bernard JK, Demory Beckler M, Almohazey D, Washington MK, Smith JJ, Frey MR. ERBB4 is over-expressed in human colon cancer and enhances cellular transformation. Carcinogenesis 2015; 36:710-8. [PMID: 25916654 DOI: 10.1093/carcin/bgv049] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/05/2015] [Indexed: 01/21/2023] Open
Abstract
The ERBB4 receptor tyrosine kinase promotes colonocyte survival. Herein, we tested whether ERBB4's antiapoptotic signaling promotes transformation and colorectal tumorigenesis. ERBB4 alterations in a The Cancer Genome Atlas colorectal cancer (CRC) data set stratified survival, and in a combined Moffitt Cancer Center and Vanderbilt Medical Center CRC expression data set, ERBB4 message levels were increased at all tumor stages. Similarly, western blot and immunohistochemistry on additional CRC tissue banks showed elevated ERBB4 protein in tumors. ERBB4 was highly expressed in aggressive, dedifferentiated CRC cell lines, and its knockdown in LIM2405 cells reduced anchorage-independent colony formation. In nude mouse xenograft studies, ERBB4 alone was insufficient to induce tumor establishment of non-transformed mouse colonocytes, but its over-expression in cells harboring Apc(min) and v-Ha-Ras caused a doubling of tumor size. ERBB4-expressing xenografts displayed increased activation of survival pathways, including epidermal growth factor receptor and Akt phosphorylation and COX-2 expression, and decreased apoptotic signals. Finally, ERBB4 deletion from mouse intestinal epithelium impaired stem cell replication and in vitro enteroid establishment. In summary, we report that ERBB4 is over-expressed in human CRC, and in experimental systems enhances the survival and growth of cells driven by Ras and/or WNT signaling. Chronic ERBB4 over-expression in the context of, for example, inflammation may contribute to colorectal carcinogenesis. Tumors with high receptor levels are likely to have enhanced cell survival signaling through epidermal growth factor receptor, PI3K and COX-2. These results suggest ERBB4 as a novel therapeutic target in a subset of CRC.
Collapse
Affiliation(s)
- Christopher S Williams
- Departments of Medicine and Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA, Department of Surgery, Veterans Affairs Tennessee Valley Health Care System, Nashville, TN 37232, USA
| | - Jessica K Bernard
- Department of Pediatrics, University of Southern California Keck School of Medicine and The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Michelle Demory Beckler
- Department of Pediatrics, University of Southern California Keck School of Medicine and The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Dana Almohazey
- Department of Pediatrics, University of Southern California Keck School of Medicine and The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, CA 90027, USA, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Mary Kay Washington
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA and
| | - Jesse J Smith
- Department of Surgery, Veterans Affairs Tennessee Valley Health Care System, Nashville, TN 37232, USA
| | - Mark R Frey
- Department of Pediatrics, University of Southern California Keck School of Medicine and The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, CA 90027, USA, Department of Biochemistry and Molecular Biology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|
27
|
Endo H, Okuyama H, Ohue M, Inoue M. Dormancy of cancer cells with suppression of AKT activity contributes to survival in chronic hypoxia. PLoS One 2014; 9:e98858. [PMID: 24905002 PMCID: PMC4048292 DOI: 10.1371/journal.pone.0098858] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/08/2014] [Indexed: 01/23/2023] Open
Abstract
A hypoxic microenvironment in tumors has been recognized as a cause of malignancy or resistance to various cancer therapies. In contrast to recent progress in understanding the acute response of cancer cells to hypoxia, the characteristics of tumor cells in chronic hypoxia remain elusive. We have identified a pancreatic cancer cell line, AsPC-1, that is exceptionally able to survive for weeks under 1% oxygen conditions while most tested cancer cell lines die after only some days under these conditions. In chronic hypoxia, AsPC-1 cells entered a state of dormancy characterized by no proliferation, no death, and metabolic suppression. They reversibly switched to active status after being placed again in optimal culture conditions. ATP turnover, an indicator of energy demand, was markedly decreased and accompanied by reduced AKT phosphorylation. Forced activation of AKT resulted in increased ATP turnover and massive cell death in vitro and a decreased number of dormant cells in vivo. In contrast to most cancer cell lines, primary-cultured colorectal cancer cells easily entered the dormant status with AKT suppression under hypoxia combined with growth factor–depleted conditions. Primary colorectal cancer cells in dormancy were resistant to chemotherapy. Thus, the ability to survive in a deteriorated microenvironment by entering into dormancy under chronic hypoxia might be a common property among cancer cells. Targeting the regulatory mechanism inducing this dormant status could provide a new strategy for treating cancer.
Collapse
Affiliation(s)
- Hiroko Endo
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Hiroaki Okuyama
- Department of Pathology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Masayuki Ohue
- Department of Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Masahiro Inoue
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
- * E-mail:
| |
Collapse
|
28
|
Salazar R, Tabernero J. New Approaches but the Same Flaws in the Search for Prognostic Signatures. Clin Cancer Res 2014; 20:2019-22. [DOI: 10.1158/1078-0432.ccr-14-0219] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|