101
|
Kahle KT, Kozono D, Ng K, Hsieh G, Zinn PO, Nitta M, Chen CC. Functional genomics to explore cancer cell vulnerabilities. Neurosurg Focus 2010; 28:E5. [PMID: 20043720 DOI: 10.3171/2009.10.focus09212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our understanding of glioblastoma multiforme (GBM), the most common form of primary brain cancer, has been significantly advanced by recent efforts to characterize the cancer genome using unbiased high-throughput sequencing analyses. While these studies have documented hundreds of mutations, gene copy alterations, and chromosomal abnormalities, only a subset of these alterations are likely to impact tumor initiation or maintenance. Furthermore, genes that are not altered at the genomic level may play essential roles in tumor initiation and maintenance. Identification of these genes is critical for therapeutic development and investigative methodologies that afford insight into biological function. This requirement has largely been fulfilled with the emergence of RNA interference (RNAi) and high-throughput screening technology. In this article, the authors discuss the application of genome-wide, high-throughput RNAi-based genetic screening as a powerful tool for the rapid and cost-effective identification of genes essential for cancer proliferation and survival. They describe how these technologies have been used to identify genes that are themselves selectively lethal to cancer cells, or synthetically lethal with other oncogenic mutations. The article is intended to provide a platform for how RNAi libraries might contribute to uncovering glioma cell vulnerabilities and provide information that is highly complementary to the structural characterization of the glioblastoma genome. The authors emphasize that unbiased, systems-level structural and functional genetic approaches are complementary efforts that should facilitate the identification of genes involved in the pathogenesis of GBM and permit the identification of novel drug targets.
Collapse
Affiliation(s)
- Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
102
|
Scatena R, Bottoni P, Pontoglio A, Giardina B. Revisiting the Warburg effect in cancer cells with proteomics. The emergence of new approaches to diagnosis, prognosis and therapy. Proteomics Clin Appl 2010; 4:143-158. [DOI: 10.1002/prca.200900157] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
103
|
Li P, Lin JE, Marszlowicz GP, Valentino MA, Chang C, Schulz S, Pitari GM, Waldman SA. GCC signaling in colorectal cancer: Is colorectal cancer a paracrine deficiency syndrome? ACTA ACUST UNITED AC 2009; 22:313-8. [PMID: 19771320 DOI: 10.1358/dnp.2009.22.6.1395254] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Guanylyl cyclase C (GCC) is the receptor expressed by intestinal cells for the paracrine hormones guanylin and uroguanylin that coordinate mucosal homeostasis and its silencing contributes to intestinal transformation. It orchestrates proliferative and metabolic circuits by limiting the cell cycle and programming metabolic transitions central to regeneration along the crypt-villus axis. Mice deficient in GCC are more susceptible to colon cancer induced by germline mutations or carcinogens. Moreover, guanylin and uroguanylin are the most commonly lost gene products in colon cancer. The role of GCC as a tumor suppressor and the universal loss of its hormones in transformation suggest a paradigm in which colorectal cancer is a disease of paracrine hormone insufficiency. Indeed, GCC signaling reverses the tumorigenic phenotype of human colon cancer cells by regulating proliferation and metabolism. These data suggest a pathophysiological hypothesis in which GCC is a tumor suppressor coordinating proliferative homeostasis whose silencing through hormone loss initiates transformation. The correlative therapeutic hypothesis suggests that colorectal cancer is a disease of hormone insufficiency that can be prevented or treated by oral hormone replacement therapy employing GCC ligands.
Collapse
Affiliation(s)
- P Li
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | |
Collapse
|
104
|
Lartigue L, Kushnareva Y, Seong Y, Lin H, Faustin B, Newmeyer DD. Caspase-independent mitochondrial cell death results from loss of respiration, not cytotoxic protein release. Mol Biol Cell 2009; 20:4871-84. [PMID: 19793916 PMCID: PMC2785731 DOI: 10.1091/mbc.e09-07-0649] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 09/17/2009] [Accepted: 09/23/2009] [Indexed: 01/29/2023] Open
Abstract
In apoptosis, mitochondrial outer membrane permeabilization (MOMP) triggers caspase-dependent death. However, cells undergo clonogenic death even if caspases are blocked. One proposed mechanism involved the release of cytotoxic proteins (e.g., AIF and endoG) from mitochondria. To initiate MOMP directly without side effects, we created a tamoxifen-switchable BimS fusion protein. Surprisingly, even after MOMP, caspase-inhibited cells replicated DNA and divided for approximately 48 h before undergoing proliferation arrest. AIF and endoG remained in mitochondria. However, cells gradually lost mitochondrial membrane potential and ATP content, and DNA synthesis slowed to a halt by 72 h. These defects resulted from a partial loss of respiratory function, occurring 4-8 h after MOMP, that was not merely due to dispersion of cytochrome c. In particular, Complex I activity was completely lost, and Complex IV activity was reduced by approximately 70%, whereas Complex II was unaffected. Later, cells exhibited a more profound loss of mitochondrial protein constituents. Thus, under caspase inhibition, MOMP-induced clonogenic death results from a progressive loss of mitochondrial function, rather than the release of cytotoxic proteins from mitochondria.
Collapse
Affiliation(s)
- Lydia Lartigue
- *La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Yulia Kushnareva
- *La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Youngmo Seong
- *La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Helen Lin
- *La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | | | | |
Collapse
|
105
|
Alford R, Ogawa M, Choyke PL, Kobayashi H. Molecular probes for the in vivo imaging of cancer. MOLECULAR BIOSYSTEMS 2009; 5:1279-91. [PMID: 19823742 PMCID: PMC3407672 DOI: 10.1039/b911307j] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Advancements in medical imaging have brought about unprecedented changes in the in vivo assessment of cancer. Positron emission tomography, single photon emission computed tomography, optical imaging, and magnetic resonance imaging are the primary tools being developed for oncologic imaging. These techniques may still be in their infancy, as recently developed chemical molecular probes for each modality have improved in vivo characterization of physiologic and molecular characteristics. Herein, we discuss advances in these imaging techniques, and focus on the major design strategies with which molecular probes are being developed.
Collapse
Affiliation(s)
- Raphael Alford
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 1B40, MSC1088, Bethesda, Maryland, MD 20892-1088, USA; Fax: +1 301-402-3191; Tel: +1 301-451-4220
- Case Western Reserve School of Medicine, Cleveland, Ohio, USA
| | - Mikako Ogawa
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 1B40, MSC1088, Bethesda, Maryland, MD 20892-1088, USA; Fax: +1 301-402-3191; Tel: +1 301-451-4220
| | - Peter L. Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 1B40, MSC1088, Bethesda, Maryland, MD 20892-1088, USA; Fax: +1 301-402-3191; Tel: +1 301-451-4220
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 1B40, MSC1088, Bethesda, Maryland, MD 20892-1088, USA; Fax: +1 301-402-3191; Tel: +1 301-451-4220
| |
Collapse
|
106
|
Abstract
PURPOSE OF REVIEW To highlight key studies providing rationale for and utility in targeting glycolysis for the treatment of hematological malignancies. RECENT FINDINGS Several therapeutic strategies are capitalizing on the diagnostic utility of 18fluoro-deoxyglucose positron emission tomography that relies on increased glycolysis and glucose utilization in tumor cells. Although aerobic glycolysis was initially proposed by Warburg to be due to mitochondrial impairment, recent studies have shown a preferential switch to glycolysis in tumor cells with functional mitochondria. Increased glucose consumption can be advantageous for a tumor cell through stimulation of cellular biosynthetic, energetic, and pro-survival pathways. We now have a greater appreciation for the utilization of glucose in specific metabolic pathways that in some aspects can be complemented with other nutrients such as glutamine. Targeting glucose consumption for the treatment of hematological malignancies seems to be a promising field that will require characterization of tumor cell specific targets to inhibit glucose uptake and/or glycolysis. It is imperative to further our understanding of the tumor cell metabolome to target cellular bioenergetics in the treatment of cancer. SUMMARY Targeting the glycolytic pathway for the treatment of hematological malignancies has sufficient rationale given the utility of fluoro-deoxyglucose positron emission tomography in diagnostic imaging. Further research is required in developing tumor cell specific therapeutics.
Collapse
Affiliation(s)
- Mala Shanmugam
- Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA.
| | | | | |
Collapse
|
107
|
Govekar RB, D'Cruz AK, Alok Pathak K, Agarwal J, Dinshaw KA, Chinoy RF, Gadewal N, Kannan S, Sirdeshmukh R, Sundaram CS, Malgundkar SA, Kane SV, Zingde SM. Proteomic profiling of cancer of the gingivo-buccal complex: Identification of new differentially expressed markers. Proteomics Clin Appl 2009; 3:1451-62. [PMID: 21136964 DOI: 10.1002/prca.200900023] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 08/18/2009] [Accepted: 09/07/2009] [Indexed: 11/11/2022]
Abstract
Tobacco-related oral cancer is the most common cancer among Indian males, gingivo-buccal complex (GBC) being the most affected subsite due to the habit of chewing tobacco. Proteins from the lysates of microdissected normal and transformed epithelium from clinically well-characterized tissue samples of the GBC were separated by two-dimensional gel electrophoresis to identify differentially expressed proteins. Eleven protein spots showed differential expression, which could withstand the stringency of statistical evaluation. The observations were confirmed with additional tissues. Nine of these differentiators were identified by MS as lactate dehydrogenase B, α-enolase, prohibitin, cathepsin D, apolipoprotein A-I, tumor protein translationally controlled-1, an SFN family protein, 14-3-3σ and tropomyosin. Cluster analysis indicated that these proteins, as a coexpressed set, could distinguish normal and transformed epithelium. Functionally, these differentiator molecules are relevant to the pathways and processes that have been previously implicated in oral carcinogenesis and could therefore be investigated further as a panel of markers for management of cancer of the GBC.
Collapse
Affiliation(s)
- Rukmini B Govekar
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, India
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Electron microscopy morphology of the mitochondrial network in human cancer. Int J Biochem Cell Biol 2009; 41:2062-8. [DOI: 10.1016/j.biocel.2009.02.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 01/08/2009] [Accepted: 02/06/2009] [Indexed: 12/17/2022]
|
109
|
de Groof AJC, te Lindert MM, van Dommelen MMT, Wu M, Willemse M, Smift AL, Winer M, Oerlemans F, Pluk H, Fransen JAM, Wieringa B. Increased OXPHOS activity precedes rise in glycolytic rate in H-RasV12/E1A transformed fibroblasts that develop a Warburg phenotype. Mol Cancer 2009; 8:54. [PMID: 19646236 PMCID: PMC2734543 DOI: 10.1186/1476-4598-8-54] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 07/31/2009] [Indexed: 12/11/2022] Open
Abstract
Background The Warburg phenotype in cancer cells has been long recognized, but there is still limited insight in the consecutive metabolic alterations that characterize its establishment. We obtained better understanding of the coupling between metabolism and malignant transformation by studying mouse embryonic fibroblast-derived cells with loss-of-senescence or H-RasV12/E1A-transformed phenotypes at different stages of oncogenic progression. Results Spontaneous immortalization or induction of senescence-bypass had only marginal effects on metabolic profiles and viability. In contrast, H-RasV12/E1A transformation initially caused a steep increase in oxygen consumption and superoxide production, accompanied by massive cell death. During prolonged culture in vitro, cell growth rate increased gradually, along with tumor forming potential in in vitro anchorage-independent growth assays and in vivo tumor formation assays in immuno-deficient mice. Notably, glucose-to-lactic acid flux increased with passage number, while cellular oxygen consumption decreased. This conversion in metabolic properties was associated with a change in mitochondrial NAD+/NADH redox, indicative of decreased mitochondrial tricarboxic acid cycle and OXPHOS activity. Conclusion The high rate of oxidative metabolism in newly transformed cells is in marked contrast with the high glycolytic rate in cells in the later tumor stage. In our experimental system, with cells growing under ambient oxygen conditions in nutrient-rich media, the shift towards this Warburg phenotype occurred as a step-wise adaptation process associated with augmented tumorigenic capacity and improved survival characteristics of the transformed cells. We hypothesize that early-transformed cells, which potentially serve as founders for new tumor masses may escape therapies aimed at metabolic inhibition of tumors with a fully developed Warburg phenotype.
Collapse
Affiliation(s)
- Ad J C de Groof
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Abstract
Up to 50% of cancer patients suffer from a progressive atrophy of adipose tissue and skeletal muscle, called cachexia, resulting in weight loss, a reduced quality of life, and a shortened survival time. Anorexia often accompanies cachexia, but appears not to be responsible for the tissue loss, particularly lean body mass. An increased resting energy expenditure is seen, possibly arising from an increased thermogenesis in skeletal muscle due to an increased expression of uncoupling protein, and increased operation of the Cori cycle. Loss of adipose tissue is due to an increased lipolysis by tumor or host products. Loss of skeletal muscle in cachexia results from a depression in protein synthesis combined with an increase in protein degradation. The increase in protein degradation may include both increased activity of the ubiquitin-proteasome pathway and lysosomes. The decrease in protein synthesis is due to a reduced level of the initiation factor 4F, decreased elongation, and decreased binding of methionyl-tRNA to the 40S ribosomal subunit through increased phosphorylation of eIF2 on the alpha-subunit by activation of the dsRNA-dependent protein kinase, which also increases expression of the ubiquitin-proteasome pathway through activation of NFkappaB. Tumor factors such as proteolysis-inducing factor and host factors such as tumor necrosis factor-alpha, angiotensin II, and glucocorticoids can all induce muscle atrophy. Knowledge of the mechanisms of tissue destruction in cachexia should improve methods of treatment.
Collapse
Affiliation(s)
- Michael J Tisdale
- Nutritional Biomedicine, School of Life and Health Sciences, Aston University, Birmingham, UK.
| |
Collapse
|
111
|
Vizán P, Alcarraz-Vizán G, Díaz-Moralli S, Solovjeva ON, Frederiks WM, Cascante M. Modulation of pentose phosphate pathway during cell cycle progression in human colon adenocarcinoma cell line HT29. Int J Cancer 2009; 124:2789-96. [PMID: 19253370 DOI: 10.1002/ijc.24262] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cell cycle regulation is dependent on multiple cellular and molecular events. Cell proliferation requires metabolic sources for the duplication of DNA and cell size. However, nucleotide reservoirs are not sufficient to support cell duplication and, therefore, biosynthetic pathways should be upregulated during cell cycle. Here, we reveal that glucose-6-phosphate dehydrogenase (G6PDH) and transketolase (TKT), the 2 key enzymes of oxidative and nonoxidative branches of the pentose phosphate pathway (PPP), respectively, which is necessary for nucleotide synthesis, are enhanced during cell cycle progression of the human colon cancer cell line HT29. These enhanced enzyme activities coincide with an increased ratio of pentose monophosphate to hexose monophosphate pool during late G1 and S phase, suggesting a potential role for pentose phosphates in proliferating signaling. Isotopomeric analysis distribution of nucleotide ribose synthesized from 1,2-(13)C(2)-glucose confirms the activation of the PPP during late G1 and S phase and reveals specific upregulation of the oxidative branch. Our data sustain the idea of a critical oxidative and nonoxidative balance in cancer cells, which is consistent with a late G1 metabolic check point. The distinctive modulation of these enzymes during cell cycle progression may represent a new strategy to inhibit proliferation in anticancer treatments.
Collapse
Affiliation(s)
- Pedro Vizán
- Department of Biochemistry and Molecular Biology, Institute of Biomedicine of University of Barcelona (IBUB), Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
112
|
Lin JE, Li P, Pitari GM, Schulz S, Waldman SA. Guanylyl cyclase C in colorectal cancer: susceptibility gene and potential therapeutic target. Future Oncol 2009; 5:509-22. [PMID: 19450179 PMCID: PMC2749695 DOI: 10.2217/fon.09.14] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer is one of the leading causes of tumor-related morbidity and mortality worldwide. While mechanisms underlying this disease have been elucidated over the past two decades, these molecular insights have failed to translate into efficacious therapy. The oncogenomic view of cancer suggests that terminal transformation reflects the sequential corruption of signal transduction circuits regulating key homeostatic mechanisms, whose multiplicity underlies the therapeutic resistance of most tumors to interventions targeting individual pathways. Conversely, the paucity of mechanistic insights into proximal pathophysiological processes that initiate and amplify oncogenic circuits preceding accumulation of mutations and transformation impedes development of effective prevention and therapy. In that context, guanylyl cyclase C (GCC), the intestinal receptor for the paracrine hormones guanylin and uroguanylin, whose early loss characterizes colorectal transformation, has emerged as a component of lineage-specific homeostatic programs organizing spatiotemporal patterning along the crypt-surface axis. Dysregulation of GCC signaling, reflecting hormone loss, promotes tumorigenesis through reprogramming of replicative and bioenergetic circuits and genomic instability. Compensatory upregulation of GCC in response to hormone loss provides a unique translational opportunity for prevention and treatment of colorectal tumors by hormone-replacement therapy.
Collapse
Affiliation(s)
- Jieru E Lin
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 132 South 10th Street, 1170 Main, Philadelphia, PA 19107, USA.
| | | | | | | | | |
Collapse
|
113
|
Yoshii Y, Furukawa T, Yoshii H, Mori T, Kiyono Y, Waki A, Kobayashi M, Tsujikawa T, Kudo T, Okazawa H, Yonekura Y, Fujibayashi Y. Cytosolic acetyl-CoA synthetase affected tumor cell survival under hypoxia: the possible function in tumor acetyl-CoA/acetate metabolism. Cancer Sci 2009; 100:821-7. [PMID: 19445015 PMCID: PMC11158093 DOI: 10.1111/j.1349-7006.2009.01099.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Understanding tumor-specific metabolism under hypoxia is important to find novel targets for antitumor drug design. Here we found that tumor cells expressed higher levels of cytosolic acetyl-CoA synthetase (ACSS2) under hypoxia than normoxia. Knockdown of ACSS2 by RNA interference (RNAi) in tumor cells enhanced tumor cell death under long-term hypoxia in vitro. Our data also demonstrated that the ACSS2 suppression slowed tumor growth in vivo. These findings showed that ACSS2 plays a significant role in tumor cell survival under hypoxia and that ACSS2 would be a potential target for tumor treatment. Furthermore, we found that tumor cells excreted acetate and the quantity increased under hypoxia: the pattern of acetate excretion followed the expression pattern of ACSS2. Additionally, the ACSS2 knockdown led to a corresponding reduction in the acetate excretion in tumor cells. These results mean that ACSS2 can conduct the reverse reaction from acetyl-CoA to acetate in tumor cells, which indicates that ACSS2 is a bi-directional enzyme in tumor cells and that ACSS2 might play a buffering role in tumor acetyl-CoA/acetate metabolism.
Collapse
Affiliation(s)
- Yukie Yoshii
- Biomedical Imaging Research Center, University of Fukui, 23-3, Shimoaizuki, Eiheiji, Fukui 910-1193, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
|
115
|
Lee SC, Delikatny EJ, Poptani H, Pickup S, Glickson JD. In vivo (1)H MRS of WSU-DLCL2 human non-Hodgkin's lymphoma xenografts: response to rituximab and rituximab plus CHOP. NMR IN BIOMEDICINE 2009; 22:259-265. [PMID: 19040203 DOI: 10.1002/nbm.1316] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
In order to identify early (1)H MRS metabolic markers of response to rituximab immunotherapy and to rituximab plus CHOP (cyclophosphamide, hydroxydoxorubicin, vincristine, and prednisone) combination therapy, we performed an in vivo MRS investigation of a non-Hodgkin's lymphoma (NHL) xenograft model. Human WSU-DLCL2 NHL cells were subcutaneously implanted into flanks of female severe combined immunodeficient mice. When tumor volumes reached approximately 600 mm(3), rituximab was administered for three weekly cycles at a dose of 25 mg/kg per cycle with or without CHOP. Before and after treatment, tumor lactate (Lac) and total choline (tCho) were detected using the selective multiple quantum coherence sequence and the stimulated echo acquisition mode sequence, respectively. Rituximab produced a small tumor growth delay ( approximately 5 days), whereas treatment with rituximab plus CHOP (RCHOP) led to approximately 20% tumor regression after three cycles of therapy. After one cycle of rituximab, the tCho/H(2)O ratio had decreased significantly (5%, P = 0.003), whereas the Lac/H(2)O ratio had not changed (P = 0.58). Both Lac/H(2)O and tCho/H(2)O had decreased after one cycle of RCHOP treatment (26%, P = 0.001; 10%, P = 0.016, respectively). After two cycles of RCHOP, Ki67 assay of histological tumor specimens indicated approximately 40% decrease in proliferation (P < 0.001) in the RCHOP-treated tumors; no change was detected after treatment with rituximab alone. This study suggests that decreases in tCho/H(2)O are more sensitive indices of response to rituximab, whereas decreases in Lac/H(2)O are more sensitive to response to CHOP combination therapy.
Collapse
Affiliation(s)
- Seung-Cheol Lee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
116
|
Mendelsohn BA, Malone JP, Townsend RR, Gitlin JD. Proteomic analysis of anoxia tolerance in the developing zebrafish embryo. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2009; 4:21-31. [PMID: 20403745 PMCID: PMC2858231 DOI: 10.1016/j.cbd.2008.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/13/2008] [Revised: 09/25/2008] [Accepted: 09/26/2008] [Indexed: 12/26/2022]
Abstract
While some species and tissue types are injured by oxygen deprivation, anoxia tolerant organisms display a protective response that has not been fully elucidated and is well-suited to genomic and proteomic analysis. However, such methodologies have focused on transcriptional responses, prolonged anoxia, or have used cultured cells or isolated tissues. In this study of intact zebrafish embryos, a species capable of >24 h survival in anoxia, we have utilized 2D difference in gel electrophoresis to identify changes in the proteomic profile caused by near-lethal anoxic durations as well as acute anoxia (1 h), a timeframe relevant to ischemic events in human disease when response mechanisms are largely limited to post-transcriptional and post-translational processes. We observed a general stabilization of the proteome in anoxia. Proteins involved in oxidative phosphorylation, antioxidant defense, transcription, and translation changed over this time period. Among the largest proteomic alterations was that of muscle cofilin 2, implicating the regulation of the cytoskeleton and actin assembly in the adaptation to acute anoxia. These studies in an intact embryo highlight proteomic components of an adaptive response to anoxia in a model organism amenable to genetic analysis to permit further mechanistic insight into the phenomenon of anoxia tolerance.
Collapse
Affiliation(s)
- Bryce A. Mendelsohn
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - James P. Malone
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - R. Reid Townsend
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Jonathan D. Gitlin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
117
|
Abstract
p53 has been referred to as the 'guardian of the genome' because of its role in protecting the cell from DNA damage. p53 performs its duties by regulating cell-cycle progression and DNA repair and, in cases of irreparable DNA damage, by executing programmed cell death. Mitochondria are an important target of transcription-dependent and -independent actions of p53 to carry out the apoptotic function. However, increasing evidence suggests that p53 activity is regulated by mitochondria. Cellular insults that alter mitochondrial function can have important consequences on p53 activity. In light of these new findings, the following review focuses on p53/mitochondria connections, in particular how reactive oxygen species generated at mitochondria regulate p53 activity. A better understanding of the mechanisms by which mitochondria regulate p53 may have an impact on our understanding of the development and progression of many diseases, especially cancer.
Collapse
Affiliation(s)
- Aaron K Holley
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K St Clair
- Graduate Center for Toxicology, 454 HSRB, University of Kentucky, Lexington, KY 40536, USA, Tel.: +1 859 257 3956, Fax: +1 859 323 1059,
| |
Collapse
|
118
|
Mendelsohn BA, Gitlin JD. Coordination of development and metabolism in the pre-midblastula transition zebrafish embryo. Dev Dyn 2008; 237:1789-98. [PMID: 18521947 DOI: 10.1002/dvdy.21584] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
To define the mechanisms that coordinate early embryonic development and metabolism, we have examined the response of zebrafish embryos to anoxia before the midblastula transition. Our findings reveal that anoxic pre-midblastula transition embryos slow the cell cycle, arrest before the midblastula transition and can recover normally if restored to a normoxic environment. Analyses of respiratory rates reveal that pre-midblastula transition embryos are less reliant on oxidative phosphorylation than older embryos. Interestingly, arrest in anoxia occurs despite inhibition of zygotic transcription, revealing a central role for maternal factors in the response to energy limitation. Consistent with this concept, we demonstrate that the posttranslational energy-sensing AMP-activated protein kinase pathway is activated in anoxia in pre-midblastula transition embryos. Taken together, these findings demonstrate a maternal program capable of coordinating developmental rate and metabolism in the absence of transcription-based pathways or cell cycle checkpoints.
Collapse
Affiliation(s)
- Bryce A Mendelsohn
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
119
|
Abstract
Transcriptional control of cellular energy metabolic pathways is achieved by the coordinated action of numerous transcription factors and associated coregulators. Several members of the nuclear receptor superfamily have been shown to play important roles in this process because they can translate hormonal, nutrient, and metabolite signals into specific gene expression networks to satisfy energy demands in response to distinct physiological cues. Estrogen-related receptor (ERR) alpha, ERRbeta, and ERRgamma are nuclear receptors that have yet to be associated with a natural ligand and are thus considered as orphan receptors. However, the transcriptional activity of the ERRs is exquisitely sensitive to the presence of coregulatory proteins known to be essential for the control of energy homeostasis, and for all intents and purposes, these coregulators function as protein ligands for the ERRs. In particular, functional genomics and biochemical studies have shown that ERRalpha and ERRgamma operate as the primary conduits for the activity of members of the family of PGC-1 coactivators. As transcription factors, the ERRs control vast gene networks involved in all aspects of energy homeostasis, including fat and glucose metabolism as well as mitochondrial biogenesis and function. Phenotypic analyses of knockout mouse models have shown that all three ERRs are indispensable for proper development and/or survival of the organism when subjected to a variety of physiological challenges. The focus of this review is on the recent and rapid advances in understanding the functions of the ERRs in regulating bioenergetic pathways, with an emphasis on their roles in the specification of energetic properties required for cell- and tissue-specific functions.
Collapse
Affiliation(s)
- Vincent Giguère
- The Rosalind and Morris Goodman Cancer Centre, Cancer Pavilion, 1160 Pine Avenue West, Montreal, Quebec, Canada H3A 1A3.
| |
Collapse
|
120
|
Abstract
Molecular imaging of tumor metabolism has gained considerable interest, since preclinical studies have indicated a close relationship between the activation of various oncogenes and alterations of cellular metabolism. Furthermore, several clinical trials have shown that metabolic imaging can significantly impact patient management by improving tumor staging, restaging, radiation treatment planning, and monitoring of tumor response to therapy. In this review, we summarize recent data on the molecular mechanisms underlying the increased metabolic activity of cancer cells and discuss imaging techniques for studies of tumor glucose, lipid, and amino acid metabolism.
Collapse
Affiliation(s)
- Christian Plathow
- Department of Nuclear Medicine, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
121
|
Lee SC, Huang MQ, Nelson DS, Pickup S, Wehrli S, Adegbola O, Poptani H, Delikatny EJ, Glickson JD. In vivo MRS markers of response to CHOP chemotherapy in the WSU-DLCL2 human diffuse large B-cell lymphoma xenograft. NMR IN BIOMEDICINE 2008; 21:723-733. [PMID: 18384181 DOI: 10.1002/nbm.1250] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
To identify 1H-MRS molecular biomarkers of early clinical therapeutic response in non-Hodgkin's lymphoma, an in vivo longitudinal study was performed on human non-Hodgkin's diffuse large B-cell lymphoma xenografts (WSU-DLCL2) grown in the flanks of female SCID mice. 31P-MRS measurements, which have been demonstrated to be prognostic clinical indices of response (Arias-Mendoza et al. Acad. Radiol. 2004; 11: 368-376) but which provide lower spatial resolution, were included for comparison. The animals received CHOP (cyclophosphamide, hydroxydoxorubicin, oncovin and prednisone) chemotherapy for three 1-week cycles, resulting in stable disease based on tumor volume. Localization of total choline and phosphorus metabolites in vivo was achieved with stimulated echo acquisition mode and image selected in vivo spectroscopy sequences, respectively. Significant decreases in lactate were detected by the selective multiple quantum coherence spectral editing technique after the first cycle of CHOP, whereas total choline and the phosphomonoester/nucleoside triphosphate ratio did not change until the third cycle. Ex vivo extract MRS of tumors corroborated the in vivo results. Histological staining with antibodies to Ki67 revealed a decrease in proliferation rate in CHOP-treated tumors that coincided with the decrease in lactate. This study demonstrates the utility of lactate as an early proliferation-sensitive indicator of therapeutic response in a mouse model of non-Hodgkin's lymphoma and serves as a basis for future clinical implementation of these methods.
Collapse
Affiliation(s)
- Seung-Cheol Lee
- Molecular Imaging Laboratory, Department of Radiology, University of Pennsylvania, and NMR Core Facility, Children's Hospital of Philadelphia, Philadelphia, PA 19104-4011, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Pickup S, Lee SC, Mancuso A, Glickson JD. Lactate imaging with Hadamard-encoded slice-selective multiple quantum coherence chemical-shift imaging. Magn Reson Med 2008; 60:299-305. [PMID: 18666110 PMCID: PMC2701382 DOI: 10.1002/mrm.21659] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Accepted: 03/17/2008] [Indexed: 11/06/2022]
Abstract
The ability to generate in vivo maps of lactate may have significant diagnostic utility in staging and treatment planning of a wide variety of cancers. The double selective multiple quantum filter technique (SelMQC) has been shown to be effective for nonlocalized detection of lactate with little or no interference from other signals. Here the SelMQC technique has been combined with longitudinal Hadamard slice selection and chemical shift imaging (CSI) to yield slice-selective images of lactate. The technique is shown to be effective in phantoms and in WSU-DLCL2 xenografts implanted in flanks of SCID mice. Tumors exhibited an annulus of elevated lactate concentration surrounding a necrotic tumor core.
Collapse
Affiliation(s)
- Stephen Pickup
- Department of Radiology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
123
|
Jiang X, Kenerson H, Aicher L, Miyaoka R, Eary J, Bissler J, Yeung RS. The tuberous sclerosis complex regulates trafficking of glucose transporters and glucose uptake. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1748-56. [PMID: 18511518 DOI: 10.2353/ajpath.2008.070958] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human cancers often display an avidity for glucose, a feature that is exploited in clinical staging and response monitoring by using (18)F-fluoro-deoxyglucose (FDG) positron emission tomography. Determinants of FDG accumulation include tumor blood flow, glucose transport, and glycolytic rate, but the underlying molecular mechanisms are incompletely understood. The phosphoinositide-3 kinase/Akt/mammalian target of rapamycin complex (mTORC) 1 pathway has been implicated in this process via the hypoxia-inducible factor alpha-dependent expression of vascular endothelial growth factor and glycolytic enzymes. Thus, we predicted that tumors with elevated mTORC1 activity would be accompanied by high FDG uptake. We tested this hypothesis in eight renal angiomyolipomas in which the loss of tuberous sclerosis complex (TSC) 1/2 function gave rise to constitutive mTORC1 activation. Surprisingly, these tumors displayed low FDG uptake on positron emission tomography. Exploring the underlying mechanisms in vitro revealed that Tsc2 regulates the membrane localization of the glucose transporter proteins (Glut)1, Glut2, and Glut4, and, therefore, glucose uptake. Down-regulation of cytoplasmic linker protein 170, an mTOR effector, rescued Glut4 trafficking in Tsc2(-/-) cells, whereas up-regulation of Akt activity in these cells was insufficient to redistribute Glut4 to the plasma membrane. The effect of mTORC1 on glucose uptake was confirmed using a liver-specific Tsc1- deletion mouse model in which FDG uptake was reduced in the livers of mutant mice compared with wild-type controls. Together, these data show that mTORC1 activity is insufficient for increased glycolysis in tumors and that constitutive mTOR activity negatively regulates glucose transporter trafficking.
Collapse
Affiliation(s)
- Xiuyun Jiang
- Department of Surgery, Box 356410, University of Washington, 1959 NE Pacific, Seattle, WA 98195, USA
| | | | | | | | | | | | | |
Collapse
|
124
|
Abstract
Developing organisms depend upon a delicate balance in the supply and demand of energy to adapt to variable oxygen availability, although the essential mechanisms determining such adaptation remain elusive. In this study, we examine reversible anoxic arrest and dynamic bioenergetic transitions during zebrafish development. Our data reveal that the duration of anoxic viability corresponds to the developmental stage and anaerobic metabolic rate. Diverse chemical inhibitors of mitochondrial oxidative phosphorylation induce a similar arrest in normoxic embryos, suggesting a pathway responsive to perturbations in aerobic energy production rather than molecular oxygen. Consistent with this concept, arrest is accompanied by rapid activation of the energy-sensing AMP-activated protein kinase pathway, demonstrating a potential link between the sensing of energy status and adaptation to oxygen availability. These observations permit mechanistic insight into energy homeostasis during development that now enable genetic and small molecule screens in this vertebrate model of anoxia tolerance.
Collapse
Affiliation(s)
- Bryce A. Mendelsohn
- Edward Mallinckrodt Department of Pediatrics Washington University School of Medicine St. Louis, Missouri 63110, USA
| | - Bethany L. Kassebaum
- Edward Mallinckrodt Department of Pediatrics Washington University School of Medicine St. Louis, Missouri 63110, USA
| | - Jonathan D. Gitlin
- Edward Mallinckrodt Department of Pediatrics Washington University School of Medicine St. Louis, Missouri 63110, USA
| |
Collapse
|
125
|
Schöder H, Ong SC. Fundamentals of molecular imaging: rationale and applications with relevance for radiation oncology. Semin Nucl Med 2008; 38:119-28. [PMID: 18243847 DOI: 10.1053/j.semnuclmed.2007.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Molecular imaging allows for the visualization and quantification biologic processes at cellular levels. This article focuses on positron emission tomography as one readily available tool for clinical molecular imaging. To prove its clinical utility in oncology, molecular imaging will ultimately have to provide valuable information in the following 4 pertinent areas: staging; assessment of extent of disease; target delineation for radiation therapy planning; response prediction and assessment and differentiation between treatment sequelae and recurrent disease. These issues are addressed in other contributions in this issue of Seminars in Nuclear Medicine. In contrast, this article will focus on the biochemical principles of cancer metabolism that provide the rationale for positron emission tomography imaging in radiation oncology.
Collapse
Affiliation(s)
- Heiko Schöder
- Department of Radiology/Nuclear Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | |
Collapse
|
126
|
Ertuk M, Van den Abbeele AD. Infrequent Tumors of the Gastrointestinal Tract Including Gastrointestinal Stromal Tumor (GIST). PET Clin 2008; 3:207-15. [DOI: 10.1016/j.cpet.2008.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
127
|
Vazquez-Martin A, Colomer R, Brunet J, Lupu R, Menendez JA. Overexpression of fatty acid synthase gene activates HER1/HER2 tyrosine kinase receptors in human breast epithelial cells. Cell Prolif 2008; 41:59-85. [PMID: 18211286 DOI: 10.1111/j.1365-2184.2007.00498.x] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES More than 50 years ago, we learned that breast cancer cells (and those of many other types of tumour) endogenously synthesize 95% of fatty acids (FAs) de novo, despite having adequate nutritional lipid supply. Today, we know that breast cancer cells benefit from this phenomenon in terms of enhanced cell proliferation, survival, chemoresistance and metastasis. However, the exact role of the major lipogenic enzyme fatty acid synthase (FASN) as cause, correlate or facilitator of breast cancer remains unidentified. MATERIALS AND METHODS To evaluate a causal effect of FASN-catalysed endogenous FA biosynthesis in the natural history of breast cancer disease, HBL100 cells (an SV40-transformed in vitro model for near-normal gene expression in the breast epithelium), and MCF10A cells (a non-transformed, near diploid, spontaneously immortalized human mammary epithelial cell line) were acutely forced to overexpress the human FASN gene. RESULTS Following transient transfection with plasmid pCMV6-XL4 carrying full-length human FASN cDNA (gi: NM 004104), HBL100 cells enhanced their endogenous lipid synthesis while acquiring canonical oncogenic properties such as increased size and number of colonies in semisolid (i.e. soft-agar) anchorage-independent cultures. Anchorage-dependent cell proliferation assays in low serum (0.1% foetal bovine serum), MTT-based assessment of cell metabolic status and cell death ELISA-based detection of apoptosis-induced DNA-histone fragmentation, together revealed that sole activation of endogenous FA biosynthesis was sufficient to significantly enhance breast epithelial cell proliferation and survival. When analysing molecular mechanisms by which acute activation of de novo FA biosynthesis triggered a transformed phenotype, HBL100 cells, transiently transfected with pCMV6-XL4/FASN, were found to exhibit a dramatic increase in the number of phosphor-tyrosine (Tyr)-containing proteins, as detected by 4G10 antiphosphor-Tyr monoclonal antibody. Phosphor-Tyr-specific antibodies recognizing the phosphorylation status of either the 1173 Tyr residue of epidermal growth factor receptor (HER1) or the 1248 Tyr residue of HER2, further revealed that FASN-induced Tyr-phosphorylation at approximately 180 kDa region mainly represented that of these key members of the HER (erbB) network, which remained switched-off in mock-transfected HBL100 cells. ELISA and immunoblotting procedures demonstrated that FASN overactivation significantly increased (> 200%) expression levels of epidermal growth factor receptor and HER2 proteins in HBL100 cells. Proteome Profilertrade mark antibody arrays capable of simultaneously detecting relative levels of phosphorylation of 42 phospho-receptor Tyr-kinases (RTKs) confirmed that acute activation of endogenous FA biosynthesis specifically promoted hyper-Tyr-phosphorylation of HER1 and HER2 in MCF10A cells. This FASN-triggered HER1/HER2-breast cancer-like phenotype was specifically inhibitable either by FASN inhibitor C75 or by Tyr-kinase inhibitors (TKIs) gefitinib (Iressa) and lapatinib (Tykerb) but not by chemotherapeutic agents such as cisplatin. Transient overexpression of FASN dramatically increased HBL100 breast epithelial cells' sensitivity to cytotoxic effects of C75, gefitinib and lapatinib (approximately 8, 10 and > 15 times, respectively), while significantly decreasing (approximately 3 times) cisplatin efficacy. CONCLUSIONS Although we cannot definitely establish FASN as a novel oncogene in breast cancer, this study reveals for the first time that exacerbated endogenous FA biosynthesis in non-cancerous epithelial cells is sufficient to induce a cancer-like phenotype functionally dependent on the HER1/HER2 duo. These findings may perhaps radically amend our current perspective of endogenously synthesized fat, as on its own, it appears to actively increase signal-to-noise ratio in the HER1/HER2-driven progression of human breast epithelial cells towards malignancy.
Collapse
Affiliation(s)
- A Vazquez-Martin
- Catalan Institute of Oncology, Health Services Division of Catalonia, Catalonia, Spain
| | | | | | | | | |
Collapse
|
128
|
Abstract
As described by Warburg more than 50 years ago, tumour cells maintain a high glycolytic rate even in conditions of adequate oxygen supply. However, most of tumours are subjected to hypoxic conditions due to the abnormal vasculature that supply them with oxygen and nutrients. Thus, glycolysis is essential for tumour survival and spread. A key step in controlling glycolytic rate is the conversion of fructose-6-P to fructose-1,6-P(2) by 6-phosphofructo-1-kinase (PFK-1). The activity of PFK-1 is allosterically controlled by fructose-2,6-P(2), the product of the enzymatic activity of a dual kinase/phosphatase family of enzymes (PFKFB1-4) that are increased in a significant number of tumour types. In turn, these enzymes are induced by hypoxia through the activation of the HIF-1 complex (hypoxia-inducible complex-1), a transcriptional activator that controls the expression of most of hypoxia-regulated genes. HIF-1 complex is overexpressed in a variety of tumours and its expression appears to correlate with poor prognosis and responses to chemo or radiotherapy. Thus, targeting PFKFB enzymes, either directly or through inhibition of HIF-1, appears as a promising approach for the treatment of certain tumours.
Collapse
Affiliation(s)
- Ramon Bartrons
- Unitat Bioquímica i Biologia Molecular, Departament de Ciències Fisiològiques, Campus de Ciències de la Salut, IDIBELL--Universitat de Barcelona, Barcelona, Spain.
| | | |
Collapse
|
129
|
Kueck A, Opipari AW, Griffith KA, Tan L, Choi M, Huang J, Wahl H, Liu JR. Resveratrol inhibits glucose metabolism in human ovarian cancer cells. Gynecol Oncol 2007; 107:450-7. [PMID: 17825886 DOI: 10.1016/j.ygyno.2007.07.065] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Revised: 07/10/2007] [Accepted: 07/18/2007] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Resveratrol is a phytoalexin found in grapes that inhibits the in vitro growth of multiple tumor cell types. We showed previously that resveratrol induces autophagic cell death in ovarian cancer cells. Because autophagy is typically an adaptive response to nutrient starvation, we hypothesized that autophagy would also be triggered when ovarian cancer cells are nutrient deprived and that resveratrol could in fact be acting by inducing a starvation-like signaling response. METHODS Ovarian cancer cells were incubated with normal media, media containing resveratrol, glucose free media, or media lacking amino acids. Growth inhibition was determined using the sulforhodamine assay. Cells were evaluated for autophagocytosis by analyzing cleavage of LC3. Glucose uptake, lactate production, and activation of glycolytic regulators pAkt and pmTOR were analyzed following resveratrol treatment. RESULTS We show here that epithelial ovarian cancer cells are highly sensitive to glucose-deprivation-induced cell death and like resveratrol, glucose deprivation induces caspase-independent cell death with hallmarks of autophagy. Consistent with the hypothesis that resveratrol treatment results in biochemical conditions that mirror a nutrient deprived state, we found that resveratrol dramatically reduces glucose uptake and lactate production. Moreover, resveratrol reduces the levels of phosphorylated Akt and mTOR, two signals that increase glucose uptake and the rate limiting steps in glycolysis. CONCLUSIONS Our findings are consistent with the hypothesis that resveratrol-induced changes in glucose utilization comprise the mechanism that underlies resveratrol-induced autophagocytosis in ovarian cancer. Inhibition of glycolysis in ovarian cancer with resveratrol or other compounds may be effective therapy for ovarian cancer.
Collapse
Affiliation(s)
- Angela Kueck
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Michigan, L4000 Women's Hospital, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | |
Collapse
|
130
|
Regulation of mitochondrial oxidative phosphorylation through cell signaling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1701-20. [DOI: 10.1016/j.bbamcr.2007.10.001] [Citation(s) in RCA: 199] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
131
|
Kim JW, Gao P, Liu YC, Semenza GL, Dang CV. Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1. Mol Cell Biol 2007; 27:7381-93. [PMID: 17785433 PMCID: PMC2169056 DOI: 10.1128/mcb.00440-07] [Citation(s) in RCA: 501] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Revised: 04/24/2007] [Accepted: 08/24/2007] [Indexed: 02/07/2023] Open
Abstract
Hypoxia is a pervasive microenvironmental factor that affects normal development as well as tumor progression. In most normal cells, hypoxia stabilizes hypoxia-inducible transcription factors (HIFs), particularly HIF-1, which activates genes involved in anaerobic metabolism and angiogenesis. As hypoxia signals a cellular deprivation state, HIF-1 has also been reported to counter the activity of MYC, which encodes a transcription factor that drives cell growth and proliferation. Since many human cancers express dysregulated MYC, we sought to determine whether HIF-1 would in fact collaborate with dysregulated MYC rather countering its function. Here, using the P493-6 Burkitt's lymphoma model with an inducible MYC, we demonstrate that HIF-1 cooperates with dysregulated c-Myc to promote glycolysis by induction of hexokinase 2, which catalyzes the first step of glycolysis, and pyruvate dehydrogenase kinase 1, which inactivates pyruvate dehydrogenase and diminishes mitochondrial respiration. We also found the collaborative induction of vascular endothelial growth factor (VEGF) by HIF-1 and dysregulated c-Myc. This study reports the previously unsuspected collaboration between HIF-1 and dysregulated MYC and thereby provides additional insights into the regulation of VEGF and the Warburg effect, which describes the propensity for cancer cells to convert glucose to lactate.
Collapse
Affiliation(s)
- Jung-whan Kim
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
132
|
Abstract
PURPOSE OF REVIEW Angiogenesis and inflammation are important features in atherosclerotic plaque destabilization. The transcription factors hypoxia-inducible factor-1alpha and Notch are key regulators of angiogenesis. In addition, hypoxia-inducible factor-1alpha has been linked to regulation of inflammatory processes and innate immunity. This review will document how hypoxia-inducible factor-mediated signaling pathways are initiated in hypoxic cells, and how the hypoxia-inducible factor and Notch-dependent signaling pathways are functionally integrated. RECENT FINDINGS Activation of the hypoxia-inducible factor-mediated signaling events by hypoxia is complex and regulated by a cascade of molecular events that will be reviewed in detail. The activated form of hypoxia-inducible factor enhances Notch-dependent activation of Notch target genes, thereby providing a mechanism by which hypoxia can regulate the differentiation status of a cell. Recent observations implicate the Notch signaling pathway in proper specification of cell identity, position and behavior in a developing blood vessel sprout, and the hypoxia-inducible factor-mediated signaling pathway is critical for induction of expression of vascular endothelial growth factor. SUMMARY Hypoxia-inducible factor and Notch transcription factors represent potentially attractive targets for regulation of angiogenesis and possibly inflammation. In view of the pleiotropic effects of these transcription factors, however, successful targeting of these signaling pathways will require the development of gene specific (and possibly tissue-specific) modulators and extensive validation in relevant model systems.
Collapse
Affiliation(s)
- Jorge L Ruas
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
133
|
Abstract
There is a renewed interest in the ultimate role of fatty acid synthase (FASN)--a key lipogenic enzyme catalysing the terminal steps in the de novo biogenesis of fatty acids--in cancer pathogenesis. Tumour-associated FASN, by conferring growth and survival advantages rather than functioning as an anabolic energy-storage pathway, appears to necessarily accompany the natural history of most human cancers. A recent identification of cross-talk between FASN and well-established cancer-controlling networks begins to delineate the oncogenic nature of FASN-driven lipogenesis. FASN, a nearly-universal druggable target in many human carcinomas and their precursor lesions, offers new therapeutic opportunities for metabolically treating and preventing cancer.
Collapse
Affiliation(s)
- Javier A Menendez
- Translational Research Unit, Catalan Institute of Oncology (ICO), Health Services Division of Catalonia, Girona Biomedical Research Institute (IdIBGi), Medical Oncology, Josep Trueta University Hospital of Girona, 17,007 Girona, Catalonia, Spain
| | | |
Collapse
|
134
|
Abstract
Rapidly growing tumors invariably contain hypoxic regions. Adaptive response to hypoxia through angiogenesis, enhanced glucose metabolism and diminished but optimized mitochondrial respiration confers survival and growth advantage to hypoxic tumor cells. In this review, the roles of hypoxia, the hypoxia inducible factors, oncogenes and tumor suppressors in metabolic adaptation of tumors are discussed. These new insights into hypoxic metabolic alterations in tumors will hopefully lead us to target tumor bioenergetics for the treatment of cancers.
Collapse
Affiliation(s)
- Jung-whan Kim
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
135
|
Galmiche A, Fueller J. RAF kinases and mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1256-62. [PMID: 17442414 DOI: 10.1016/j.bbamcr.2006.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 10/19/2006] [Accepted: 10/20/2006] [Indexed: 01/29/2023]
Abstract
Over the past decade, several investigators reported that a fraction of the RAF kinases are recruited to the mitochondria. Although we are still far from a global understanding of the molecular consequences of RAF translocation on mitochondrial physiology and metabolism, the recent description of some molecular interactions that are established by C-RAF in this organelle, principally with the proteins Bcl-2 and Bag-1, provides some clues. Here, we discuss the possible contribution of RAF targeting to mitochondria to their modulation of apoptosis signaling, as well as to this organelle's physiology. In addition, we discuss the possible modulation of the mitochondrial metabolism by RAF oncogenes in the context of cancer.
Collapse
Affiliation(s)
- Antoine Galmiche
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), University of Würzburg, D-97078 Würzburg, Germany.
| | | |
Collapse
|
136
|
López-Lázaro M. Digitoxin as an anticancer agent with selectivity for cancer cells: possible mechanisms involved. Expert Opin Ther Targets 2007; 11:1043-53. [PMID: 17665977 DOI: 10.1517/14728222.11.8.1043] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Accumulating preclinical and clinical data suggest that the cardiac drug digitoxin might be used in cancer therapy. Recent reports have shown that digitoxin can inhibit the growth and induce apoptosis in cancer cells at concentrations commonly found in the plasma of cardiac patients treated with this drug. Several mechanisms have been associated with the anticancer activity of digitoxin, yet at present it is unknown why malignant cells are more susceptible to this cardiac glycoside than non-malignant cells. This report analyses the possible anticancer mechanisms of digitoxin and proposes that the inhibition of glycolysis may be a key mechanism by which this natural product selectively targets cancer cells. Finally, whether or not there is enough evidence to support the clinical evaluation of digitoxin in patients with cancer is discussed.
Collapse
Affiliation(s)
- Miguel López-Lázaro
- University of Seville, Department of Pharmacology, Faculty of Pharmacy, Sevilla, Spain.
| |
Collapse
|
137
|
Bensaad K, Vousden KH. p53: new roles in metabolism. Trends Cell Biol 2007; 17:286-91. [PMID: 17481900 DOI: 10.1016/j.tcb.2007.04.004] [Citation(s) in RCA: 228] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 03/07/2007] [Accepted: 04/23/2007] [Indexed: 11/22/2022]
Abstract
Virtually all cancers show metabolic changes that result in upregulation of glycolysis and glucose consumption. Although discovered in the 1920s, how this glycolytic switch happens, and whether it is a cause or a consequence of the malignant process, has remained a matter of debate. The p53 tumor suppressor gene, discovered some 30 years ago, has a key role in preventing cancer development. Recent discoveries revealing new functions for p53 in the regulation of glucose metabolism and oxidative stress have brought together these two venerable fields of cancer biology. These activities of p53 appear to be key in tumor suppression, and shed some light on the pathways that underlie the metabolic changes in cancer cells.
Collapse
Affiliation(s)
- Karim Bensaad
- The Beatson Institute for Cancer Research, Glasgow, UK
| | | |
Collapse
|
138
|
Lum JJ, Bui T, Gruber M, Gordan JD, DeBerardinis RJ, Covello KL, Simon MC, Thompson CB. The transcription factor HIF-1alpha plays a critical role in the growth factor-dependent regulation of both aerobic and anaerobic glycolysis. Genes Dev 2007; 21:1037-49. [PMID: 17437992 PMCID: PMC1855230 DOI: 10.1101/gad.1529107] [Citation(s) in RCA: 321] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 03/08/2007] [Indexed: 01/20/2023]
Abstract
Mammalian cells are believed to have a cell-intrinsic ability to increase glucose metabolism in response to hypoxia. Here we show that the ability of hematopoietic cells to up-regulate anaerobic glycolysis in response to hypoxia is dependent on receptor-mediated signal transduction. In the absence of growth factor signaling, hematopoietic cells fail to express hypoxia-inducible transcription factor (Hif-1alpha) mRNA. Growth factor-deprived hematopoietic cells do not engage in glucose-dependent anabolic synthesis and neither express Hif-1alpha mRNA nor require HIF-1alpha protein to regulate cell survival in response to hypoxia. However, HIF-1alpha is adaptive for the survival of growth factor-stimulated cells, as suppression of HIF-1alpha results in death when growing cells are exposed to hypoxia. Growth factor-dependent HIF-1alpha expression reprograms the intracellular fate of glucose, resulting in decreased glucose-dependent anabolic synthesis and increased lactate production, an effect that is enhanced when HIF-1alpha protein is stabilized by hypoxia. Together, these data suggest that HIF-1alpha contributes to the regulation of growth factor-stimulated glucose metabolism even in the absence of hypoxia.
Collapse
Affiliation(s)
- Julian J. Lum
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Thi Bui
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Michaela Gruber
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - John D. Gordan
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Ralph J. DeBerardinis
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Division of Child Development, Rehabilitation Medicine and Metabolic Disease, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Kelly L. Covello
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - M. Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Philadelphia, Pennsylvania 19104, USA
| | - Craig B. Thompson
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
139
|
Wu R, Hendrix-Lucas N, Kuick R, Zhai Y, Schwartz DR, Akyol A, Hanash S, Misek DE, Katabuchi H, Williams BO, Fearon ER, Cho KR. Mouse model of human ovarian endometrioid adenocarcinoma based on somatic defects in the Wnt/beta-catenin and PI3K/Pten signaling pathways. Cancer Cell 2007; 11:321-33. [PMID: 17418409 DOI: 10.1016/j.ccr.2007.02.016] [Citation(s) in RCA: 244] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Revised: 01/05/2007] [Accepted: 02/21/2007] [Indexed: 01/09/2023]
Abstract
One histologic subtype of ovarian carcinoma, ovarian endometrioid adenocarcinoma (OEA), frequently harbors mutations that constitutively activate Wnt/beta-catenin-dependent signaling. We now show that defects in the PI3K/Pten and Wnt/beta-catenin signaling pathways often occur together in a subset of human OEAs, suggesting their cooperation during OEA pathogenesis. Deregulation of these two pathways in the murine ovarian surface epithelium by conditional inactivation of the Pten and Apc tumor suppressor genes results in the formation of adenocarcinomas morphologically similar to human OEAs with 100% penetrance, short latency, and rapid progression to metastatic disease in upwards of 75% of mice. The biological behavior and gene expression patterns of the murine cancers resemble those of human OEAs with defects in the Wnt/beta-catenin and PI3K/Pten pathways.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/genetics
- Adenocarcinoma, Clear Cell/metabolism
- Adenocarcinoma, Clear Cell/pathology
- Adenocarcinoma, Mucinous/genetics
- Adenocarcinoma, Mucinous/metabolism
- Adenocarcinoma, Mucinous/pathology
- Adenomatous Polyposis Coli Protein/genetics
- Adenomatous Polyposis Coli Protein/physiology
- Animals
- Carcinoma, Endometrioid/genetics
- Carcinoma, Endometrioid/metabolism
- Carcinoma, Endometrioid/pathology
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/pathology
- Disease Models, Animal
- Epithelium/metabolism
- Epithelium/pathology
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Mice
- Mutation
- Neoplasm Staging
- Oligonucleotide Array Sequence Analysis
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Ovary/metabolism
- Ovary/pathology
- PTEN Phosphohydrolase/genetics
- PTEN Phosphohydrolase/physiology
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Signal Transduction
- Survival Rate
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Wnt1 Protein/genetics
- Wnt1 Protein/metabolism
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Rong Wu
- Department of Pathology, Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Abstract
The first identified biochemical hallmark of tumor cells was a shift in glucose metabolism from oxidative phosphorylation to aerobic glycolysis. We now know that much of this metabolic conversion is controlled by specific transcriptional programs. Recent studies suggest that activation of the hypoxia-inducible factor (HIF) is a common consequence of a wide variety of mutations underlying human cancer. HIF stimulates expression of glycolytic enzymes and decreases reliance on mitochondrial oxidative phosphorylation in tumor cells, which occurs even under aerobic conditions. In addition, recent efforts have also connected the master metabolic regulator AMP-activated protein kinase (AMPK) to several human tumor suppressors. Several promising therapeutic strategies based on modulation of AMPK, HIF and other metabolic targets have been proposed to exploit the addiction of tumor cells to increased glucose uptake and glycolysis.
Collapse
Affiliation(s)
- Reuben J Shaw
- Dulbecco Center for Cancer Research, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|