Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Endocrinology ; 164(4)2023 02 11.
Article in English | MEDLINE | ID: mdl-36763043

ABSTRACT

Pheochromocytomas (PCC) and paragangliomas (PGL) are rare neuroendocrine tumors with limited curative treatment options outside of surgical resection. Patients with mutations in succinate dehydrogenase subunit B (SDHB) are at an increased risk of malignant and aggressive disease. As cation channels are associated with tumorigenesis, we studied the expression and activity of cation channels from the Degenerin superfamily in a progenitor cell line derived from a human PCC. hPheo1 wild-type (WT) and SDHB knockdown (KD) cells were studied to investigate whether epithelial sodium channels (ENaC) and acid-sensing ion channels (ASIC) are regulated by the activity of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). First, we performed targeted metabolomic studies and quantified changes in glycolysis pathway intermediates and citric acid cycle intermediates using hPheo1 WT cells and SDHB KD cells. Next, we performed protein biochemistry and electrophysiology studies to characterize the protein expression and activity, respectively, of these ion channels. Our western blot experiments show both ENaC alpha and ASIC1/2 are expressed in both hPheo1 WT and SDHB KD cells, with lower levels of a cleaved 60 kDa form of ENaC in SDHB KD cells. Single-channel patch clamp studies corroborate these results and further indicate channel activity is decreased in SDHB KD cells. Additional experiments showed a more significant decreased membrane potential in SDHB KD cells, which were sensitive to amiloride compared to WT cells. We provide evidence for the differential expression and activity of ENaC and ASIC hybrid channels in hPheo1 WT and SDHB KD cells, providing an important area of investigation in understanding SDHB-related disease.


Subject(s)
Adrenal Gland Neoplasms , Pheochromocytoma , Humans , Epithelial Sodium Channels/metabolism , Acid Sensing Ion Channels/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Cations/metabolism , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism
2.
Metabolites ; 12(9)2022 Aug 27.
Article in English | MEDLINE | ID: mdl-36144205

ABSTRACT

Burn injury remains a significant public health issue worldwide. Metabolic derangements are a major complication of burn injury and negatively affect the clinical outcomes of severely burned patients. These metabolic aberrations include muscle wasting, hypermetabolism, hyperglycemia, hyperlactatemia, insulin resistance, and mitochondrial dysfunction. However, little is known about the impact of burn injury on the metabolome profile in skeletal muscle. We have previously shown that farnesyltransferase inhibitor (FTI) reverses burn injury-induced insulin resistance, mitochondrial dysfunction, and the Warburg effect in mouse skeletal muscle. To evaluate metabolome composition, targeted quantitative analysis was performed using capillary electrophoresis mass spectrometry in mouse skeletal muscle. Principal component analysis (PCA), partial least squares discriminant analysis (PLS-DA), and hierarchical cluster analysis demonstrated that burn injury induced a global change in metabolome composition. FTI treatment almost completely prevented burn injury-induced alterations in metabolite levels. Pathway analysis revealed that the pathways most affected by burn injury were purine, glutathione, ß-alanine, glycine, serine, and threonine metabolism. Burn injury induced a suppressed oxidized to reduced nicotinamide adenine dinucleotide (NAD+/NADH) ratio as well as oxidative stress and adenosine triphosphate (ATP) depletion, all of which were reversed by FTI. Moreover, our data raise the possibility that burn injury may lead to increased glutaminolysis and reductive carboxylation in mouse skeletal muscle.

3.
Front Oncol ; 12: 968351, 2022.
Article in English | MEDLINE | ID: mdl-36059707

ABSTRACT

Glioblastoma (GBM), similar to most cancers, is dependent on fermentation metabolism for the synthesis of biomass and energy (ATP) regardless of the cellular or genetic heterogeneity seen within the tumor. The transition from respiration to fermentation arises from the documented defects in the number, the structure, and the function of mitochondria and mitochondrial-associated membranes in GBM tissue. Glucose and glutamine are the major fermentable fuels that drive GBM growth. The major waste products of GBM cell fermentation (lactic acid, glutamic acid, and succinic acid) will acidify the microenvironment and are largely responsible for drug resistance, enhanced invasion, immunosuppression, and metastasis. Besides surgical debulking, therapies used for GBM management (radiation, chemotherapy, and steroids) enhance microenvironment acidification and, although often providing a time-limited disease control, will thus favor tumor recurrence and complications. The simultaneous restriction of glucose and glutamine, while elevating non-fermentable, anti-inflammatory ketone bodies, can help restore the pH balance of the microenvironment while, at the same time, providing a non-toxic therapeutic strategy for killing most of the neoplastic cells.

5.
Metabolism ; 110: 154297, 2020 09.
Article in English | MEDLINE | ID: mdl-32562798

ABSTRACT

BACKGROUND: Pheochromocytomas (PCCs) and paragangliomas (PGLs) are neuroendocrine tumors that are mostly benign. Metastatic disease does occur in about 10% of cases of PCC and up to 25% of PGL, and for these patients no effective therapies are available. Patients with mutations in the succinate dehydrogenase subunit B (SDHB) gene tend to have metastatic disease. We hypothesized that a down-regulation in the active succinate dehydrogenase B subunit should result in notable changes in cellular metabolic profile and could present a vulnerability point for successful pharmacological targeting. METHODS: Metabolomic analysis was performed on human hPheo1 cells and shRNA SDHB knockdown hPheo1 (hPheo1 SDHB KD) cells. Additional analysis of 115 human fresh frozen samples was conducted. In vitro studies using N1,N11-diethylnorspermine (DENSPM) and N1,N12- diethylspermine (DESPM) treatments were carried out. DENSPM efficacy was assessed in human cell line derived mouse xenografts. RESULTS: Components of the polyamine pathway were elevated in hPheo1 SDHB KD cells compared to wild-type cells. A similar observation was noted in SDHx PCC/PGLs tissues compared to their non-mutated counterparts. Specifically, spermidine, and spermine were significantly elevated in SDHx-mutated PCC/PGLs, with a similar trend in hPheo1 SDHB KD cells. Polyamine pathway inhibitors DENSPM and DESPM effectively inhibited growth of hPheo1 cells in vitro as well in mouse xenografts. CONCLUSIONS: This study demonstrates overactive polyamine pathway in PCC/PGL with SDHB mutations. Treatment with polyamine pathway inhibitors significantly inhibited hPheo1 cell growth and led to growth suppression in xenograft mice treated with DENSPM. These studies strongly implicate the polyamine pathway in PCC/PGL pathophysiology and provide new foundation for exploring the role for polyamine analogue inhibitors in treating metastatic PCC/PGL. PRéCIS: Cell line metabolomics on hPheo1 cells and PCC/PGL tumor tissue indicate that the polyamine pathway is activated. Polyamine inhibitors in vitro and in vivo demonstrate that polyamine inhibitors are promising for malignant PCC/PGL treatment. However, further research is warranted.


Subject(s)
Adrenal Gland Neoplasms/drug therapy , Biogenic Polyamines/antagonists & inhibitors , Paraganglioma/drug therapy , Pheochromocytoma/drug therapy , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Animals , Biogenic Polyamines/metabolism , Cell Line, Tumor , Humans , Male , Metabolomics , Mice , Mutation , Paraganglioma/genetics , Paraganglioma/metabolism , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Succinate Dehydrogenase/genetics , Xenograft Model Antitumor Assays
6.
Neurochem Res ; 44(10): 2392-2404, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31025151

ABSTRACT

No major advances have been made in improving overall survival for glioblastoma (GBM) in almost 100 years. The current standard of care (SOC) for GBM involves immediate surgical resection followed by radiotherapy with concomitant temozolomide chemotherapy. Corticosteroid (dexamethasone) is often prescribed to GBM patients to reduce tumor edema and inflammation. The SOC disrupts the glutamate-glutamine cycle thus increasing availability of glucose and glutamine in the tumor microenvironment. Glucose and glutamine are the prime fermentable fuels that underlie therapy resistance and drive GBM growth through substrate level phosphorylation in the cytoplasm and the mitochondria, respectively. Emerging evidence indicates that ketogenic metabolic therapy (KMT) can reduce glucose availability while elevating ketone bodies that are neuroprotective and non-fermentable. Information is presented from preclinical and case report studies showing how KMT could target tumor cells without causing neurochemical damage thus improving progression free and overall survival for patients with GBM.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Ketone Bodies/metabolism , Standard of Care , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Glucose/metabolism , Glutamine/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Temozolomide/therapeutic use , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
7.
Biochim Biophys Acta Bioenerg ; 1859(9): 975-983, 2018 09.
Article in English | MEDLINE | ID: mdl-29580805

ABSTRACT

Succinate is known to act as an inflammatory signal in classically activated macrophages through stabilization of HIF-1α leading to IL-1ß production. Relevant to this, hypoxia is known to drive succinate accumulation and release into the extracellular milieu. The metabolic alterations associated with succinate release during inflammation and under hypoxia are poorly understood. Data are presented showing that Mycoplasma arginini infection of VM-M3 cancer cells enhances the Warburg effect associated with succinate production in mitochondria and eventual release into the extracellular milieu. We investigated how succinate production and release was related to the changes of other soluble metabolites, including itaconate and 2-HG. Furthermore, we found that hypoxia alone could induce succinate release from the VM-M3 cells and that this could occur in the absence of glucose-driven lactate production. Our results elucidate metabolic pathways responsible for succinate accumulation and release in cancer cells, thus identifying potential targets involved in both inflammation and hypoxia. This article is part of a Special Issue entitled 20th European Bioenergetics Conference, edited by László Zimányi and László Tretter.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Hypoxia/complications , Inflammation/complications , Mycoplasma Infections/complications , Mycoplasma/pathogenicity , Succinates/metabolism , Animals , Brain Neoplasms/etiology , Brain Neoplasms/metabolism , Energy Metabolism , Glioblastoma/etiology , Glioblastoma/metabolism , Metabolome , Mice , Tumor Cells, Cultured
8.
Cell Rep ; 18(4): 991-1004, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28122247

ABSTRACT

Targeting defects in metabolism is an underutilized strategy for the treatment of cancer. Arginine auxotrophy resulting from the silencing of argininosuccinate synthetase 1 (ASS1) is a common metabolic alteration reported in a broad range of aggressive cancers. To assess the metabolic effects that arise from acute and chronic arginine starvation in ASS1-deficient cell lines, we performed metabolite profiling. We found that pharmacologically induced arginine depletion causes increased serine biosynthesis, glutamine anaplerosis, oxidative phosphorylation, and decreased aerobic glycolysis, effectively inhibiting the Warburg effect. The reduction of glycolysis in cells otherwise dependent on aerobic glycolysis is correlated with reduced PKM2 expression and phosphorylation and upregulation of PHGDH. Concurrent arginine deprivation and glutaminase inhibition was found to be synthetic lethal across a spectrum of ASS1-deficient tumor cell lines and is sufficient to cause in vivo tumor regression in mice. These results identify two synthetic lethal therapeutic strategies exploiting metabolic vulnerabilities of ASS1-negative cancers.


Subject(s)
Argininosuccinate Synthase/genetics , Glutamine/metabolism , Serine/biosynthesis , Animals , Arginine/chemistry , Argininosuccinate Synthase/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Citric Acid Cycle/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Glucose/metabolism , Glucose/pharmacology , Glutaminase/antagonists & inhibitors , Glutaminase/genetics , Glutaminase/metabolism , Glutamine/pharmacology , Glycolysis/drug effects , Humans , Hydrolases/pharmacology , Membrane Proteins/metabolism , Metabolomics , Mice , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Phosphorylation/drug effects , Polyethylene Glycols/pharmacology , RNA Interference , Thyroid Hormones/metabolism , Up-Regulation/drug effects , Thyroid Hormone-Binding Proteins
9.
Cancer Res ; 76(15): 4430-42, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27280394

ABSTRACT

Intratumoral hypoxia stimulates enrichment of breast cancer stem cells (BCSC), which are critical for metastasis and patient mortality. Here we report a metabolic adaptation that is required for hypoxia-induced BCSC enrichment and metastasis. Hypoxia-inducible factors coordinately regulate expression of genes encoding phosphoglycerate dehydrogenase (PHGDH) and five downstream enzymes in the serine synthesis pathway and mitochondrial one-carbon (folate) cycle. RNAi-mediated silencing of PHGDH expression in both estrogen receptor-positive and negative breast cancer cells led to decreased NADPH levels, disturbed mitochondrial redox homeostasis, and increased apoptosis, which abrogated BCSC enrichment under hypoxic conditions. PHGDH-deficient cells exhibited increased oxidant levels and apoptosis, as well as loss of BCSC enrichment, in response to treatment with carboplatin or doxorubicin. PHGDH-deficient cells were relatively weakly tumorigenic and tumors that did form were deficient in BCSCs, abolishing metastatic capacity. Our findings highlight a role for PHGDH in the formation of secondary (recurrent or metastatic) tumors, with potential implications for therapeutic targeting of advanced cancers. Cancer Res; 76(15); 4430-42. ©2016 AACR.


Subject(s)
Breast Neoplasms/genetics , Lung Neoplasms/secondary , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Phosphoglycerate Dehydrogenase/metabolism , Breast Neoplasms/pathology , Female , Humans , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Transfection
10.
Cell Metab ; 23(5): 867-80, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27133130

ABSTRACT

Mammary epithelial cells transition between periods of proliferation and quiescence during development, menstrual cycles, and pregnancy, and as a result of oncogenic transformation. Utilizing an organotypic 3D tissue culture model coupled with quantitative metabolomics and proteomics, we identified significant differences in glutamate utilization between proliferating and quiescent cells. Relative to quiescent cells, proliferating cells catabolized more glutamate via transaminases to couple non-essential amino acid (NEAA) synthesis to α-ketoglutarate generation and tricarboxylic acid (TCA) cycle anaplerosis. As cells transitioned to quiescence, glutamine consumption and transaminase expression were reduced, while glutamate dehydrogenase (GLUD) was induced, leading to decreased NEAA synthesis. Highly proliferative human tumors display high transaminase and low GLUD expression, suggesting that proliferating cancer cells couple glutamine consumption to NEAA synthesis to promote biosynthesis. These findings describe a competitive and partially redundant relationship between transaminases and GLUD, and they reveal how coupling of glutamate-derived carbon and nitrogen metabolism can be regulated to support cell proliferation.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Glutamic Acid/metabolism , Mammary Glands, Human/cytology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Proliferation , Cells, Cultured , Female , Glutamate Dehydrogenase/metabolism , Humans , Metabolomics , Models, Biological , Nitrogen Isotopes , Phosphatidylinositol 3-Kinases/metabolism , Proteomics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transaminases/metabolism
11.
Cancer Metab ; 3: 6, 2015.
Article in English | MEDLINE | ID: mdl-26023330

ABSTRACT

BACKGROUND: Systematic quantitative methodologies are needed to understand the heterogeneity of cell metabolism across cell types in normal physiology, disease, and treatment. Metabolic flux analysis (MFA) can be used to infer steady state fluxes, but it does not apply for transient dynamics. Kinetic flux profiling (KFP) can be used in the context of transient dynamics, and it is the current gold standard. However, KFP requires measurements at several time points, limiting its use in high-throughput applications. RESULTS: Here we propose transient MFA (tMFA) as a cost-effective methodology to quantify metabolic fluxes using metabolomics and isotope tracing. tMFA exploits the time scale separation between the dynamics of different metabolites to obtain mathematical equations relating metabolic fluxes to metabolite concentrations and isotope fractions. We show that the isotope fractions of serine and glycine are at steady state 8 h after addition of a tracer, while those of purines and glutathione are following a transient dynamics with an approximately constant turnover rate per unit of metabolite, supporting the application of tMFA to the analysis of folate metabolism. Using tMFA, we investigate the heterogeneity of folate metabolism and the response to the antifolate methotrexate in breast cancer cells. Our analysis indicates that methotrexate not only inhibits purine synthesis but also induces an increase in the AMP/ATP ratio, activation of AMP kinase (AMPK), and the inhibition of protein and glutathione synthesis. We also find that in some cancer cells, the generation of one-carbon units from serine exceeds the biosynthetic demand. CONCLUSIONS: This work validates tMFA as a cost-effective methodology to investigate cell metabolism. Using tMFA, we have shown that the effects of treatment with the antifolate methotrexate extend beyond inhibition of purine synthesis and propagate to other pathways in central metabolism.

12.
Nature ; 520(7547): 363-7, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25855294

ABSTRACT

Cancer cells adapt their metabolic processes to support rapid proliferation, but less is known about how cancer cells alter metabolism to promote cell survival in a poorly vascularized tumour microenvironment. Here we identify a key role for serine and glycine metabolism in the survival of brain cancer cells within the ischaemic zones of gliomas. In human glioblastoma multiforme, mitochondrial serine hydroxymethyltransferase (SHMT2) and glycine decarboxylase (GLDC) are highly expressed in the pseudopalisading cells that surround necrotic foci. We find that SHMT2 activity limits that of pyruvate kinase (PKM2) and reduces oxygen consumption, eliciting a metabolic state that confers a profound survival advantage to cells in poorly vascularized tumour regions. GLDC inhibition impairs cells with high SHMT2 levels as the excess glycine not metabolized by GLDC can be converted to the toxic molecules aminoacetone and methylglyoxal. Thus, SHMT2 is required for cancer cells to adapt to the tumour environment, but also renders these cells sensitive to glycine cleavage system inhibition.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Glycine Hydroxymethyltransferase/metabolism , Glycine/metabolism , Ischemia/metabolism , Acetone/analogs & derivatives , Acetone/metabolism , Acetone/toxicity , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/enzymology , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Female , Glioblastoma/blood supply , Glioblastoma/enzymology , Glycine Dehydrogenase (Decarboxylating)/antagonists & inhibitors , Glycine Dehydrogenase (Decarboxylating)/metabolism , Humans , Ischemia/enzymology , Ischemia/pathology , Mice , Necrosis , Oxygen Consumption , Pyruvaldehyde/metabolism , Pyruvaldehyde/toxicity , Pyruvate Kinase/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
13.
PLoS One ; 8(3): e58601, 2013.
Article in English | MEDLINE | ID: mdl-23505541

ABSTRACT

We report that a single growth factor, NM23-H1, enables serial passaging of both human ES and iPS cells in the absence of feeder cells, their conditioned media or bFGF in a fully defined xeno-free media on a novel defined, xeno-free surface. Stem cells cultured in this system show a gene expression pattern indicative of a more "naïve" state than stem cells grown in bFGF-based media. NM23-H1 and MUC1* growth factor receptor cooperate to control stem cell self-replication. By manipulating the multimerization state of NM23-H1, we override the stem cell's inherent programming that turns off pluripotency and trick the cells into continuously replicating as pluripotent stem cells. Dimeric NM23-H1 binds to and dimerizes the extra cellular domain of the MUC1* transmembrane receptor which stimulates growth and promotes pluripotency. Inhibition of the NM23-H1/MUC1* interaction accelerates differentiation and causes a spike in miR-145 expression which signals a cell's exit from pluripotency.


Subject(s)
NM23 Nucleoside Diphosphate Kinases/pharmacology , Stem Cells/drug effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Binding, Competitive , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Culture Media, Conditioned/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Ligands , MicroRNAs/genetics , MicroRNAs/metabolism , Mucin-1/immunology , Mucin-1/metabolism , NM23 Nucleoside Diphosphate Kinases/chemistry , NM23 Nucleoside Diphosphate Kinases/metabolism , Protein Binding/drug effects , Protein Multimerization , Stem Cells/cytology , Stem Cells/metabolism
14.
Epilepsy Res ; 100(3): 310-26, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21885251

ABSTRACT

Malignant brain cancer persists as a major disease of morbidity and mortality. The failure to recognize brain cancer as a disease of energy metabolism has contributed in large part to the failure in management. As long as brain tumor cells have access to glucose and glutamine, the disease will progress. The current standard of care provides brain tumors with access to glucose and glutamine. The high fat low carbohydrate ketogenic diet (KD) will target glucose availability and possibly that of glutamine when administered in carefully restricted amounts to reduce total caloric intake and circulating levels of glucose. The restricted KD (RKD) targets major signaling pathways associated with glucose and glutamine metabolism including the IGF-1/PI3K/Akt/Hif pathway. The RKD is anti-angiogenic, anti-invasive, anti-inflammatory, and pro-apoptotic when evaluated in mice with malignant brain cancer. The therapeutic efficacy of the restricted KD can be enhanced when combined with drugs that also target glucose and glutamine. Therapeutic efficacy of the RKD was also seen against malignant gliomas in human case reports. Hence, the RKD can be an effective non-toxic therapeutic option to the current standard of care for inhibiting the growth and invasive properties of malignant brain cancer.


Subject(s)
Brain Neoplasms/diet therapy , Caloric Restriction , Diet, Ketogenic , Energy Metabolism/drug effects , Glioblastoma/diet therapy , Animals , Glucose/metabolism , Humans
15.
Biochim Biophys Acta ; 1807(6): 577-94, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20804725

ABSTRACT

Malignant brain tumors are a significant health problem in children and adults. Conventional therapeutic approaches have been largely unsuccessful in providing long-term management. As primarily a metabolic disease, malignant brain cancer can be managed through changes in metabolic environment. In contrast to normal neurons and glia, which readily transition to ketone bodies (ß-hydroxybutyrate) for energy under reduced glucose, malignant brain tumors are strongly dependent on glycolysis for energy. The transition from glucose to ketone bodies as a major energy source is an evolutionary conserved adaptation to food deprivation that permits the survival of normal cells during extreme shifts in nutritional environment. Only those cells with a flexible genome and normal mitochondria can effectively transition from one energy state to another. Mutations restrict genomic and metabolic flexibility thus making tumor cells more vulnerable to energy stress than normal cells. We propose an alternative approach to brain cancer management that exploits the metabolic flexibility of normal cells at the expense of the genetically defective and metabolically challenged tumor cells. This approach to brain cancer management is supported from recent studies in mice and humans treated with calorie restriction and the ketogenic diet. Issues of implementation and use protocols are presented for the metabolic management of brain cancer.


Subject(s)
Brain Neoplasms/therapy , Energy Metabolism/physiology , Glioblastoma/therapy , Adult , Animals , Brain Neoplasms/complications , Brain Neoplasms/diet therapy , Brain Neoplasms/metabolism , Caloric Restriction , Child , Diet, Ketogenic , Disease Models, Animal , Glioblastoma/complications , Glioblastoma/diet therapy , Glioblastoma/metabolism , Humans , Mice , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/therapy , Models, Biological
17.
ASN Neuro ; 2(3): e00038, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20664705

ABSTRACT

GBM (glioblastoma multiforme) is the most aggressive and invasive form of primary human brain cancer. We recently developed a novel brain cancer model in the inbred VM mouse strain that shares several characteristics with human GBM. Using bioluminescence imaging, we tested the efficacy of CR (calorie restriction) for its ability to reduce tumour size and invasion. CR targets glycolysis and rapid tumour cell growth in part by lowering circulating glucose levels. The VM-M3 tumour cells were implanted intracerebrally in the syngeneic VM mouse host. Approx. 12-15 days post-implantation, brains were removed and both ipsilateral and contralateral hemispheres were imaged to measure bioluminescence of invading tumour cells. CR significantly reduced the invasion of tumour cells from the implanted ipsilateral hemisphere into the contralateral hemisphere. The total percentage of Ki-67-stained cells within the primary tumour and the total number of blood vessels was also significantly lower in the CR-treated mice than in the mice fed ad libitum, suggesting that CR is anti-proliferative and anti-angiogenic. Our findings indicate that the VM-M3 GBM model is a valuable tool for studying brain tumour cell invasion and for evaluating potential therapeutic approaches for managing invasive brain cancer. In addition, we show that CR can be effective in reducing malignant brain tumour growth and invasion.


Subject(s)
Brain Neoplasms/diet therapy , Caloric Restriction/methods , Disease Models, Animal , Glioblastoma/diet therapy , Neoplasm Invasiveness , Animals , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , Glioblastoma/pathology , Glioblastoma/prevention & control , Male , Mice , Mice, Inbred Strains , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control
18.
Int J Cancer ; 127(10): 2478-85, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20473919

ABSTRACT

Metastatic cancer is a major cause of morbidity and mortality. Current therapeutic options consist of chemotherapy, radiation or targeted therapies. However, these therapies are often toxic, effective over a small range of cancer types or result in drug resistance. Therefore, a more global, less toxic strategy for the management of metastatic cancer is required. Although most cancers display increased glucose metabolism, glutamine is also a major energy substrate for many cancers. We evaluated the antimetastatic potential of 6-diazo-5-oxo-L-norleucine (DON), a glutamine analog, using the new VM mouse model of systemic metastasis. We found that primary tumor growth was ∼20-fold less in DON-treated mice than in untreated control mice. We also found that DON treatment inhibited metastasis to liver, lung and kidney as detected by bioluminescence imaging and histology. Our findings provide proof of concept that metabolic therapies targeting glutamine metabolism can manage systemic metastatic cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Brain Neoplasms/drug therapy , Diazooxonorleucine/pharmacology , Glutamine/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Caloric Restriction , Cell Growth Processes/drug effects , Cell Line, Tumor , Cerebrum/metabolism , Cerebrum/pathology , Drug Delivery Systems , Female , Glucose/deficiency , Glucose/metabolism , Glutamine/deficiency , Male , Mice , Neoplasm Metastasis
19.
Nutr Metab (Lond) ; 7: 7, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20181022

ABSTRACT

Emerging evidence indicates that impaired cellular energy metabolism is the defining characteristic of nearly all cancers regardless of cellular or tissue origin. In contrast to normal cells, which derive most of their usable energy from oxidative phosphorylation, most cancer cells become heavily dependent on substrate level phosphorylation to meet energy demands. Evidence is reviewed supporting a general hypothesis that genomic instability and essentially all hallmarks of cancer, including aerobic glycolysis (Warburg effect), can be linked to impaired mitochondrial function and energy metabolism. A view of cancer as primarily a metabolic disease will impact approaches to cancer management and prevention.

20.
J Neurooncol ; 99(2): 165-76, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20069342

ABSTRACT

Glioblastoma multiforme (GBM) is a rapidly progressive disease of morbidity and mortality and is the most common form of primary brain cancer in adults. Lack of appropriate in vivo models has been a major roadblock to developing effective therapies for GBM. A new highly invasive in vivo GBM model is described that was derived from a spontaneous brain tumor (VM-M3) in the VM mouse strain. Highly invasive tumor cells could be identified histologically on the hemisphere contralateral to the hemisphere implanted with tumor cells or tissue. Tumor cells were highly expressive for the chemokine receptor CXCR4 and the proliferation marker Ki-67 and could be identified invading through the pia mater, the vascular system, the ventricular system, around neurons, and over white matter tracts including the corpus callosum. In addition, the brain tumor cells were labeled with the firefly luciferase gene, allowing for non-invasive detection and quantitation through bioluminescent imaging. The VM-M3 tumor has a short incubation time with mortality occurring in 100% of the animals within approximately 15 days. The VM-M3 brain tumor model therefore can be used in a pre-clinical setting for the rapid evaluation of novel anti-invasive therapies.


Subject(s)
Astrocytoma/pathology , Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Disease Models, Animal , Neurons/pathology , Animals , Astrocytoma/genetics , Astrocytoma/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Adhesion , Immunoenzyme Techniques , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neurons/metabolism , RNA, Messenger/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...