41 results on '"Mariia Yuneva"'
Search Results
2. Adapt and conquer: Metabolic flexibility in cancer growth, invasion and evasion
- Author
-
Peter Kreuzaler, Yulia Panina, Joanna Segal, and Mariia Yuneva
- Subjects
Internal medicine ,RC31-1245 - Abstract
Background: It has been known for close to a century that, on average, tumors have a metabolism that is different from those found in healthy tissues. Typically, tumors show a biosynthetic metabolism that distinguishes itself by engaging in large scale aerobic glycolysis, heightened flux through the pentose phosphate pathway, and increased glutaminolysis among other means. However, it is becoming equally clear that non tumorous tissues at times can engage in similar metabolism, while tumors show a high degree of metabolic flexibility reacting to cues, and stresses in their local environment. Scope of the review: In this review, we want to scrutinize historic and recent research on metabolism, comparing and contrasting oncogenic and physiological metabolic states. This will allow us to better define states of bona fide tumor metabolism. We will further contextualize the stress response and the metabolic evolutionary trajectory seen in tumors, and how these contribute to tumor progression. Lastly, we will analyze the implications of these characteristics with respect to therapy response. Major conclusions: In our review, we argue that there is not one single oncogenic state, but rather a diverse set of oncogenic states. These are grounded on a physiological proliferative/wound healing program but distinguish themselves due to their large scale of proliferation, mutations, and transcriptional changes in key metabolic pathways, and the adaptations to widespread stress signals within tumors. We find evidence for the necessity of metabolic flexibility and stress responses in tumor progression and how these responses in turn shape oncogenic progression. Lastly, we find evidence for the notion that the metabolic adaptability of tumors frequently frustrates therapeutic interventions. Keywords: Tumourigenesis, Tumour metabolism, Metabolic flexibility, Central carbon metabolism
- Published
- 2020
- Full Text
- View/download PDF
3. [18F]-(2S,4R)4-Fluoroglutamine PET Imaging of Glutamine Metabolism in Murine Models of Hepatocellular Carcinoma (HCC)
- Author
-
Youngho Seo, Miranda C. Craig, Stephanie T. Murphy, Jinjin Feng, Xin Chen, and Mariia Yuneva
- Subjects
Biology (General) ,QH301-705.5 ,Medical technology ,R855-855.5 - Abstract
Purpose. Quantitative in vivo [18F]-(2S,4R)4-fluoroglutamine ([18F]4-FGln or more simply [18F]FGln) metabolic kinetic parameters are compared with activity levels of glutamine metabolism in different types of hepatocellular carcinoma (HCC). Methods. For this study, we used two transgenic mouse models of HCC induced by protooncogenes, MYC, and MET. Biochemical data have shown that tumors induced by MYC have increased levels of glutamine metabolism compared to those induced by MET. One-hour dynamic [18F]FGln PET data were acquired and reconstructed for fasted MYC mice (n=11 tumors from 7 animals), fasted MET mice (n=8 tumors from 6 animals), fasted FVBN controls (n=8 normal liver regions from 6 animals), nonfasted MYC mice (n=16 tumors from 6 animals), and nonfasted FVBN controls (n=8 normal liver regions from 3 animals). The influx rate constants (K1) using the one-tissue compartment model were derived for each tumor with the left ventricular blood pool input function. Results. Influx rate constants were significantly higher for MYC tumors (K1=0.374±0.133) than for MET tumors (K1=0.141±0.058) under fasting conditions (P=0.0002). Rate constants were also significantly lower for MET tumors (K1=0.141±0.135) than normal livers (K1=0.332±0.179) under fasting conditions (P=0.0123). Fasting conditions tested for MYC tumors and normal livers did not result in any significant difference with P values > 0.005. Conclusion. Higher influx rate constants corresponded to elevated levels of glutamine metabolism as determined by biochemical assays. The data showed that there is a distinctive difference in glutamine metabolism between MYC and MET tumors. Our study has demonstrated the potential of [18F]FGln PET imaging as a tool to assess glutamine metabolism in HCC tumors in vivo with a caution that it may not be able to clearly distinguish HCC tumors from normal liver tissue.
- Published
- 2022
- Full Text
- View/download PDF
4. De novo MYC addiction as an adaptive response of cancer cells to CDK4/6 inhibition
- Author
-
Míriam Tarrado‐Castellarnau, Pedro de Atauri, Josep Tarragó‐Celada, Jordi Perarnau, Mariia Yuneva, Timothy M Thomson, and Marta Cascante
- Subjects
13C metabolic flux analysis ,CDK4/6 ,glutaminase ,MYC ,tumor metabolic reprogramming ,Biology (General) ,QH301-705.5 ,Medicine (General) ,R5-920 - Abstract
Abstract Cyclin‐dependent kinases (CDK) are rational cancer therapeutic targets fraught with the development of acquired resistance by tumor cells. Through metabolic and transcriptomic analyses, we show that the inhibition of CDK4/6 leads to a metabolic reprogramming associated with gene networks orchestrated by the MYC transcription factor. Upon inhibition of CDK4/6, an accumulation of MYC protein ensues which explains an increased glutamine metabolism, activation of the mTOR pathway and blunting of HIF‐1α‐mediated responses to hypoxia. These MYC‐driven adaptations to CDK4/6 inhibition render cancer cells highly sensitive to inhibitors of MYC, glutaminase or mTOR and to hypoxia, demonstrating that metabolic adaptations to antiproliferative drugs unveil new vulnerabilities that can be exploited to overcome acquired drug tolerance and resistance by cancer cells.
- Published
- 2017
- Full Text
- View/download PDF
5. The big picture: exploring the metabolic cross-talk in cancer
- Author
-
Almut Schulze and Mariia Yuneva
- Subjects
Medicine ,Pathology ,RB1-214 - Abstract
Metabolic reprogramming is now well established as one of the hallmarks of cancer. The renewed interest in this topic has spurred a remarkable advance in our understanding of the metabolic alterations in cancer cells and in the tumour microenvironment. Initially, this research focussed on identifying the metabolic processes that provided cancer cells with building blocks for growth or to prevent oxidative damage and death. In addition to providing detailed insight into the mechanisms by which oncogenic signalling pathways modulate metabolic processes, this research also revealed multiple nodes within the metabolic network that can be targeted for the selective elimination of cancer cells. However, recent years have seen a paradigm shift in the field of cancer metabolism; while early studies focussed mainly on the metabolic processes within a cancer cell, recent approaches also consider the impact of metabolic cross-talk between different cell types within the tumour or study cancer within the organismal metabolic context. The Review articles presented in this themed Special Collection of Disease Models & Mechanisms aim to provide an overview of the recent advances in the field. The Collection also contains research articles that describe how metabolic inhibition can improve the efficacy of targeted therapy and introduce a new zebrafish model to study metabolic tumour-host interactions. We also present ‘A model for life’ interviews: a new interview with Karen Vousden and a previously published one with Lewis Cantley that provide insight into these two leaders' personal scientific journeys that resulted in seminal discoveries in the field of cancer metabolism. In this Editorial, we summarise some of the key insights obtained from studying cancer metabolism. We also describe some of the many exciting developments in the field and discuss its future challenges.
- Published
- 2018
- Full Text
- View/download PDF
6. The contributions of cancer cell metabolism to metastasis
- Author
-
Gloria Pascual, Diana Domínguez, Salvador Aznar Benitah, Almut Schulze, and Mariia Yuneva
- Subjects
Cancer ,Metabolism ,Metastasis ,Epigenetics ,Medicine ,Pathology ,RB1-214 - Abstract
Metastasis remains the leading cause of cancer-related deaths worldwide, and our inability to identify the tumour cells that colonize distant sites hampers the development of effective anti-metastatic therapies. However, with recent research advances we are beginning to distinguish metastasis-initiating cells from their non-metastatic counterparts. Importantly, advances in genome sequencing indicate that the acquisition of metastatic competency does not involve the progressive accumulation of driver mutations; moreover, in the early stages of tumorigenesis, cancer cells harbour combinations of driver mutations that endow them with metastatic competency. Novel findings highlight that cells can disseminate to distant sites early during primary tumour growth, remaining dormant and untreatable for long periods before metastasizing. Thus, metastatic cells must require local and systemic influences to generate metastases. This hypothesis suggests that factors derived from our lifestyle, such as our diet, exert a strong influence on tumour progression, and that such factors could be modulated if understood. Here, we summarize the recent findings on how specific metabolic cues modulate the behaviour of metastatic cells and how they influence the genome and epigenome of metastatic cells. We also discuss how crosstalk between metabolism and the epigenome can be harnessed to develop new anti-metastatic therapies.
- Published
- 2018
- Full Text
- View/download PDF
7. A novel zebrafish intestinal tumor model reveals a role for cyp7a1-dependent tumor–liver crosstalk in causing adverse effects on the host
- Author
-
Sora Enya, Koichi Kawakami, Yutaka Suzuki, Shinpei Kawaoka, Almut Schulze, and Mariia Yuneva
- Subjects
Intestinal tumor ,Hepatomegaly ,Liver inflammation ,Growth defect ,cyp7a1 ,Cholesterol metabolism ,Medicine ,Pathology ,RB1-214 - Abstract
The nature of host organs and genes that underlie tumor-induced physiological disruption on the host remains ill-defined. Here, we establish a novel zebrafish intestinal tumor model that is suitable for addressing this issue, and find that hepatic cyp7a1, the rate-limiting factor for synthesizing bile acids [or, in the case of zebrafish, bile alcohol (BA)], is such a host gene. Inducing krasG12D by Gal4 specifically expressed in the posterior intestine resulted in the formation of an intestinal tumor. The local intestinal tumor caused systemic detrimental effects on the host, including liver inflammation, hepatomegaly, growth defects and organismal death. Whole-organism-level gene expression analysis and metabolite measurements revealed that the intestinal tumor reduced total BA levels, possibly via altered expression of hepatic cyp7a1. Genetically overexpressing cyp7a1 in the liver restored BA synthesis and ameliorated tumor-induced liver inflammation, but not other tumor-dependent phenotypes. Thus, we found a previously unknown role of cyp7a1 as the host gene that links the intestinal tumor, hepatic cholesterol–BA metabolism and liver inflammation in tumor-bearing zebrafish larvae. Our model provides an important basis to discover host genes responsible for tumor-induced phenotypes and to uncover mechanisms underlying how tumors adversely affect host organisms.
- Published
- 2018
- Full Text
- View/download PDF
8. SCD1 Expression is dispensable for hepatocarcinogenesis induced by AKT and Ras oncogenes in mice.
- Author
-
Lei Li, Chunmei Wang, Diego F Calvisi, Matthias Evert, Maria G Pilo, Lijie Jiang, Mariia Yuneva, and Xin Chen
- Subjects
Medicine ,Science - Abstract
Increased de novo lipogenesis is one of the major metabolic events in cancer. In human hepatocellular carcinoma (HCC), de novo lipogenesis has been found to be increased and associated with the activation of AKT/mTOR signaling. In mice, overexpression of an activated form of AKT results in increased lipogenesis and hepatic steatosis, ultimately leading to liver tumor development. Hepatocarcinogenesis is dramatically accelerated when AKT is co-expressed with an oncogenic form of N-Ras. SCD1, the major isoform of stearoyl-CoA desaturases, catalyzing the conversion of saturated fatty acids (SFA) into monounsaturated fatty acids (MUFA), is a key enzyme involved in de novo lipogenesis. While many studies demonstrated the requirement of SCD1 for tumor cell growth in vitro, whether SCD1 is necessary for tumor development in vivo has not been previously investigated. Here, we show that genetic ablation of SCD1 neither inhibits lipogenesis and hepatic steatosis in AKT-overexpressing mice nor affects liver tumor development in mice co-expressing AKT and Ras oncogenes. Molecular analysis showed that SCD2 was strongly upregulated in liver tumors from AKT/Ras injected SCD1(-/-) mice. Noticeably, concomitant silencing of SCD1 and SCD2 genes was highly detrimental for the growth of AKT/Ras cells in vitro. Altogether, our study provides the evidence, for the first time, that SCD1 expression is dispensable for AKT/mTOR-dependent hepatic steatosis and AKT/Ras-induced hepatocarcinogenesis in mice. Complete inhibition of stearoyl-CoA desaturase activity may be required to efficiently suppress liver tumor development.
- Published
- 2013
- Full Text
- View/download PDF
9. Figure S4 from Glucose Catabolism in Liver Tumors Induced by c-MYC Can Be Sustained by Various PKM1/PKM2 Ratios and Pyruvate Kinase Activities
- Author
-
Xin Chen, Mariia Yuneva, Diego F. Calvisi, Darjus F. Tschaharganeh, Paul C. Driscoll, Chencheng Xie, Jingxiao Wang, Jiaoyuan Jia, Xinhua Song, Li Che, Junjie Hu, Xiaolei Li, and Andrés Méndez-Lucas
- Abstract
Study design
- Published
- 2023
10. Data from Glucose Catabolism in Liver Tumors Induced by c-MYC Can Be Sustained by Various PKM1/PKM2 Ratios and Pyruvate Kinase Activities
- Author
-
Xin Chen, Mariia Yuneva, Diego F. Calvisi, Darjus F. Tschaharganeh, Paul C. Driscoll, Chencheng Xie, Jingxiao Wang, Jiaoyuan Jia, Xinhua Song, Li Che, Junjie Hu, Xiaolei Li, and Andrés Méndez-Lucas
- Abstract
Different pyruvate kinase isoforms are expressed in a tissue-specific manner, with pyruvate kinase M2 (PKM2) suggested to be the predominant isoform in proliferating cells and cancer cells. Because of differential regulation of enzymatic activities, PKM2, but not PKM1, has been thought to favor cell proliferation. However, the role of PKM2 in tumorigenesis has been recently challenged. Here we report that increased glucose catabolism through glycolysis and increased pyruvate kinase activity in c-MYC-driven liver tumors are associated with increased expression of both PKM1 and PKM2 isoforms and decreased expression of the liver-specific isoform of pyruvate kinase, PKL. Depletion of PKM2 at the time of c-MYC overexpression in murine livers did not affect c-MYC–induced tumorigenesis and resulted in liver tumor formation with decreased pyruvate kinase activity and decreased catabolism of glucose into alanine and the Krebs cycle. An increased PKM1/PKM2 ratio by ectopic PKM1 expression further decreased glucose flux into serine biosynthesis and increased flux into lactate and the Krebs cycle, resulting in reduced total levels of serine. However, these changes also did not affect c-MYC–induced liver tumor development. These results suggest that increased expression of PKM2 is not required to support c-MYC–induced tumorigenesis in the liver and that various PKM1/PKM2 ratios and pyruvate kinase activities can sustain glucose catabolism required for this process. Cancer Res; 77(16); 4355–64. ©2017 AACR.
- Published
- 2023
11. Inhibition of Stearoyl-CoA Desaturase Has Anti-Leukemic Properties in Acute Myeloid Leukemia
- Author
-
Vilma Dembitz, Hannah Lawson, Celine Philippe, Richard J. Burt, Sophie James, Aoife S.M. Magee, Keith Woodley, Jozef Durko, Joana Campos, Michael James Austin, Ana Rio-Machin, Pedro Casado Izquierdo, Findlay Bewicke-Copley, Giovanny Rodriguez Blanco, Bela Patel, Lori Hazlehurst, Barrie Peck, Andrew Finch, Pedro Cutillas, Jude Fitzgibbon, Mariia Yuneva, Kevin Rouault-Pierre, John Copland, Kamil Kranc, and Paolo Gallipoli
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
12. Cold exposure as anti-cancer therapy
- Author
-
Mariia Yuneva
- Subjects
Cancer Research ,Oncology - Published
- 2022
13. Discovery of multi-state gene cluster switches determining the adaptive mitochondrial and metabolic landscape of breast cancer
- Author
-
Michela Menegollo, Robert B. Bentham, Tiago Henriques, Seow Qi Ng, Ziyu Ren, Clarinde Esculier, Sia Agarwal, Emily Tong, Clement Lo, Sanjana Ilangovan, Zorka Szabadkai, Matteo Suman, Neill Patani, Avinash Ghanate, Kevin Bryson, Robert C. Stein, Mariia Yuneva, and Gyorgy Szabadkai
- Abstract
SummaryAdaptive metabolic switches are proposed to underlie conversions between cellular states during normal development as well as in cancer evolution, where they represent important therapeutic targets. However, the full spectrum, characteristics and regulation of existing metabolic switches are unknown. We propose that metabolic switches can be recognised by locating large alternating gene expression patterns and associate them with specific metabolic states. We developed a method to identify interspersed genesets by massive correlated biclustering (MCbiclust) and to predict their metabolic wiring. Testing the method on major breast cancer transcriptome datasets we discovered a series of gene sets with switch-like behaviour, predicting mitochondrial content, activity and central carbon fluxes in tumours associated with different switch positions. The predictions were experimentally validated by bioenergetic profiling and metabolic flux analysis of13C-labelled substrates and were ultimately extended by geneset analysis to link metabolic alterations to cellular states, thus predicting tumour pathology, prognosis and chemosensitivity. The method is applicable to any large and heterogeneous transcriptome dataset to discover metabolic and associated pathophysiological states.
- Published
- 2023
14. ALDH1L2 regulation of formate, formyl-methionine, and ROS controls cancer cell migration and metastasis
- Author
-
Marc Hennequart, Steven E. Pilley, Christiaan F. Labuschagne, Jack Coomes, Loic Mervant, Paul C. Driscoll, Nathalie M. Legrave, Younghwan Lee, Peter Kreuzaler, Benedict Macintyre, Yulia Panina, Julianna Blagih, David Stevenson, Douglas Strathdee, Deborah Schneider-Luftman, Eva Grönroos, Eric C. Cheung, Mariia Yuneva, Charles Swanton, and Karen H. Vousden
- Subjects
Model organisms ,Chemical Biology & High Throughput ,Human Biology & Physiology ,FOS: Clinical medicine ,Genome Integrity & Repair ,Immunology ,Infectious Disease ,Cell Biology ,Tumour Biology ,Biochemistry & Proteomics ,General Biochemistry, Genetics and Molecular Biology ,Signalling & Oncogenes ,Metabolism ,Ecology,Evolution & Ethology ,Genetics & Genomics ,Developmental Biology ,Computational & Systems Biology - Abstract
Mitochondrial 10-formyltetrahydrofolate (10-formyl-THF) is utilized by three mitochondrial enzymes to produce formate for nucleotide synthesis, NADPH for antioxidant defense, and formyl-methionine (fMet) to initiate mitochondrial mRNA translation. One of these enzymes-aldehyde dehydrogenase 1 family member 2 (ALDH1L2)-produces NADPH by catabolizing 10-formyl-THF into CO2 and THF. Using breast cancer cell lines, we show that reduction of ALDH1L2 expression increases ROS levels and the production of both formate and fMet. Both depletion of ALDH1L2 and direct exposure to formate result in enhanced cancer cell migration that is dependent on the expression of the formyl-peptide receptor (FPR). In various tumor models, increased ALDH1L2 expression lowers formate and fMet accumulation and limits metastatic capacity, while human breast cancer samples show a consistent reduction of ALDH1L2 expression in metastases. Together, our data suggest that loss of ALDH1L2 can support metastatic progression by promoting formate and fMet production, resulting in enhanced FPR-dependent signaling.
- Published
- 2023
15. Dinner is served, sir: Fighting cancer with the right diet
- Author
-
Andrés Méndez-Lucas and Mariia Yuneva
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2021
16. Adapt and conquer: Metabolic flexibility in cancer growth, invasion and evasion
- Author
-
Joanna Segal, Mariia Yuneva, Peter A. Kreuzaler, and Yulia Panina
- Subjects
0301 basic medicine ,lcsh:Internal medicine ,Citric Acid Cycle ,030209 endocrinology & metabolism ,Biology ,Article ,Pentose Phosphate Pathway ,03 medical and health sciences ,Metabolic flexibility ,Central carbon metabolism ,0302 clinical medicine ,Tumour metabolism ,Neoplasms ,medicine ,Humans ,Neoplasm Invasiveness ,lcsh:RC31-1245 ,Molecular Biology ,Cell Proliferation ,Flexibility (engineering) ,Glutaminolysis ,Cancer ,Cell Biology ,medicine.disease ,Evasion (ethics) ,Tumourigenesis ,3. Good health ,Metabolic pathway ,Glucose ,030104 developmental biology ,Tumor progression ,Anaerobic glycolysis ,Mutation ,Tumor Escape ,Energy Metabolism ,Glycolysis ,Neuroscience ,Flux (metabolism) ,Metabolic Networks and Pathways - Abstract
Background: It has been known for close to a century that, on average, tumors have a metabolism that is different from those found in healthy tissues. Typically, tumors show a biosynthetic metabolism that distinguishes itself by engaging in large scale aerobic glycolysis, heightened flux through the pentose phosphate pathway, and increased glutaminolysis among other means. However, it is becoming equally clear that non tumorous tissues at times can engage in similar metabolism, while tumors show a high degree of metabolic flexibility reacting to cues, and stresses in their local environment. Scope of the review: In this review, we want to scrutinize historic and recent research on metabolism, comparing and contrasting oncogenic and physiological metabolic states. This will allow us to better define states of bona fide tumor metabolism. We will further contextualize the stress response and the metabolic evolutionary trajectory seen in tumors, and how these contribute to tumor progression. Lastly, we will analyze the implications of these characteristics with respect to therapy response. Major conclusions: In our review, we argue that there is not one single oncogenic state, but rather a diverse set of oncogenic states. These are grounded on a physiological proliferative/wound healing program but distinguish themselves due to their large scale of proliferation, mutations, and transcriptional changes in key metabolic pathways, and the adaptations to widespread stress signals within tumors. We find evidence for the necessity of metabolic flexibility and stress responses in tumor progression and how these responses in turn shape oncogenic progression. Lastly, we find evidence for the notion that the metabolic adaptability of tumors frequently frustrates therapeutic interventions. Keywords: Tumourigenesis, Tumour metabolism, Metabolic flexibility, Central carbon metabolism
- Published
- 2020
17. Defining a metabolic landscape of tumours: genome meets metabolism
- Author
-
Sharavan Vishaan Venkateswaran, Chandan Seth Nanda, Neill Patani, and Mariia Yuneva
- Subjects
Cancer Research ,Carcinogenesis ,Inflammation ,Computational biology ,Biology ,medicine.disease_cause ,Genome ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,medicine ,Humans ,Gene ,030304 developmental biology ,Epigenomics ,0303 health sciences ,Genome, Human ,Genetic heterogeneity ,3. Good health ,Crosstalk (biology) ,Oncology ,030220 oncology & carcinogenesis ,medicine.symptom ,Signal Transduction - Abstract
Cancer is a complex disease of multiple alterations occuring at the epigenomic, genomic, transcriptomic, proteomic and/or metabolic levels. The contribution of genetic mutations in cancer initiation, progression and evolution is well understood. However, although metabolic changes in cancer have long been acknowledged and considered a plausible therapeutic target, the crosstalk between genetic and metabolic alterations throughout cancer types is not clearly defined. In this review, we summarise the present understanding of the interactions between genetic drivers of cellular transformation and cancer-associated metabolic changes, and how these interactions contribute to metabolic heterogeneity of tumours. We discuss the essential question of whether changes in metabolism are a cause or a consequence in the formation of cancer. We highlight two modes of how metabolism contributes to tumour formation. One is when metabolic reprogramming occurs downstream of oncogenic mutations in signalling pathways and supports tumorigenesis. The other is where metabolic reprogramming initiates transformation being either downstream of mutations in oncometabolite genes or induced by chronic wounding, inflammation, oxygen stress or metabolic diseases. Finally, we focus on the factors that can contribute to metabolic heterogeneity in tumours, including genetic heterogeneity, immunomodulatory factors and tissue architecture. We believe that an in-depth understanding of cancer metabolic reprogramming, and the role of metabolic dysregulation in tumour initiation and progression, can help identify cellular vulnerabilities that can be exploited for therapeutic use.
- Published
- 2019
18. PPAR-delta acts as a metabolic master checkpoint for metastasis in pancreatic cancer
- Author
-
Quan Zheng, Shanthini M. Crusz, Jiajia Tang, Angel Lanas, Sarah Courtois, Meng-Lay Lin, Andres Cano-Galiano, Sara Compte-Sancerni, Christopher Heeschen, Laure Penin-Peyta, Zhenyang Guo, Patricia Sancho, Mariia Yuneva, Beatriz Parejo-Alonso, Bruno Sainz, Pilar Espiau-Romera, Laura Ruiz-Canas, Ulf Schmitz, Petra Jagust, Sergio Lopez-Escalona, David Barneda, Minchun Chen, Sara Trabulo, and Pilar Irún
- Subjects
Paracrine signalling ,Tumor progression ,Pancreatic cancer ,medicine ,Regulator ,Cancer research ,Peroxisome proliferator-activated receptor delta ,Glycolysis ,Biology ,medicine.disease ,Phenotype ,Metastasis - Abstract
SummaryIn pancreatic cancer, emerging evidence suggests that PPAR-δ overexpression is associated with tumor progression and metastasis, but a mechanistic link is still missing. Here we now show that PPAR-δ acts as the integrating upstream regulator for the metabolic rewiring, which is preceding the subsequent initiation of an invasive/metastatic program. Specifically, paracrine and metabolic cues regularly found in the metastasis-promoting tumor stroma consistently enhance, via induction of PPAR-δ activity, the glycolytic capacity and reserve of pancreatic cancer cells, respectively, accompanied by decreased mitochondrial oxygen consumption. Consequently, genetic or pharmacological inhibition of PPAR-δ results in reduced invasiveness and metastasis. Mechanistically, PPAR-δ acts by shifting the MYC/PGC1A balance towards MYC, enhancing metabolic plasticity. Targeting MYC similarly prevents the metabolic switch and subsequent initiation of invasiveness. Therefore, our data demonstrate that PPAR-δ is a key initiator for the metabolic reprogramming in pancreatic cancer, thereby acting as a checkpoint for the phenotypic change towards invasiveness. These findings provide compelling evidence for a novel treatment strategy to combat pancreatic cancer progression.
- Published
- 2021
19. Cysteine and Folate metabolism are targetable vulnerabilities of metastatic colorectal cancer
- Author
-
Míriam Tarrado-Castellarnau, Josep Tarragó-Celada, Jordi Perarnau, Pedro de Atauri, Fionnuala Morrish, Roger R. Gomis, Mariia Yuneva, Xavier Hernández-Alias, Silvia Marin, Marta Cascante, Carles Foguet, David M. Hockenbery, and Eytan Ruppin
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,Folate Metabolism ,genome-scale metabolic models ,colorectal cancer ,lcsh:RC254-282 ,Article ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Metàstasi ,Càncer colorectal ,medicine ,metastasis ,Gene ,Redox homeostasis ,business.industry ,redox metabolism ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,digestive system diseases ,Metabolisme ,030104 developmental biology ,Metabolism ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Colon adenocarcinoma ,business ,Cysteine - Abstract
Simple Summary In this work, we studied the metabolic reprogramming of same-patient-derived cell lines with increasing metastatic potential to develop new therapeutic approaches against metastatic colorectal cancer. Using a novel systems biology approach to integrate multiple layers of omics data, we predicted and validated that cystine uptake and folate metabolism, two key pathways related to redox metabolism, are potential targets against metastatic colorectal cancer. Our findings indicate that metastatic cell lines are selectively dependent on redox homeostasis, paving the way for new targeted therapies. Abstract With most cancer-related deaths resulting from metastasis, the development of new therapeutic approaches against metastatic colorectal cancer (mCRC) is essential to increasing patient survival. The metabolic adaptations that support mCRC remain undefined and their elucidation is crucial to identify potential therapeutic targets. Here, we employed a strategy for the rational identification of targetable metabolic vulnerabilities. This strategy involved first a thorough metabolic characterisation of same-patient-derived cell lines from primary colon adenocarcinoma (SW480), its lymph node metastasis (SW620) and a liver metastatic derivative (SW620-LiM2), and second, using a novel multi-omics integration workflow, identification of metabolic vulnerabilities specific to the metastatic cell lines. We discovered that the metastatic cell lines are selectively vulnerable to the inhibition of cystine import and folate metabolism, two key pathways in redox homeostasis. Specifically, we identified the system xCT and MTHFD1 genes as potential therapeutic targets, both individually and combined, for combating mCRC.
- Published
- 2021
20. Training a neural network to learn other dimensionality reduction removes data size restrictions in bioinformatics and provides a new route to exploring data representations
- Author
-
Ian S. Gilmore, Piotr Grabowski, Mariia Yuneva, Arafath Kaja Najumudeen, Bin Yan, Panina Y, Teresa Murta, Andrew D. Campbell, Chelsea J. Nikula, Simon T. Barry, Alex Dexter, Owen J. Sansom, Kenneth N. Robinson, Richard J. A. Goodwin, Josephine Bunch, Spencer A. Thomas, John G. Swales, Adam J. Taylor, Zoltan Takats, Efstathios A Elia, Gregory Hamm, and Rory T. Steven
- Subjects
Artificial neural network ,Computer science ,business.industry ,Dimensionality reduction ,Deep learning ,Feature extraction ,Hyperspectral imaging ,Sample (statistics) ,Machine learning ,computer.software_genre ,Embedding ,Artificial intelligence ,business ,computer ,Sentence - Abstract
High dimensionality omics and hyperspectral imaging datasets present difficult challenges for feature extraction and data mining due to huge numbers of features that cannot be simultaneously examined. The sample numbers and variables of these methods are constantly growing as new technologies are developed, and computational analysis needs to evolve to keep up with growing demand. Current state of the art algorithms can handle some routine datasets but struggle when datasets grow above a certain size. We present a training deep learning via neural networks on non-linear dimensionality reduction, in particular t-distributed stochastic neighbour embedding (t-SNE), to overcome prior limitations of these methods.One Sentence SummaryAnalysis of prohibitively large datasets by combining deep learning via neural networks with non-linear dimensionality reduction.
- Published
- 2020
21. Hyperpolarised 13C MRI: a new horizon for non-invasive diagnosis of aggressive breast cancer
- Author
-
Mariia Yuneva, Teresita Beeston, Ramla Awais, David G. Gadian, Robert Stein, Matthias Glaser, Erik Årstad, Arash Latifoltojar, Hassan Jeraj, Gyorgi Szabadkai, Fiona Gong, Rafat Chowdhury, Frazer Twyman, Neill Patani, Arnaud Comment, Lorna Smith, Christopher Holt, Oshaani Abeyakoon, Shonit Punwani, Joey Clemente, James M. O'Callaghan, Steve Halligan, David Atkinson, and Marianthi-Vasiliki Papoutsaki
- Subjects
Oncology ,Model organisms ,medicine.medical_specialty ,Human Biology & Physiology ,medicine.diagnostic_test ,business.industry ,Non invasive ,Genome Integrity & Repair ,Case Report ,General Medicine ,Cancer detection ,medicine.disease ,Breast cancer screening ,Signalling & Oncogenes ,Breast cancer ,Metabolism ,Internal medicine ,medicine ,Breast MRI ,Over treatment ,Imaging technique ,business ,Public awareness ,Computational & Systems Biology - Abstract
Hyperpolarised 13C MRI (HP-MRI) is a novel imaging technique that allows real-time analysis of metabolic pathways in vivo. 1 The technology to conduct HP-MRI in humans has recently become available and is starting to be clinically applied. As knowledge of molecular biology advances, it is increasingly apparent that cancer cell metabolism is related to disease outcomes, with lactate attracting specific attention. 2 Recent reviews of breast cancer screening programs have raised concerns and increased public awareness of over treatment. The scientific community needs to shift focus from improving cancer detection alone to pursuing novel methods of distinguishing aggressive breast cancers from those which will remain indolent. HP-MRI offers the opportunity to identify aggressive tumour phenotypes and help monitor/predict therapeutic response. Here we report one of the first cases of breast cancer imaged using HP-MRI alongside correlative conventional imaging, including breast MRI.
- Published
- 2020
- Full Text
- View/download PDF
22. Glucose Catabolism in Liver Tumors Induced by c-MYC Can Be Sustained by Various PKM1/PKM2 Ratios and Pyruvate Kinase Activities
- Author
-
Mariia Yuneva, Andrés Méndez-Lucas, Diego F. Calvisi, Xin Chen, Darjus F. Tschaharganeh, Junjie Hu, Paul C. Driscoll, Jiaoyuan Jia, Chencheng Xie, Jingxiao Wang, Xinhua Song, Xiaolei Li, and Li Che
- Subjects
0301 basic medicine ,Pyruvate decarboxylation ,Cancer Research ,Pyruvate dehydrogenase kinase ,Oncology and Carcinogenesis ,Pyruvate Kinase ,Genes, myc ,Pyruvate dehydrogenase phosphatase ,Biology ,PKM2 ,Article ,Proto-Oncogene Proteins c-myc ,Experimental ,Mice ,03 medical and health sciences ,Liver Neoplasms, Experimental ,0302 clinical medicine ,2.1 Biological and endogenous factors ,Animals ,Glycolysis ,Oncology & Carcinogenesis ,Aetiology ,Phosphorylation ,Cancer ,Liver Disease ,Liver Neoplasms ,myc ,Pyruvate carboxylase ,Isoenzymes ,Glucose ,030104 developmental biology ,Genes ,Oncology ,Gluconeogenesis ,Biochemistry ,030220 oncology & carcinogenesis ,Digestive Diseases ,Pyruvate kinase - Abstract
Different pyruvate kinase isoforms are expressed in a tissue-specific manner, with pyruvate kinase M2 (PKM2) suggested to be the predominant isoform in proliferating cells and cancer cells. Because of differential regulation of enzymatic activities, PKM2, but not PKM1, has been thought to favor cell proliferation. However, the role of PKM2 in tumorigenesis has been recently challenged. Here we report that increased glucose catabolism through glycolysis and increased pyruvate kinase activity in c-MYC-driven liver tumors are associated with increased expression of both PKM1 and PKM2 isoforms and decreased expression of the liver-specific isoform of pyruvate kinase, PKL. Depletion of PKM2 at the time of c-MYC overexpression in murine livers did not affect c-MYC–induced tumorigenesis and resulted in liver tumor formation with decreased pyruvate kinase activity and decreased catabolism of glucose into alanine and the Krebs cycle. An increased PKM1/PKM2 ratio by ectopic PKM1 expression further decreased glucose flux into serine biosynthesis and increased flux into lactate and the Krebs cycle, resulting in reduced total levels of serine. However, these changes also did not affect c-MYC–induced liver tumor development. These results suggest that increased expression of PKM2 is not required to support c-MYC–induced tumorigenesis in the liver and that various PKM1/PKM2 ratios and pyruvate kinase activities can sustain glucose catabolism required for this process. Cancer Res; 77(16); 4355–64. ©2017 AACR.
- Published
- 2017
23. Hopefully devoted to Q: targeting glutamine addiction in cancer
- Author
-
Emma R Still and Mariia Yuneva
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,Drug discovery ,Autophagy ,Cancer ,Context (language use) ,Review ,Carbohydrate metabolism ,Biology ,Bioinformatics ,medicine.disease ,3. Good health ,Glutamine ,03 medical and health sciences ,030104 developmental biology ,Cell metabolism ,Oncology ,Biochemistry ,medicine ,glutamine ,cancer ,metabolism - Abstract
Altered cell metabolism enables tumours to sustain their increased energetic and biosynthetic needs. Although tumour metabolism has long been considered a promising discipline in the development of cancer therapeutics, the majority of work has focused on changes in glucose metabolism. However, the complexity of cellular metabolism means that very rarely is an individual metabolite required for a single purpose, and thus understanding the overall metabolic requirements of tumours is vital. Over the past 30 years, increasing evidence has shown that many tumours require glutamine as well as glucose for their proliferation and survival. In this minireview, we explore the complexity of glutamine metabolism in tumour cells, discussing how the overall context of the tumour dictates the requirement for glutamine and how this can affect the design of effective therapeutic strategies.
- Published
- 2017
24. Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity
- Author
-
Sebastian Munck, Kazuaki Yoshinaga, Mariia Yuneva, Gianmarco Rinaldi, Kwok-Kin Wong, Juan Fernández-García, Brandon Faubert, Chris Verslype, Thomas G. P. Grunewald, Sweta Parik, Mustapha Najimi, Catherine M. Verfaillie, Carmen Escalona-Noguero, Leanne Hodson, Kim Vriens, Sylvia Grünewald, Stefan Christen, Charlotte Kopitz, Catriona Charlton, Andrés Méndez-Lucas, Axelle Kerstens, Naohiro Gotoh, Matteo Rossi, Johannes V. Swinnen, David Cassiman, Arndt Schmitz, Laura Beretta, Ting Chen, Ruben Boon, Etienne Sokal, Patrick Steigemann, Andrea Hägebarth, Jonas Dehairs, Roberta Schmieder, Thomas Cornfield, Dorien Broekaert, Sarah-Maria Fendt, Jo A. Van Ginderachter, Martin F. Orth, Suet Ying Kwan, Laura Romero-Pérez, Sven Christian, Ali Talebi, Rita Derua, Ralph J. DeBerardinis, and Joao A.G. Duarte
- Subjects
0301 basic medicine ,Fatty Acid Desaturases ,Male ,Palmitates ,Oleic Acids ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,sapienate ,Neoplasms ,lipid metabolism ,chemistry.chemical_classification ,Human Biology & Physiology ,Multidisciplinary ,Genome Integrity & Repair ,Fatty Acids ,fatty acid metabolism ,Biochemistry ,030220 oncology & carcinogenesis ,Female ,Metabolic Networks and Pathways ,Stearoyl-CoA Desaturase ,Model organisms ,Palmitic Acids ,fatty acid desaturation ,Article ,liver cancer ,03 medical and health sciences ,Signalling & Oncogenes ,Biosynthesis ,Cell Line, Tumor ,medicine ,FADS2 ,cancer ,Animals ,Humans ,Computational & Systems Biology ,Cell Proliferation ,Fatty acid metabolism ,Cell growth ,HEK 293 cells ,Cell Membrane ,Fatty acid ,Cancer ,medicine.disease ,SCD ,lung cancer ,030104 developmental biology ,Metabolism ,HEK293 Cells ,chemistry ,Cell culture ,Cancer cell - Abstract
Most tumours have an aberrantly activated lipid metabolism1,2 that enables them to synthesize, elongate and desaturate fatty acids to support proliferation. However, only particular subsets of cancer cells are sensitive to approaches that target fatty acid metabolism and, in particular, fatty acid desaturation3. This suggests that many cancer cells contain an unexplored plasticity in their fatty acid metabolism. Here we show that some cancer cells can exploit an alternative fatty acid desaturation pathway. We identify various cancer cell lines, mouse hepatocellular carcinomas, and primary human liver and lung carcinomas that desaturate palmitate to the unusual fatty acid sapienate to support membrane biosynthesis during proliferation. Accordingly, we found that sapienate biosynthesis enables cancer cells to bypass the known fatty acid desaturation pathway that is dependent on stearoyl-CoA desaturase. Thus, only by targeting both desaturation pathways is the in vitro and in vivo proliferation of cancer cells that synthesize sapienate impaired. Our discovery explains metabolic plasticity in fatty acid desaturation and constitutes an unexplored metabolic rewiring in cancers. In several human and mouse cancer cell lines and carcinomas, a sapienate biosynthesis pathway underpins metabolic plasticity by allowing these cells to bypass stearoyl-CoA desaturase-dependent fatty acid desaturation.
- Published
- 2019
25. Low catalytic activity is insufficient to induce disease pathology in triosephosphate isomerase deficiency
- Author
-
Joanna Segal, Lillian Garrett, Manuela Scholze-Wittler, Dirk H. Busch, Bernd Timmermann, Eckhard Wolf, Markus Ralser, Thure Adler, Nana-Maria Grüning, Martin Hrabě de Angelis, Helmut Fuchs, Jan Rozman, Thomas Klopstock, Antje Krüger, Julia Calzada-Wack, Michael Mülleder, Sabine M. Hölter, Steve Michel, Hans Lehrach, Ingo Voigt, Ludger Hartmann, Mariia Yuneva, Frauke Neff, Ildiko Racz, Lore Becker, Martin Klingenspor, Ralf Fischer, Wolfgang Wurst, Heinrich Schrewe, Valerie Gailus-Durner, Beata Lukaszewska‐McGreal, and Birgit Rathkolb
- Subjects
Male ,Hemolytic anemia ,site‐directed mutagenesis ,Mutant ,enzymology [Carbohydrate Metabolism, Inborn Errors] ,medicine.disease_cause ,Triosephosphate isomerase ,Mice ,Catalytic Domain ,Enzyme Stability ,pathology [Anemia, Hemolytic, Congenital Nonspherocytic] ,Triosephosphate isomerase deficiency ,hemolytic anemia ,Genetics (clinical) ,glycolytic enzymopathy ,triosephosphate isomerase deficiency ,0303 health sciences ,Mutation ,Behavior, Animal ,Chemistry ,genetics [Triose-Phosphate Isomerase] ,030305 genetics & heredity ,Genetic disorder ,Anemia, Hemolytic, Congenital Nonspherocytic ,3. Good health ,ddc ,enzymology [Anemia, Hemolytic, Congenital Nonspherocytic] ,Original Article ,Female ,Active Site Mutation ,Glycolytic Enzymopathy ,Hemolytic Anemia ,Protein Stability Disorder ,Site-directed Mutagenesis ,Triosephosphate Isomerase Deficiency ,Carbohydrate Metabolism, Inborn Errors ,Triose-Phosphate Isomerase ,deficiency [Triose-Phosphate Isomerase] ,genetics [Catalytic Domain] ,active site mutation ,03 medical and health sciences ,protein stability disorder ,pathology [Carbohydrate Metabolism, Inborn Errors] ,Valine ,parasitic diseases ,Genetics ,medicine ,Animals ,Humans ,ddc:610 ,030304 developmental biology ,Original Articles ,medicine.disease ,Molecular biology ,Mice, Inbred C57BL ,Disease Models, Animal ,Protein Multimerization ,Isoleucine - Abstract
Triosephosphate isomerase (TPI) deficiency is a fatal genetic disorder characterized by hemolytic anemia and neurological dysfunction. Although the enzyme defect in TPI was discovered in the 1960s, the exact etiology of the disease is still debated. Some aspects indicate the disease could be caused by insufficient enzyme activity, whereas other observations indicate it could be a protein misfolding disease with tissue‐specific differences in TPI activity. We generated a mouse model in which exchange of a conserved catalytic amino acid residue (isoleucine to valine, Ile170Val) reduces TPI specific activity without affecting the stability of the protein dimer. TPIIle170Val/Ile170Val mice exhibit an approximately 85% reduction in TPI activity consistently across all examined tissues, which is a stronger average, but more consistent, activity decline than observed in patients or symptomatic mouse models that carry structural defect mutant alleles. While monitoring protein expression levels revealed no evidence for protein instability, metabolite quantification indicated that glycolysis is affected by the active site mutation. TPIIle170Val/Ile170Val mice develop normally and show none of the disease symptoms associated with TPI deficiency. Therefore, without the stability defect that affects TPI activity in a tissue‐specific manner, a strong decline in TPI catalytic activity is not sufficient to explain the pathological onset of TPI deficiency., A new mouse model shows that reducing enzyme activity, without affecting the overall structure of triosephopshate isomerase (TPI), does not induce TPI deficiency‐like symptoms in mice.
- Published
- 2018
26. Metabolic reprogramming and Notch activity distinguish between non-small cell lung cancer subtypes
- Author
-
Thaddeus D. Allen, Nourdine Bah, Teresa W.-M. Fan, Richard M. Higashi, Andrew N. Lane, Yelena Chernyavskaya, Mariia Yuneva, Michael Bousamra, Katherine Sellers, Wei Lin, James I. MacRae, Jinlian Tan, and Andrés Méndez-Lucas
- Subjects
Cancer Research ,Lung Neoplasms ,Cell ,Adenocarcinoma of Lung ,Biology ,Article ,Transcriptome ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Carcinoma, Non-Small-Cell Lung ,Gene expression ,medicine ,Tumor Microenvironment ,Metabolomics ,Animals ,Humans ,Lung cancer ,Gene ,Receptors, Notch ,Gene signature ,medicine.disease ,Cancer metabolism ,3. Good health ,stomatognathic diseases ,medicine.anatomical_structure ,Editorial ,Mechanisms of disease ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Carcinoma, Squamous Cell ,Non-small-cell lung cancer - Abstract
Background Previous studies suggested that the metabolism is differently reprogrammed in the major subtypes of non-small cell lung cancer (NSCLC), squamous cell carcinomas (SCC) and adenocarcinomas (AdC). However, a comprehensive analysis of this differential metabolic reprogramming is lacking. Methods Publicly available gene expression data from human lung cancer samples and cell lines were analysed. Stable isotope resolved metabolomics were performed on SCC and ADC tumours in human patients and in freshly resected tumour slices. Results Analysis of multiple transcriptomics data from human samples identified a SCC-distinguishing enzyme gene signature. SCC tumours from patients infused with [U-13C]-glucose and SCC tissue slices incubated with stable isotope tracers demonstrated differential glucose and glutamine catabolism compared to AdCs or non-cancerous lung, confirming increased activity through pathways defined by the SCC metabolic gene signature. Furthermore, the upregulation of Notch target genes was a distinguishing feature of SCCs, which correlated with the metabolic signature. Notch and MYC-driven murine lung tumours recapitulated the SCC-distinguishing metabolic reprogramming. However, the differences between SCCs and AdCs disappear in established cell lines in 2D culture. Conclusions Our data emphasise the importance of studying lung cancer metabolism in vivo. They also highlight potential targets for therapeutic intervention in SCC patients including differentially expressed enzymes that catalyse reactions in glycolysis, glutamine catabolism, serine, nucleotide and glutathione biosynthesis.
- Published
- 2018
27. A novel zebrafish intestinal tumor model reveals a role for cyp7a1-dependent tumor-liver crosstalk in tumors adverse effects on host
- Author
-
Sora Enya, Koichi Kawakami, Yutaka Suzuki, Shinpei Kawaoka, Almut Schulze, and Mariia Yuneva
- Subjects
0301 basic medicine ,Male ,lcsh:Medicine ,Medicine (miscellaneous) ,medicine.disease_cause ,Epithelium ,Animals, Genetically Modified ,Immunology and Microbiology (miscellaneous) ,Gene expression ,Bile ,Cholesterol 7-alpha-Hydroxylase ,Zebrafish ,biology ,Phenotype ,Cell biology ,Intestines ,Crosstalk (biology) ,Liver ,Larva ,Female ,KRAS ,medicine.symptom ,Intestinal tumor ,lcsh:RB1-214 ,Research Article ,Hepatomegaly ,Growth defect ,Neuroscience (miscellaneous) ,Inflammation ,Cholesterol 7 alpha-hydroxylase ,General Biochemistry, Genetics and Molecular Biology ,Liver inflammation ,03 medical and health sciences ,cyp7a1 ,Intestinal Neoplasms ,lcsh:Pathology ,medicine ,Cholesterol metabolism ,Animals ,Gene ,Crosses, Genetic ,Cell Proliferation ,lcsh:R ,Metabolism ,Zebrafish Proteins ,biology.organism_classification ,Disease Models, Animal ,030104 developmental biology ,Gene Expression Regulation ,Immunology ,Cancer research - Abstract
The nature of host organs and genes that underlie tumor-induced physiological disruption on the host remains ill-defined. Here, we establish a novel zebrafish intestinal tumor model that is suitable for addressing this issue, and find that hepatic cyp7a1, the rate-limiting factor for synthesizing bile acids [or, in the case of zebrafish, bile alcohol (BA)], is such a host gene. Inducing krasG12D by Gal4 specifically expressed in the posterior intestine resulted in the formation of an intestinal tumor. The local intestinal tumor caused systemic detrimental effects on the host, including liver inflammation, hepatomegaly, growth defects and organismal death. Whole-organism-level gene expression analysis and metabolite measurements revealed that the intestinal tumor reduced total BA levels, possibly via altered expression of hepatic cyp7a1. Genetically overexpressing cyp7a1 in the liver restored BA synthesis and ameliorated tumor-induced liver inflammation, but not other tumor-dependent phenotypes. Thus, we found a previously unknown role of cyp7a1 as the host gene that links the intestinal tumor, hepatic cholesterol–BA metabolism and liver inflammation in tumor-bearing zebrafish larvae. Our model provides an important basis to discover host genes responsible for tumor-induced phenotypes and to uncover mechanisms underlying how tumors adversely affect host organisms., Summary: The authors genetically dissect the adverse effects on the host in a new model of zebrafish intestinal tumor.
- Published
- 2017
- Full Text
- View/download PDF
28. Correction: Hopefully devoted to Q: targeting glutamine addiction in cancer
- Author
-
Emma R Still and Mariia Yuneva
- Subjects
Cancer Research ,Glycosylation ,Glutamine ,media_common.quotation_subject ,MEDLINE ,Bioinformatics ,Neoplasms ,Drug Discovery ,Autophagy ,Tumor Microenvironment ,Humans ,Medicine ,License ,media_common ,business.industry ,TOR Serine-Threonine Kinases ,Addiction ,Published Erratum ,Correction ,Cancer ,medicine.disease ,Risk factors ,Oncology ,business ,Metabolic Networks and Pathways - Abstract
Altered cell metabolism enables tumours to sustain their increased energetic and biosynthetic needs. Although tumour metabolism has long been considered a promising discipline in the development of cancer therapeutics, the majority of work has focused on changes in glucose metabolism. However, the complexity of cellular metabolism means that very rarely is an individual metabolite required for a single purpose, and thus understanding the overall metabolic requirements of tumours is vital. Over the past 30 years, increasing evidence has shown that many tumours require glutamine as well as glucose for their proliferation and survival. In this minireview, we explore the complexity of glutamine metabolism in tumour cells, discussing how the overall context of the tumour dictates the requirement for glutamine and how this can affect the design of effective therapeutic strategies.
- Published
- 2019
29. Reciprocal regulation of microRNA-122 and c-Myc in hepatocellular cancer: Role of E2F1 and transcription factor dimerization partner 2
- Author
-
Ramesh K. Ganju, Xinmei Wang, Mariia Yuneva, Hemant K. Bid, Bo Wang, Jianhua Yu, Shu-Hao Hsu, Samson T. Jacob, Kalpana Ghoshal, and Huban Kutay
- Subjects
Regulation of gene expression ,Hepatology ,Coactivator ,Gene expression ,Cancer research ,biology.protein ,RNA polymerase II ,Ectopic expression ,Promoter ,Biology ,Chromatin immunoprecipitation ,Molecular biology ,Transcription factor - Abstract
c-Myc is a well-known oncogene frequently up-regulated in different malignancies, whereas liver-specific microRNA (miR)-122, a bona fide tumor suppressor, is down-regulated in hepatocellular cancer (HCC). Here we explored the underlying mechanism of reciprocal regulation of these two genes. Real-time reverse-transcription polymerase chain reaction (RT-PCR) and northern blot analysis demonstrated reduced expression of the primary, precursor, and mature miR-122 in c-MYC-induced HCCs compared to the benign livers, indicating transcriptional suppression of miR-122 upon MYC overexpression. Indeed, chromatin immunoprecipitation (ChIP) assay showed significantly reduced association of RNA polymerase II and histone H3K9Ac, markers of active chromatin, with the miR-122 promoter in tumors relative to the c-MYC-uninduced livers, indicating transcriptional repression of miR-122 in c-MYC-overexpressing tumors. The ChIP assay also demonstrated a significant increase in c-Myc association with the miR-122 promoter region that harbors a conserved noncanonical c-Myc binding site in tumors compared to the livers. Ectopic expression and knockdown studies showed that c-Myc indeed suppresses expression of primary and mature miR-122 in hepatic cells. Additionally, Hnf-3β, a liver enriched transcription factor that activates miR-122 gene, was suppressed in c-MYC-induced tumors. Notably, miR-122 also repressed c-Myc transcription by targeting transcriptional activator E2f1 and coactivator Tfdp2, as evident from ectopic expression and knockdown studies and luciferase reporter assays in mouse and human hepatic cells. Conclusion: c-Myc represses miR-122 gene expression by associating with its promoter and by down-regulating Hnf-3β expression, whereas miR-122 indirectly inhibits c-Myc transcription by targeting Tfdp2 and E2f1. In essence, these results suggest a double-negative feedback loop between a tumor suppressor (miR-122) and an oncogene (c-Myc). (Hepatology 2014;59:555–566)
- Published
- 2013
30. Finding an 'Achilles’ heel' of cancer: The role of glucose and glutamine metabolism in the survival of transformed cells
- Author
-
Mariia Yuneva
- Subjects
Cell Survival ,Glutamine ,Cell ,Cell Biology ,Biology ,medicine.disease_cause ,Mice ,Metabolic pathway ,Cell Transformation, Neoplastic ,Glucose ,Cell metabolism ,medicine.anatomical_structure ,Biochemistry ,In vivo ,Apoptosis ,Neoplasms ,medicine ,Animals ,Humans ,Viability assay ,Carcinogenesis ,Molecular Biology ,Developmental Biology - Abstract
Multiple lines of evidence indicate that the process of tumorigenesis is often associated with altered metabolism of two major nutrients, glucose and glutamine. These two nutrients are engaged in multiple metabolic pathways that can be required for cell viability. The roles of glucose and glutamine in the survival of transformed cells both in vitro and in vivo have been separately evaluated in various cell systems, and glucose as the major cellular energy source has received most of the attention. At the same time, data suggests that the inclusion of glucose and glutamine into specific metabolic pathways and cellular sensitivity to the availability of either of these nutrients depends on the cell origin and the combination and nature of transforming events. Exploiting cell metabolism to develop selective cancer therapeutics requires consideration of these factors and evaluation of the requirement of glucose and glutamine metabolism for survival of different transformed cells. Here we discuss possible molecular mechanisms underlying oncogene-induced sensitivity to deprivation of these nutrients.
- Published
- 2008
31. Deficiency in glutamine but not glucose induces MYC-dependent apoptosis in human cells
- Author
-
Mariia Yuneva, Ravi Sachidanandam, Yuri Lazebnik, Nicola Zamboni, and Peter J. Oefner
- Subjects
Male ,Glutamine ,Cell Culture Techniques ,Apoptosis ,Carbohydrate metabolism ,Biology ,Proto-Oncogene Mas ,Article ,Cell Line ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,0302 clinical medicine ,Transduction, Genetic ,medicine ,Humans ,Viability assay ,Lung ,Research Articles ,Cells, Cultured ,030304 developmental biology ,Skin ,0303 health sciences ,Cancer ,Cell Biology ,Fibroblasts ,medicine.disease ,3. Good health ,Cell biology ,Citric acid cycle ,Glucose ,Retroviridae ,Gene Expression Regulation ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell - Abstract
The idea that conversion of glucose to ATP is an attractive target for cancer therapy has been supported in part by the observation that glucose deprivation induces apoptosis in rodent cells transduced with the proto-oncogene MYC, but not in the parental line. Here, we found that depletion of glucose killed normal human cells irrespective of induced MYC activity and by a mechanism different from apoptosis. However, depletion of glutamine, another major nutrient consumed by cancer cells, induced apoptosis depending on MYC activity. This apoptosis was preceded by depletion of the Krebs cycle intermediates, was prevented by two Krebs cycle substrates, but was unrelated to ATP synthesis or several other reported consequences of glutamine starvation. Our results suggest that the fate of normal human cells should be considered in evaluating nutrient deprivation as a strategy for cancer therapy, and that understanding how glutamine metabolism is linked to cell viability might provide new approaches for treatment of cancer.
- Published
- 2007
32. Deregulation of the energy metabolism in the hepatocellular cancer developed in miR-122 knockout mice
- Author
-
Chowdhary, Vivek, Mal, Tapas, Hsu, Shu-Hao, Kun-Yu Teng, Wang, Bo, Huban Kutay, Mariia Yuneva, Fan, Teresa, and Ghoshal, Kalpana
- Published
- 2014
- Full Text
- View/download PDF
33. Pyruvate carboxylase is critical for non-small-cell lung cancer proliferation
- Author
-
Yali Wang, Mariia Yuneva, Rahul Deshpande, Richard M. Higashi, Teresa W.-M. Fan, Andrew N. Lane, Stephen P. Slone, Donald M. Miller, Katherine Sellers, Michael Bousamra, Jun Yan, and Matthew P. Fox
- Subjects
Male ,Stromal cell ,Lung Neoplasms ,Citric Acid Cycle ,Biology ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,medicine ,Animals ,Humans ,Radioactive Tracers ,Lung cancer ,030304 developmental biology ,Cell Proliferation ,Pyruvate Carboxylase ,0303 health sciences ,Cell growth ,Glutaminase ,Nucleotides ,General Medicine ,medicine.disease ,Lipid Metabolism ,Glutathione ,3. Good health ,Pyruvate carboxylase ,Neoplasm Proteins ,Citric acid cycle ,Gene Expression Regulation, Neoplastic ,Glucose ,HEK293 Cells ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Commentary ,Female ,Growth inhibition - Abstract
Anabolic biosynthesis requires precursors supplied by the Krebs cycle, which in turn requires anaplerosis to replenish precursor intermediates. The major anaplerotic sources are pyruvate and glutamine, which require the activity of pyruvate carboxylase (PC) and glutaminase 1 (GLS1), respectively. Due to their rapid proliferation, cancer cells have increased anabolic and energy demands; however, different cancer cell types exhibit differential requirements for PC- and GLS-mediated pathways for anaplerosis and cell proliferation. Here, we infused patients with early-stage non-small-cell lung cancer (NSCLC) with uniformly 13C-labeled glucose before tissue resection and determined that the cancerous tissues in these patients had enhanced PC activity. Freshly resected paired lung tissue slices cultured in 13C6-glucose or 13C5,15N2-glutamine tracers confirmed selective activation of PC over GLS in NSCLC. Compared with noncancerous tissues, PC expression was greatly enhanced in cancerous tissues, whereas GLS1 expression showed no trend. Moreover, immunohistochemical analysis of paired lung tissues showed PC overexpression in cancer cells rather than in stromal cells of tumor tissues. PC knockdown induced multinucleation, decreased cell proliferation and colony formation in human NSCLC cells, and reduced tumor growth in a mouse xenograft model. Growth inhibition was accompanied by perturbed Krebs cycle activity, inhibition of lipid and nucleotide biosynthesis, and altered glutathione homeostasis. These findings indicate that PC-mediated anaplerosis in early-stage NSCLC is required for tumor survival and proliferation.
- Published
- 2013
34. The Metabolic Profile of Tumors Depends on both the Responsible Genetic Lesion and Tissue Type
- Author
-
Francisco J. Alonso, Chunmei Wang, J. Michael Bishop, José M. Matés, Thaddeus D. Allen, Xin Chen, Richard M. Higashi, Mariia Yuneva, Youngho Seo, Dana Ferraris, Teresa W.-M. Fan, and Takashi Tsukamoto
- Subjects
Lung Neoplasms ,Physiology ,Glutamine ,Blotting, Western ,Citric Acid Cycle ,Immunoblotting ,Biology ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Liver Neoplasms, Experimental ,Glutamine synthetase ,Cell Line, Tumor ,Glucokinase ,Animals ,Humans ,Lactic Acid ,Molecular Biology ,Nuclear Magnetic Resonance, Biomolecular ,030304 developmental biology ,DNA Primers ,0303 health sciences ,Catabolism ,Glutaminase ,Cell Biology ,Metabolism ,Proto-Oncogene Proteins c-met ,Immunohistochemistry ,3. Good health ,Citric acid cycle ,Cell Transformation, Neoplastic ,Glucose ,030220 oncology & carcinogenesis ,Isotope Labeling ,Cancer research ,Metabolome ,RNA Interference - Abstract
SummaryThe altered metabolism of tumors has been considered a target for anticancer therapy. However, the relationship between distinct tumor-initiating lesions and anomalies of tumor metabolism in vivo has not been addressed. We report that MYC-induced mouse liver tumors significantly increase both glucose and glutamine catabolism, whereas MET-induced liver tumors use glucose to produce glutamine. Increased glutamine catabolism in MYC-induced liver tumors is associated with decreased levels of glutamine synthetase (Glul) and the switch from Gls2 to Gls1 glutaminase. In contrast to liver tumors, MYC-induced lung tumors display increased expression of both Glul and Gls1 and accumulate glutamine. We also show that inhibition of Gls1 kills cells that overexpress MYC and catabolize glutamine. Our results suggest that the metabolic profiles of tumors are likely to depend on both the genotype and tissue of origin and have implications regarding the design of therapies targeting tumor metabolism.
- Published
- 2012
35. Therapeutic potential of a synthetic lethal interaction between the MYC proto-oncogene and inhibition of aurora-B kinase
- Author
-
Suwon Kim, J. Michael Bishop, Mariia Yuneva, Andrei Goga, Hong Liu, and Dun Yang
- Subjects
DNA Replication ,Lymphoma ,Aurora inhibitor ,Aurora B kinase ,Apoptosis ,Biology ,Protein Serine-Threonine Kinases ,Proto-Oncogene Mas ,Piperazines ,Proto-Oncogene Proteins c-myc ,Mice ,Aurora kinase ,Aurora Kinases ,Autophagy ,Animals ,Aurora Kinase B ,Humans ,Mitosis ,Cytokinesis ,Multidisciplinary ,Oncogene ,Kinase ,Biological Sciences ,Rats ,Disease Models, Animal ,Microscopy, Electron ,Cancer research ,Neoplasm Transplantation - Abstract
The Myc protein and proteins that participate in mitosis represent attractive targets for cancer therapy. However, their potential is presently compromised by the threat of side effects and by a lack of pharmacological inhibitors of Myc. Here we report that a circumscribed exposure to the aurora kinase inhibitor, VX-680, selectively kills cells that overexpress Myc. This synthetic lethal interaction is attributable to inhibition of aurora-B kinase, with consequent disabling of the chromosomal passenger protein complex (CPPC) and ensuing DNA replication in the absence of cell division; executed by sequential apoptosis and autophagy; not reliant on the tumor suppressor protein p53; and effective against mouse models for B-cell and T-cell lymphomas initiated by transgenes of MYC . Our findings cast light on how inhibitors of aurora-B kinase may kill tumor cells, implicate Myc in the induction of a lethal form of autophagy, indicate that expression of Myc be a useful biomarker for sensitivity of tumor cells to inhibition of the CPPC, dramatize the virtue of bimodal killing by a single therapeutic agent, and suggest a therapeutic strategy for killing tumor cells that overexpress Myc while sparing normal cells.
- Published
- 2010
36. Abstract 1424: Enhanced pyruvate carboxylation is crucial to non-small cell lung cancer proliferation and anabolism
- Author
-
Teresa W.-M. Fan, Jun Yan, Richard M. Higashi, Andrew N. Lane, Michael Bousamra, Matthew P. Fox, Mariia Yuneva, and Katherine E. Sellers
- Subjects
Citric acid cycle ,A549 cell ,Glutamine ,Cancer Research ,Glutaminolysis ,Oncology ,Anabolism ,Biochemistry ,Glutaminase ,Cancer cell ,Cancer research ,Biology ,Pyruvate carboxylase - Abstract
Proliferating cancer cells require an active Krebs cycle for generating anabolic precursors, in addition to energy production. Diversion of the Krebs cycle intermediates to meet anabolic demands cannot be sustained without anaplerosis. The two major anaplerotic pathways are the deamidation of glutamine to form glutamate catalyzed by glutaminase (GLS) or glutaminolysis, and the carboxylation of pyruvate to oxaloacetate by pyruvate carboxylase (PC). We determined the expression of PC and GLS proteins in fresh tumor tissue and paired adjacent benign tissue from 86 human NSCLC patients. PC was elevated (median 8-10 fold) in 94% of the tumor tissues, whereas GLS expression did not differ significantly between tumor and paired non tumorous lung tissue. Using 13C6 glucose as tracer and stable isotope-resolved metabolomics (SIRM), we also observed elevated PC activity in vivo in human patients with early stage NSCLC. To examine the importance of PC in the growth and survival of NSCLC, 5 NSCLC cell lines were transduced with a lentiviral vector containing an shRNA against PC. PC knockdown slowed proliferation, induced multinucleation, and reduced colony formation in soft agar. To validate attenuated PC activity and to profile the metabolic effect of PC knockdown, A549 cells were grown in 13C6 glucose or 13C5 glutamine and the incorporation of 13C into various metabolic pathways was measured by NMR and MS by SIRM. We found reduced entry of both glucose and glutamine carbon into the Krebs cycle metabolites, fatty acyl chains of lipids, and nucleotides, suggesting that both energy production and anabolic pathways were hindered and blocking the PC pathway was not compensated by GLS activity. In addition, glutathione biosynthesis and homeostasis were perturbed by PC suppression, leading to compromised anti-oxidation capacity. We further found that PC knockdown in A549 cells reduced their growth rate in mouse xenografts, and recapitulate the metabolic perturbations seen both in cell culture and in human patients. Together, these results suggest that PC is indispensible for the growth and anabolism of NSCLC. This work was funded by 5P20RR018733, 1R01CA118434-01A2, 1P01CA163223-01A1, 1R01ES022191-01, and 3R01CA118434-02S1; and the KLCRP, CTSPGP, and the KY Challenge for Excellence. Citation Format: Katherine E. Sellers, Matthew P. Fox, Michael Bousamra, Jun Yan, Mariia Yuneva, Richard M. Higashi, Andrew N. Lane, Teresa WM Fan. Enhanced pyruvate carboxylation is crucial to non-small cell lung cancer proliferation and anabolism. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1424. doi:10.1158/1538-7445.AM2014-1424
- Published
- 2014
37. Glutamine Sensitivity Analysis Identifies the xCT Antiporter as a Common Triple-Negative Breast Tumor Therapeutic Target
- Author
-
Joe W. Gray, Raymond J. Louie, Thomas Holton, Laura J. van't Veer, Denise A. Chan, Frank McCormick, Luika A. Timmerman, Mariia Yuneva, Kornelia Polyak, Stephen P. Ethier, Mercè Padró, Min Hu, and Anneleen Daemen
- Subjects
Cancer Research ,Amino Acid Transport System y+ ,Glutamine ,Antiporter ,Triple Negative Breast Neoplasms ,SLC7A11 ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,In vivo ,Sulfasalazine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,RNA, Small Interfering ,030304 developmental biology ,0303 health sciences ,biology ,Gene Expression Profiling ,Cancer ,Cell Biology ,Prognosis ,medicine.disease ,Glutathione ,3. Good health ,Gene Expression Regulation, Neoplastic ,Oncology ,Drug development ,030220 oncology & carcinogenesis ,Immunology ,Disease Progression ,biology.protein ,Cancer research ,Female ,Reactive Oxygen Species ,medicine.drug - Abstract
SummaryA handful of tumor-derived cell lines form the mainstay of cancer therapeutic development, yielding drugs with an impact typically measured as months to disease progression. To develop more effective breast cancer therapeutics and more readily understand their clinical impact, we constructed a functional metabolic portrait of 46 independently derived breast cell lines. Our analysis of glutamine uptake and dependence identified a subset of triple-negative samples that are glutamine auxotrophs. Ambient glutamine indirectly supports environmental cystine acquisition via the xCT antiporter, which is expressed on one-third of triple-negative tumors in vivo. xCT inhibition with the clinically approved anti-inflammatory sulfasalazine decreases tumor growth, revealing a therapeutic target in breast tumors of poorest prognosis and a lead compound for rapid, effective drug development.
- Full Text
- View/download PDF
38. MYC/PGC-1α Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer Stem Cells
- Author
-
Katherine Sellers, Catarina R. Viera, Christopher Heeschen, David Barneda, Mariia Yuneva, Patricia Sancho, Tony Bou Kheir, Bruno Sainz, Osvaldo Graña, Petra Jagust, Matthieu Schoenhals, Alejandra Tavera, Emma Burgos-Ramos, and Ramon Campos-Olivas
- Subjects
Mitochondrial ROS ,Physiology ,Cancer de pancreas ,Mice, Nude ,Antineoplastic Agents ,Biology ,Mitochondrion ,MYC/PGC-1a ,Oxidative Phosphorylation ,Proto-Oncogene Proteins c-myc ,Mice ,Antigens, CD ,Pancreatic cancer ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,AC133 Antigen ,Molecular Biology ,Gene Library ,Glycoproteins ,Vitamin K 3 ,Cancer ,Cell Biology ,medicine.disease ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Phenotype ,Metformin ,Mitochondria ,3. Good health ,Pancreatic Neoplasms ,Drug Resistance, Neoplasm ,Apoptosis ,Neoplastic Stem Cells ,Cancer research ,RNA Interference ,Stem cell ,Peptides ,Reactive Oxygen Species ,Transcription Factors ,medicine.drug - Abstract
SummaryThe anti-diabetic drug metformin targets pancreatic cancer stem cells (CSCs), but not their differentiated progenies (non-CSCs), which may be related to distinct metabolic phenotypes. Here we conclusively demonstrate that while non-CSCs were highly glycolytic, CSCs were dependent on oxidative metabolism (OXPHOS) with very limited metabolic plasticity. Thus, mitochondrial inhibition, e.g., by metformin, translated into energy crisis and apoptosis. However, resistant CSC clones eventually emerged during treatment with metformin due to their intermediate glycolytic/respiratory phenotype. Mechanistically, suppression of MYC and subsequent increase of PGC-1α were identified as key determinants for the OXPHOS dependency of CSCs, which was abolished in resistant CSC clones. Intriguingly, no resistance was observed for the mitochondrial ROS inducer menadione and resistance could also be prevented/reversed for metformin by genetic/pharmacological inhibition of MYC. Thus, the specific metabolic features of pancreatic CSCs are amendable to therapeutic intervention and could provide the basis for developing more effective therapies to combat this lethal cancer.
- Full Text
- View/download PDF
39. Adapt and conquer: Metabolic flexibility in cancer growth, invasion and evasion
- Author
-
Kreuzaler, Peter, Panina, Yulia, Segal, Joanna, and Mariia Yuneva
- Subjects
Model organisms ,Signalling & Oncogenes ,Human Biology & Physiology ,Metabolism ,Genome Integrity & Repair ,3. Good health ,Computational & Systems Biology - Abstract
BACKGROUND: It has been known for close to a century that, on average, tumors have a metabolism that is different from those found in healthy tissues. Typically, tumors show a biosynthetic metabolism that distinguishes itself by engaging in large scale aerobic glycolysis, heightened flux through the pentose phosphate pathway, and increased glutaminolysis among other means. However, it is becoming equally clear that non tumorous tissues at times can engage in similar metabolism, while tumors show a high degree of metabolic flexibility reacting to cues, and stresses in their local environment. SCOPE OF THE REVIEW: In this review, we want to scrutinize historic and recent research on metabolism, comparing and contrasting oncogenic and physiological metabolic states. This will allow us to better define states of bona fide tumor metabolism. We will further contextualize the stress response and the metabolic evolutionary trajectory seen in tumors, and how these contribute to tumor progression. Lastly, we will analyze the implications of these characteristics with respect to therapy response. MAJOR CONCLUSIONS: In our review, we argue that there is not one single oncogenic state, but rather a diverse set of oncogenic states. These are grounded on a physiological proliferative/wound healing program but distinguish themselves due to their large scale of proliferation, mutations, and transcriptional changes in key metabolic pathways, and the adaptations to widespread stress signals within tumors. We find evidence for the necessity of metabolic flexibility and stress responses in tumor progression and how these responses in turn shape oncogenic progression. Lastly, we find evidence for the notion that the metabolic adaptability of tumors frequently frustrates therapeutic interventions.
40. Low catalytic activity is insufficient to induce disease pathology in triosephosphate isomerase deficiency
- Author
-
Segal, Joanna, Mülleder, Michael, Krüger, Antje, Adler, Thure, Scholze-Wittler, Manuela, Becker, Lore, Calzada-Wack, Julia, Garrett, Lillian, Hölter, Sabine M, Rathkolb, Birgit, Rozman, Jan, Racz, Ildiko, Fischer, Ralf, Busch, Dirk H, Neff, Frauke, Klingenspor, Martin, Klopstock, Thomas, Nana-Maria Grüning, Michel, Steve, Lukaszewska-McGreal, Beata, Voigt, Ingo, Hartmann, Ludger, Timmermann, Bernd, Lehrach, Hans, Wolf, Eckhard, Wurst, Wolfgang, Gailus-Durner, Valérie, Fuchs, Helmut, Angelis, Martin H De, Schrewe, Heinrich, Mariia Yuneva, and Ralser, Markus
- Subjects
Model organisms ,Chemical Biology & High Throughput ,Signalling & Oncogenes ,Human Biology & Physiology ,Metabolism ,Ecology,Evolution & Ethology ,parasitic diseases ,Genome Integrity & Repair ,Synthetic Biology ,3. Good health ,Computational & Systems Biology - Abstract
Triosephosphate isomerase (TPI) deficiency is a fatal genetic disorder characterized by haemolytic anaemia and neurological dysfunction. Although the enzyme defect in TPI was discovered in the 1960s, the exact etiology of the disease is still debated. Some aspects indicate the disease could be caused by insufficient enzyme activity, whereas other observations indicate it could be a protein misfolding disease with tissue-specific differences in TPI activity. We generated a mouse model in which exchange of a conserved catalytic amino acid residue (isoleucine to valine, Ile170Val) reduces TPI specific activity without affecting the stability of the protein dimer. TPIIle170Val/Ile170Val mice exhibit an ~85% reduction in TPI activity consistently across all examined tissues, which is a stronger average activity decline than observed in patients or symptomatic mouse models that carry structural defect mutant alleles. While monitoring protein expression levels revealed no evidence for protein instability, metabolite quantification indicated that glycolysis is affected by the active site mutation. TPIIle170Val/Ile170Val mice develop normally and show none of the disease symptoms associated with TPI deficiency. Therefore, without the stability defect that penetrates in a tissue-specific manner, a strong decline in TPI catalytic activity is not sufficient to explain the pathological onset of TPI deficiency. This article is protected by copyright. All rights reserved.
41. Low catalytic activity is insufficient to induce disease pathology in triosephosphate isomerase deficiency
- Author
-
Segal, Joanna, Mülleder, Michael, Krüger, Antje, Adler, Thure, Scholze-Wittler, Manuela, Becker, Lore, Calzada-Wack, Julia, Garrett, Lillian, Hölter, Sabine M, Rathkolb, Birgit, Rozman, Jan, Racz, Ildiko, Fischer, Ralf, Busch, Dirk H, Neff, Frauke, Klingenspor, Martin, Klopstock, Thomas, Nana-Maria Grüning, Michel, Steve, Lukaszewska-McGreal, Beata, Voigt, Ingo, Hartmann, Ludger, Timmermann, Bernd, Lehrach, Hans, Wolf, Eckhard, Wurst, Wolfgang, Gailus-Durner, Valérie, Fuchs, Helmut, Angelis, Martin H De, Schrewe, Heinrich, Mariia Yuneva, and Ralser, Markus
- Subjects
Model organisms ,Chemical Biology & High Throughput ,Signalling & Oncogenes ,Human Biology & Physiology ,Metabolism ,Ecology,Evolution & Ethology ,parasitic diseases ,Genome Integrity & Repair ,Synthetic Biology ,3. Good health ,Computational & Systems Biology - Abstract
Triosephosphate isomerase (TPI) deficiency is a fatal genetic disorder characterized by haemolytic anaemia and neurological dysfunction. Although the enzyme defect in TPI was discovered in the 1960s, the exact etiology of the disease is still debated. Some aspects indicate the disease could be caused by insufficient enzyme activity, whereas other observations indicate it could be a protein misfolding disease with tissue-specific differences in TPI activity. We generated a mouse model in which exchange of a conserved catalytic amino acid residue (isoleucine to valine, Ile170Val) reduces TPI specific activity without affecting the stability of the protein dimer. TPIIle170Val/Ile170Val mice exhibit an ~85% reduction in TPI activity consistently across all examined tissues, which is a stronger average activity decline than observed in patients or symptomatic mouse models that carry structural defect mutant alleles. While monitoring protein expression levels revealed no evidence for protein instability, metabolite quantification indicated that glycolysis is affected by the active site mutation. TPIIle170Val/Ile170Val mice develop normally and show none of the disease symptoms associated with TPI deficiency. Therefore, without the stability defect that penetrates in a tissue-specific manner, a strong decline in TPI catalytic activity is not sufficient to explain the pathological onset of TPI deficiency. This article is protected by copyright. All rights reserved.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.