1
|
Odin E, Carlsson G, Saksena P, Edsjö A, Di Cara A, Tell R, Gustavsson B, Wettergren Y. Folate-Associated Gene Expression in Primary Tumors Is Associated With Tumor Response and Progression-Free Survival of Patients With Metastatic Colorectal Cancer Undergoing 5-FU/Leucovorin-Based Combination Chemotherapy. Cancer Med 2025; 14:e70895. [PMID: 40357991 PMCID: PMC12070377 DOI: 10.1002/cam4.70895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 03/21/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND 5-Fluorouracil (5-FU) and the Folate Leucovorin (LV) form the chemotherapy backbone for metastatic colorectal cancer (mCRC). Tumoral expression of specific folate-associated genes is associated with the risk of recurrence in stage III CRC following adjuvant 5-FU/LV (FLV)-based combination chemotherapy according to the Nordic bolus regimen. The aim was to evaluate whether expression of folate-associated genes in Pre-therapeutic tumor samples is associated with outcomes of patients with mCRC undergoing palliative FLV-based combination chemotherapy. PATIENTS AND METHODS Patients treated with FLV (n = 113), FLV + oxaliplatin (FLOX, n = 102), or FLV + irinotecan (FLIRI, n = 75) were included. ABCC3, RFC-1, PCFT, MFT, MTHFD2, and TYMS expression was determined by qPCR and related to tumor response and 3-year progression-free survival (PFS). Analyses were conducted on the entire cohort and on subgroups (group 1: stage I-III; group 2: stage IV, at primary surgery). Multivariate Cox proportional hazard models were applied to assess associations between covariates and PFS. RESULTS Low TYMS and high MFT expression in group 1, and high ABCC3 expression in group 2 correlated with better PFS (HR 1.37 (1.04-1.82), HR 0.49 (0.30-0.80), and HR 0.74 (0.60-0.93)), respectively. In addition, high MFT expression was associated with better PFS of patients treated with FLIRI (HR 0.47 (0.27-0.81), p = 0.007) whereas high expression of ABCC3 was associated with better PFS of patients treated with FLOX (HR 0.46 (0.23-0.92), p = 0.029). CONCLUSION While pretherapeutic tumoral expression of specific folate-associated genes may not serve as a universal predictive marker for FLV-based treatment, it might predict response and outcomes in patients receiving FLOX or FLIRI. Evaluating the impact of gene expression in primary tumors should consider subgrouping of patients by disease stage at diagnosis as well as the applied chemotherapy regimen. Prospective clinical studies on patients with mCRC are warranted to validate these findings.
Collapse
Affiliation(s)
- Elisabeth Odin
- Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Göran Carlsson
- Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of SurgerySahlgrenska University Hospital, Region Västra GötalandGothenburgSweden
| | - Pushpa Saksena
- Department of PathologySahlgrenska University HospitalGothenburgSweden
| | - Anders Edsjö
- Department of Clinical Genetics, Pathology, and Molecular DiagnosticsOffice for Medical Services, Region SkåneLundSweden
- Division of Pathology, Department of Clinical SciencesLund UniversityLundSweden
| | | | | | - Bengt Gustavsson
- Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of SurgerySahlgrenska University Hospital, Region Västra GötalandGothenburgSweden
| | - Yvonne Wettergren
- Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Department of SurgerySahlgrenska University Hospital, Region Västra GötalandGothenburgSweden
| |
Collapse
|
2
|
Jin Q, Wang F, Ye W, Wang Q, Xu S, Jiang S, Li X, Yue M, Yu D, Jin M, Fu A, Li W. Compound Bacillus improves eggshell quality and egg metabolites of hens by promoting the metabolism balance of calcium and phosphorus and uterine cell proliferation. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:355-369. [PMID: 39640545 PMCID: PMC11617893 DOI: 10.1016/j.aninu.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 12/07/2024]
Abstract
Probiotics have beneficial effects on improving egg quality, but there is little research about the effect of probiotics on metabolite composition, and the mechanisms are not yet fully understood. The aim of this study was to investigate the potential mechanisms by which compound Bacillus improves egg quality and metabolite composition. A total of 20,000 Jingfen No. 6 laying hens at 381 d old were randomly divided into two treatments: control group with a basal diet, and the basal diet with 5 × 108 CFU/kg compound Bacillus supplementation (Ba) group. The trial lasted eight weeks. The results showed that compound Bacillus improved the gloss and strength of eggshells and reduced the ratio of sand-shell eggs by 23.8%. Specifically, the effective layer of eggshell was thicker and its calcite column was closely connected. Compound Bacillus increased the contents of beneficial fatty acids in the egg yolk, and lipids and lipid-like molecules in the albumen (P < 0.01), while decreased the contents of total cholesterol, triglycerides, and benzene ring compounds in the egg yolk and organic oxygen compounds in the albumen (P < 0.01). In addition, the compound Bacillus increased the calcium absorption in the duodenum by up-regulating the expression of transporters and serum hormone synergism (P < 0.05), and promoted metabolic balance of calcium and phosphorus. Simultaneously, uterine transcriptome showed that the expression of ChaC glutathione specific gamma-glutamylcyclotransferase 1 (CHAC1), glycoprotein-N-acetylgalactosamine 3-beta-galactosyltransferase 1 (C1GALT1), phosphatidylinositol-4-phosphate 5-kinase type 1 beta (PIP5K1B), methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), brain enriched myelin associated protein 1 (BCAS1), and squalene epoxidase (SQLE) genes were increased (P < 0.01), indicating that nutrient metabolism activity was enhanced. The expression of the BCAS1, C1GALT1, KLF transcription factor 13 (KLF13), and leucine rich repeat neuronal 1 (LRRN1) was increased (P < 0.01), indicating that the cell proliferation was enhanced, which slowed uterus aging. In conclusion, compound Bacillus improved the eggshell strength and metabolite composition in the egg by promoting metabolic balance of calcium and phosphorus, cell proliferation, and nutrient metabolism in the uterus.
Collapse
Affiliation(s)
- Qian Jin
- Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya 572000, Hainan Province, China
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Fei Wang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Weisheng Ye
- Agriculture and Rural Bureau, Yunhe County 323600, Zhejiang Province, China
| | - Qi Wang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Shujie Xu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Shaoxiong Jiang
- Agriculture and Rural Bureau, Yunhe County 323600, Zhejiang Province, China
| | - Xiang Li
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Min Yue
- Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya 572000, Hainan Province, China
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Dongyou Yu
- Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya 572000, Hainan Province, China
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Mingliang Jin
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Aikun Fu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Weifen Li
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| |
Collapse
|
3
|
Jamerson LE, Bradshaw PC. The Roles of White Adipose Tissue and Liver NADPH in Dietary Restriction-Induced Longevity. Antioxidants (Basel) 2024; 13:820. [PMID: 39061889 PMCID: PMC11273496 DOI: 10.3390/antiox13070820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Dietary restriction (DR) protocols frequently employ intermittent fasting. Following a period of fasting, meal consumption increases lipogenic gene expression, including that of NADPH-generating enzymes that fuel lipogenesis in white adipose tissue (WAT) through the induction of transcriptional regulators SREBP-1c and CHREBP. SREBP-1c knockout mice, unlike controls, did not show an extended lifespan on the DR diet. WAT cytoplasmic NADPH is generated by both malic enzyme 1 (ME1) and the pentose phosphate pathway (PPP), while liver cytoplasmic NADPH is primarily synthesized by folate cycle enzymes provided one-carbon units through serine catabolism. During the daily fasting period of the DR diet, fatty acids are released from WAT and are transported to peripheral tissues, where they are used for beta-oxidation and for phospholipid and lipid droplet synthesis, where monounsaturated fatty acids (MUFAs) may activate Nrf1 and inhibit ferroptosis to promote longevity. Decreased WAT NADPH from PPP gene knockout stimulated the browning of WAT and protected from a high-fat diet, while high levels of NADPH-generating enzymes in WAT and macrophages are linked to obesity. But oscillations in WAT [NADPH]/[NADP+] from feeding and fasting cycles may play an important role in maintaining metabolic plasticity to drive longevity. Studies measuring the WAT malate/pyruvate as a proxy for the cytoplasmic [NADPH]/[NADP+], as well as studies using fluorescent biosensors expressed in the WAT of animal models to monitor the changes in cytoplasmic [NADPH]/[NADP+], are needed during ad libitum and DR diets to determine the changes that are associated with longevity.
Collapse
Affiliation(s)
| | - Patrick C. Bradshaw
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
4
|
Yu Y, Martins LM. Mitochondrial One-Carbon Metabolism and Alzheimer's Disease. Int J Mol Sci 2024; 25:6302. [PMID: 38928008 PMCID: PMC11203557 DOI: 10.3390/ijms25126302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
Mitochondrial one-carbon metabolism provides carbon units to several pathways, including nucleic acid synthesis, mitochondrial metabolism, amino acid metabolism, and methylation reactions. Late-onset Alzheimer's disease is the most common age-related neurodegenerative disease, characterised by impaired energy metabolism, and is potentially linked to mitochondrial bioenergetics. Here, we discuss the intersection between the molecular pathways linked to both mitochondrial one-carbon metabolism and Alzheimer's disease. We propose that enhancing one-carbon metabolism could promote the metabolic processes that help brain cells cope with Alzheimer's disease-related injuries. We also highlight potential therapeutic avenues to leverage one-carbon metabolism to delay Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Yizhou Yu
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - L. Miguel Martins
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
5
|
Rork AM, Bala AS, Renner T. Dynamic evolution of the mTHF gene family associated with primary metabolism across life. BMC Genomics 2024; 25:432. [PMID: 38693486 PMCID: PMC11064299 DOI: 10.1186/s12864-024-10159-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 02/25/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND The folate cycle of one-carbon (C1) metabolism, which plays a central role in the biosynthesis of nucleotides and amino acids, demonstrates the significance of metabolic adaptation. We investigated the evolutionary history of the methylenetetrahydrofolate dehydrogenase (mTHF) gene family, one of the main drivers of the folate cycle, across life. RESULTS Through comparative genomic and phylogenetic analyses, we found that several lineages of Archaea lacked domains vital for folate cycle function such as the mTHF catalytic and NAD(P)-binding domains of FolD. Within eukaryotes, the mTHF gene family diversified rapidly. For example, several duplications have been observed in lineages including the Amoebozoa, Opisthokonta, and Viridiplantae. In a common ancestor of Opisthokonta, FolD and FTHFS underwent fusion giving rise to the gene MTHFD1, possessing the domains of both genes. CONCLUSIONS Our evolutionary reconstruction of the mTHF gene family associated with a primary metabolic pathway reveals dynamic evolution, including gene birth-and-death, gene fusion, and potential horizontal gene transfer events and/or amino acid convergence.
Collapse
Affiliation(s)
- Adam M Rork
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA.
- Department of Entomology, Purdue University, West Lafayette, Indiana, 47907, USA.
| | - Arthi S Bala
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, 20007, USA
- School of Medicine, Georgetown University, Washington, DC, 20007, USA
| | - Tanya Renner
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA.
| |
Collapse
|
6
|
Wu D, Zhang K, Khan FA, Pandupuspitasari NS, Guan K, Sun F, Huang C. A comprehensive review on signaling attributes of serine and serine metabolism in health and disease. Int J Biol Macromol 2024; 260:129607. [PMID: 38253153 DOI: 10.1016/j.ijbiomac.2024.129607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024]
Abstract
Serine is a metabolite with ever-expanding metabolic and non-metabolic signaling attributes. By providing one‑carbon units for macromolecule biosynthesis and functional modifications, serine and serine metabolism largely impinge on cellular survival and function. Cancer cells frequently have a preference for serine metabolic reprogramming to create a conducive metabolic state for survival and aggressiveness, making intervention of cancer-associated rewiring of serine metabolism a promising therapeutic strategy for cancer treatment. Beyond providing methyl donors for methylation in modulation of innate immunity, serine metabolism generates formyl donors for mitochondrial tRNA formylation which is required for mitochondrial function. Interestingly, fully developed neurons lack the machinery for serine biosynthesis and rely heavily on astrocytic l-serine for production of d-serine to shape synaptic plasticity. Here, we recapitulate recent discoveries that address the medical significance of serine and serine metabolism in malignancies, mitochondrial-associated disorders, and neurodegenerative pathologies. Metabolic control and epigenetic- and posttranslational regulation of serine metabolism are also discussed. Given the metabolic similarities between cancer cells, neurons and germ cells, we further propose the relevance of serine metabolism in testicular homeostasis. Our work provides valuable hints for future investigations that will lead to a deeper understanding of serine and serine metabolism in cellular physiology and pathology.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat 10340, Indonesia
| | | | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| |
Collapse
|
7
|
Schiuma G, Lara D, Clement J, Narducci M, Rizzo R. NADH: the redox sensor in aging-related disorders. Antioxid Redox Signal 2024. [PMID: 38366731 DOI: 10.1089/ars.2023.0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
SIGNIFICANCE NADH represents the reduced form of NAD+, and together they constitute the two forms of the Nicotinamide adenine dinucleotide whose balance is named as the NAD+/NADH ratio. NAD+/NADH ratio is mainly involved in redox reactions since both the molecules are responsible for carrying electrons to maintain redox homeostasis. NADH acts as a reducing agent and one of the most known processes exploiting NADH function is energy metabolism. The two main pathways generating energy and involving NADH are Glycolysis and Oxidative phosphorylation, occurring in cell cytosol and in the mitochondrial matrix, respectively. RECENT ADVANCES Although NADH is primarily produced through the reduction of NAD+ and consumed by its own oxidation, several are the biosynthetic and consumption pathways, reflecting the NADH role in multiple cellular processes. CRITICAL ISSUES This review gathers all the main current data referring to NADH in correlation with metabolic and cellular pathways, such as its coenzyme activity, effect in cell death and on modulating redox and calcium homeostasis. Data were selected following eligibility criteria accordingly to the reviewed topic. A set of electronic databases (Medline/PubMed, Scopus, Web of Sciences (WOS), Cochrane Library) have been used for a systematic search until January 2024 using MeSH keywords/terms (i.e., NADH, NAD+/NADH and NADH/NAD+ ratio, redox homeostasis, energy metabolism, aging, aging-related disorders, therapies). FUTURE DIRECTION Gene expression control, as well as to the potential impact on neurodegenerative, cardiac disorders and infections suggest NADH application in clinical settings.
Collapse
Affiliation(s)
| | - Djidjell Lara
- University of Ferrara, 9299, Ferrara, FE, Italy
- BetterHumans, Gainesville, Florida, United States;
| | - James Clement
- Betterhumans Inc., Gainesville, Florida, United States
- University of Ferrara, 9299, Ferrara, FE, Italy;
| | - Marco Narducci
- University of Ferrara, 9299, Ferrara, FE, Italy
- BetterHumans, Gainesville, Florida, United States
- Temple University Japan Campus, 83908, Minato-ku, Tokyo, Japan;
| | - Roberta Rizzo
- University of Ferrara, 9299, Via Luigi Borsari 46, Ferrara, Ferrara, FE, Italy, 44121;
| |
Collapse
|
8
|
Li L, Zhang Y, Hu W, Zou F, Ning J, Rao T, Ruan Y, Yu W, Cheng F. MTHFD2 promotes PD-L1 expression via activation of the JAK/STAT signalling pathway in bladder cancer. J Cell Mol Med 2023; 27:2922-2936. [PMID: 37480214 PMCID: PMC10538262 DOI: 10.1111/jcmm.17863] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023] Open
Abstract
Although combination chemotherapy is widely used for bladder cancer (BC) treatment, the recurrence and progression rates remain high. Therefore, novel therapeutic targets are required. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) contributes to tumourigenesis and immune evasion in several cancers; however, its biological function in BC remains unknown. This study aimed to investigate the expression, prognostic value and protumoural function of MTHFD2 in BC and elucidate the mechanism of programmed death-ligand 1 (PD-L1) upregulation by MTHFD2. An analysis using publicly available databases revealed that a high MTHFD2 expression was correlated with clinical features and a poor prognosis in BC. Furthermore, MTHFD2 promoted the growth, migration, invasion and tumourigenicity and decreased the apoptosis of BC cells in vivo and in vitro. The results obtained from databases showed that MTHFD2 expression was correlated with immune infiltration levels, PD-L1 expression, and the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. The expression of MTHFD2, PD-L1 and JAK/STAT signalling pathway-related proteins increased after interferon gamma treatment and decreased after MTHFD2 knockdown. Moreover, addition of a JAK/STAT pathway activator partially reduced the effect of MTHFD2 knockdown on BC cells. Collectively, our findings suggest that MTHFD2 promotes the expression of PD-L1 through the JAK/STAT signalling pathway in BC.
Collapse
Affiliation(s)
- Linzhi Li
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yunlong Zhang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Weimin Hu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Fan Zou
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jinzhuo Ning
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Ting Rao
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yuan Ruan
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Weimin Yu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Fan Cheng
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
9
|
Tang YC, Chuang YJ, Chang HH, Juang SH, Yen GC, Chang JY, Kuo CC. How to deal with frenemy NRF2: Targeting NRF2 for chemoprevention and cancer therapy. J Food Drug Anal 2023; 31:387-407. [PMID: 39666284 PMCID: PMC10629913 DOI: 10.38212/2224-6614.3463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/09/2023] [Indexed: 12/13/2024] Open
Abstract
Induction of antioxidant proteins and phase 2 detoxifying enzymes that neutralize reactive electrophiles are important mechanisms for protection against carcinogenesis. Normal cells provide multifaceted pathways to tightly control NF-E2-related factor 2 (NRF2)-mediated gene expression in response to an assault by a range of endogenous and exogenous oncogenic molecules. Transient activation of NRF2 by its activators is able to induce ARE-mediated cytoprotective proteins which are essential for protection against various toxic and oxidative damages, and NRF2 activators thereby have efficacy in cancer chemoprevention. Because NRF2 has a cytoprotective function, it can protect normal cells from carcinogens like an angel, but when the protective effect acts on cancer cells, it will give rise to invincible cancer cells and play a devilish role in tumor progression. Indeed, aberrant activation of NRF2 has been found in a variety of cancers that create a favorable environment for the proliferation and survival of cancer cells and leads to drug resistance, ultimately leading to the poor clinical prognosis of patients. Therefore, pharmacological inhibition of NRF2 signaling has emerged as a promising approach for cancer therapy. This review aims to compile the regulatory mechanisms of NRF2 and its double-edged role in cancer. In addition, we also summarize the research progress of NRF2 modulators, especially phytochemicals, in chemoprevention and cancer therapy.
Collapse
Affiliation(s)
- Ya-Chu Tang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli,
Taiwan
| | - Yung-Jen Chuang
- School of Medicine, National Tsing Hua University, Hsinchu,
Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu,
Taiwan
| | - Hsin-Huei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli,
Taiwan
| | - Shin-Hun Juang
- School of Pharmacy, China Medical University, Taichung,
Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung,
Taiwan
| | - Jang-Yang Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli,
Taiwan
- Taipei Cancer Center, Taipei Medical University Hospital, Taipei,
Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei,
Taiwan
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli,
Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung,
Taiwan
| |
Collapse
|
10
|
Wu D, Zhang K, Khan FA, Wu Q, Pandupuspitasari NS, Tang Y, Guan K, Sun F, Huang C. The emerging era of lactate: A rising star in cellular signaling and its regulatory mechanisms. J Cell Biochem 2023; 124:1067-1081. [PMID: 37566665 DOI: 10.1002/jcb.30458] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/19/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Cellular metabolites are ancient molecules with pleiotropic implications in health and disease. Beyond their cognate roles, they have signaling functions as the ligands for specific receptors and the precursors for epigenetic or posttranslational modifications. Lactate has long been recognized as a metabolic waste and fatigue product mainly produced from glycolytic metabolism. Recent evidence however suggests lactate is an unique molecule with diverse signaling attributes in orchestration of numerous biological processes, including tumor immunity and neuronal survival. The copious metabolic and non-metabolic functions of lactate mediated by its bidirectional shuttle between cells or intracellular organelles lead to a phenotype called "lactormone." Importantly, the mechanisms of lactate signaling, via acting as a molecular sensor and a regulator of NAD+ metabolism and AMP-activated protein kinase signaling, and via the newly identified lactate-driven lactylation, have been discovered. Further, we include a brief discussion about the autocrine regulation of efferocytosis by lactate in Sertoli cells which favoraerobic glycolysis. By emphasizing a repertoire of the most recent discovered mechanisms of lactate signaling, this review will open tantalizing avenues for future investigations cracking the regulatory topology of lactate signaling covered in the veil of mystery.
Collapse
Affiliation(s)
- Di Wu
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Kejia Zhang
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, Ministry of Research and Technology National Research and Innovation Agency, Jakarta, Indonesia
| | - Qin Wu
- Jinan Second People's Hospital & The Ophthalmologic Hospital of Jinan, Jinan, China
| | | | - Yuan Tang
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing, China
| | - Fei Sun
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Chunjie Huang
- School of Medicine, Institute of Reproductive Medicine, Nantong University, Nantong, China
| |
Collapse
|
11
|
Cappello A, Zuccotti A, Mancini M, Tosetti G, Fania L, Ricci F, Melino G, Candi E. Serine and one-carbon metabolism sustain non-melanoma skin cancer progression. Cell Death Discov 2023; 9:102. [PMID: 36964165 PMCID: PMC10039038 DOI: 10.1038/s41420-023-01398-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/26/2023] Open
Abstract
Non-melanoma skin cancer (NMSC) is a tumor that arises from human keratinocytes, showing abnormal control of cell proliferation and aberrant stratification. Cutaneous basal cell carcinoma (cBCC) and cutaneous squamous cell carcinoma (cSCC) are the most common sub-types of NMSC. From a molecular point of view, we are still far from fully understanding the molecular mechanisms behind the onset and progression of NMSC and to unravel targetable vulnerabilities to leverage for their treatment, which is still essentially based on surgery. Under this assumption, it is still not elucidated how the central cellular metabolism, a potential therapeutical target, is involved in NMSC progression. Therefore, our work is based on the characterization of the serine anabolism/catabolism and/or one-carbon metabolism (OCM) role in NMSC pathogenesis. Expression and protein analysis of normal skin and NMSC samples show the alteration of the expression of two enzymes involved in the serine metabolism and OCM, the Serine Hydroxy-Methyl Transferase 2 (SHMT2) and Methylen-ThetraHydroFolate dehydrogenase/cyclohydrolase 2 (MTHFD2). Tissues analysis shows that these two enzymes are mainly expressed in the proliferative areas of cBCC and in the poorly differentiated areas of cSCC, suggesting their role in tumor proliferation maintenance. Moreover, in vitro silencing of SHMT2 and MTHFD2 impairs the proliferation of epidermoid cancer cell line. Taken together these data allow us to link the central cellular metabolism (serine and/or OCM) and NMSC proliferation and progression, offering the opportunity to modulate pharmacologically the involved enzymes activity against this type of human cancer.
Collapse
Affiliation(s)
- Angela Cappello
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, 00167, Rome, Italy
| | | | - Mara Mancini
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, 00167, Rome, Italy
| | - Giulia Tosetti
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Luca Fania
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, 00167, Rome, Italy
| | - Francesco Ricci
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, 00167, Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy.
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, 00167, Rome, Italy.
| |
Collapse
|
12
|
Mann JP, Duan X, Patel S, Tábara LC, Scurria F, Alvarez-Guaita A, Haider A, Luijten I, Page M, Protasoni M, Lim K, Virtue S, O'Rahilly S, Armstrong M, Prudent J, Semple RK, Savage DB. A mouse model of human mitofusin-2-related lipodystrophy exhibits adipose-specific mitochondrial stress and reduced leptin secretion. eLife 2023; 12:e82283. [PMID: 36722855 PMCID: PMC9937658 DOI: 10.7554/elife.82283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/30/2023] [Indexed: 02/02/2023] Open
Abstract
Mitochondrial dysfunction has been reported in obesity and insulin resistance, but primary genetic mitochondrial dysfunction is generally not associated with these, arguing against a straightforward causal relationship. A rare exception, recently identified in humans, is a syndrome of lower body adipose loss, leptin-deficient severe upper body adipose overgrowth, and insulin resistance caused by the p.Arg707Trp mutation in MFN2, encoding mitofusin 2. How the resulting selective form of mitochondrial dysfunction leads to tissue- and adipose depot-specific growth abnormalities and systemic biochemical perturbation is unknown. To address this, Mfn2R707W/R707W knock-in mice were generated and phenotyped on chow and high fat diets. Electron microscopy revealed adipose-specific mitochondrial morphological abnormalities. Oxidative phosphorylation measured in isolated mitochondria was unperturbed, but the cellular integrated stress response was activated in adipose tissue. Fat mass and distribution, body weight, and systemic glucose and lipid metabolism were unchanged, however serum leptin and adiponectin concentrations, and their secretion from adipose explants were reduced. Pharmacological induction of the integrated stress response in wild-type adipocytes also reduced secretion of leptin and adiponectin, suggesting an explanation for the in vivo findings. These data suggest that the p.Arg707Trp MFN2 mutation selectively perturbs mitochondrial morphology and activates the integrated stress response in adipose tissue. In mice, this does not disrupt most adipocyte functions or systemic metabolism, whereas in humans it is associated with pathological adipose remodelling and metabolic disease. In both species, disproportionate effects on leptin secretion may relate to cell autonomous induction of the integrated stress response.
Collapse
Affiliation(s)
- Jake P Mann
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Xiaowen Duan
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Satish Patel
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Luis Carlos Tábara
- Medical Research Council Mitochondrial Biology Unit, University of CambridgeCambridgeUnited Kingdom
| | - Fabio Scurria
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Anna Alvarez-Guaita
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Afreen Haider
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Ineke Luijten
- Centre for Cardiovascular Science, University of EdinburghEdinburghUnited Kingdom
| | | | - Margherita Protasoni
- Medical Research Council Mitochondrial Biology Unit, University of CambridgeCambridgeUnited Kingdom
| | - Koini Lim
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Sam Virtue
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Stephen O'Rahilly
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | | | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of CambridgeCambridgeUnited Kingdom
| | - Robert K Semple
- Centre for Cardiovascular Science, University of EdinburghEdinburghUnited Kingdom
- MRC Human Genetics Unit, University of EdinburghEdinburghUnited Kingdom
| | - David B Savage
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
13
|
Scaletti ER, Gustafsson Westergren R, Andersson Y, Wiita E, Henriksson M, Homan EJ, Jemth A, Helleday T, Stenmark P. The First Structure of Human MTHFD2L and Its Implications for the Development of Isoform-Selective Inhibitors. ChemMedChem 2022; 17:e202200274. [PMID: 35712863 PMCID: PMC9796130 DOI: 10.1002/cmdc.202200274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/15/2022] [Indexed: 01/01/2023]
Abstract
Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) is a mitochondrial 1-carbon metabolism enzyme, which is an attractive anticancer drug target as it is highly upregulated in cancer but is not expressed in healthy adult cells. Selective MTHFD2 inhibitors could therefore offer reduced side-effects during treatment, which are common with antifolate drugs that target other 1C-metabolism enzymes. This task is challenging however, as MTHFD2 shares high sequence identity with the constitutively expressed isozymes cytosolic MTHFD1 and mitochondrial MTHFD2L. In fact, one of the most potent MTHFD2 inhibitors reported to date, TH7299, is actually more active against MTHFD1 and MTHFD2L. While structures of MTHFD2 and MTHFD1 exist, no MTHFD2L structures are available. We determined the first structure of MTHFD2L and its complex with TH7299, which reveals the structural basis for its highly potent MTHFD2L inhibition. Detailed analysis of the MTHFD2L structure presented here clearly highlights the challenges associated with developing truly isoform-selective MTHFD2 inhibitors.
Collapse
Affiliation(s)
- Emma R. Scaletti
- Department of Biochemistry and BiophysicsStockholm UniversitySvante Arrhenius väg 16 CStockholm106 91Sweden
| | | | - Yasmin Andersson
- Drug Discovery and Development Platform, Science for Life Laboratory School of BiotechnologyRoyal Institute of TechnologyTomtebodavägen 23aStockholm17165Sweden
| | - Elisee Wiita
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
| | - Martin Henriksson
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
| | - Evert J. Homan
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
| | - Ann‐Sofie Jemth
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
| | - Thomas Helleday
- Science for Life LaboratoryDepartment of Oncology-PathologyKarolinska InstituteTomtebodavägen 23aStockholm171 65Sweden
- Department of Oncology and MetabolismThe University of SheffieldBeech Hill RoadSheffieldS10 2RXUK
| | - Pål Stenmark
- Department of Biochemistry and BiophysicsStockholm UniversitySvante Arrhenius väg 16 CStockholm106 91Sweden
| |
Collapse
|
14
|
ATF4/MYC Regulates MTHFD2 to Promote NSCLC Progression by Mediating Redox Homeostasis. DISEASE MARKERS 2022; 2022:7527996. [PMID: 36051358 PMCID: PMC9425107 DOI: 10.1155/2022/7527996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022]
Abstract
Purpose. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) has been reported to be overexpressed in non-small-cell lung cancer (NSCLC) and to correlate with malignant proliferation. However, the mechanism of high MTHFD2 expression in NSCLC has not been clarified. Methods. qPCR, western blot, and immunofluorescence experiments were used to measure the expression of related mRNAs and proteins. Cell apoptosis was measured by flow cytometry and TUNEL assays. The CCK-8 assay was used to determine cell viability. Flow cytometry was used to analyze the cell cycle. ROS, H2O2, MDA, SOD, and NADPH/NADP+ were evaluated by relevant assay kits. Transfection of siRNA or vectors was used to downregulate or upregulate gene expression. Dual-luciferase reporter gene assays were used to evaluate the regulated relationship between MTHFD2 and ATF4 or MYC. Results. MTHFD2 was highly expressed in NSCLC cells. Knockdown of MTHFD2 inhibited proliferation and increased apoptosis. Furthermore, oxidative factors significantly increased, while antioxidant factors significantly decreased in NSCLC cells with MTHFD2 knockdown, indicating that MTHFD2 was involved in NSCLC progression through the redox pathway. Although MTHFD2 was downregulated with ATF4 silencing, the dual-luciferase reporter assay suggested that ATF4 did not directly mediate MTHFD2 transcription. Further studies revealed that MYC had a transcriptional effect on MTHFD2 and was also regulated by ATF4. PCR, and western blotting experiments with ATF4 knockdown and MYC overexpression as well as ATF4 overexpression and MYC knockdown proved that ATF4 stimulated MTHFD2 through MYC mediation. Conclusions. ATF4 promoted high expression of MTHFD2 in NSCLC dependent on MYC.
Collapse
|
15
|
Koju N, Qin ZH, Sheng R. Reduced nicotinamide adenine dinucleotide phosphate in redox balance and diseases: a friend or foe? Acta Pharmacol Sin 2022; 43:1889-1904. [PMID: 35017669 PMCID: PMC9343382 DOI: 10.1038/s41401-021-00838-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022]
Abstract
The nicotinamide adenine dinucleotide (NAD+/NADH) and nicotinamide adenine dinucleotide phosphate (NADP+/NADPH) redox couples function as cofactors or/and substrates for numerous enzymes to retain cellular redox balance and energy metabolism. Thus, maintaining cellular NADH and NADPH balance is critical for sustaining cellular homeostasis. The sources of NADPH generation might determine its biological effects. Newly-recognized biosynthetic enzymes and genetically encoded biosensors help us better understand how cells maintain biosynthesis and distribution of compartmentalized NAD(H) and NADP(H) pools. It is essential but challenging to distinguish how cells sustain redox couple pools to perform their integral functions and escape redox stress. However, it is still obscure whether NADPH is detrimental or beneficial as either deficiency or excess in cellular NADPH levels disturbs cellular redox state and metabolic homeostasis leading to redox stress, energy stress, and eventually, to the disease state. Additional study of the pathways and regulatory mechanisms of NADPH generation in different compartments, and the means by which NADPH plays a role in various diseases, will provide innovative insights into its roles in human health and may find a value of NADPH for the treatment of certain diseases including aging, Alzheimer's disease, Parkinson's disease, cardiovascular diseases, ischemic stroke, diabetes, obesity, cancer, etc.
Collapse
Affiliation(s)
- Nirmala Koju
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
16
|
The catalytic mechanism of the mitochondrial methylenetetrahydrofolate dehydrogenase/cyclohydrolase (MTHFD2). PLoS Comput Biol 2022; 18:e1010140. [PMID: 35613161 PMCID: PMC9173628 DOI: 10.1371/journal.pcbi.1010140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/07/2022] [Accepted: 04/26/2022] [Indexed: 11/19/2022] Open
Abstract
Methylenetetrahydrofolate dehydrogenase/cyclohydrolase (MTHFD2) is a new drug target that is expressed in cancer cells but not in normal adult cells, which provides an Achilles heel to selectively kill cancer cells. Despite the availability of crystal structures of MTHFD2 in the inhibitor- and cofactor-bound forms, key information is missing due to technical limitations, including (a) the location of absolutely required Mg2+ ion, and (b) the substrate-bound form of MTHFD2. Using computational modeling and simulations, we propose that two magnesium ions are present at the active site whereby (i) Arg233, Asp225, and two water molecules coordinate MgA2+, while MgA2+ together with Arg233 stabilize the inorganic phosphate (Pi); (ii) Asp168 and three water molecules coordinate MgB2+, and MgB2+ further stabilizes Pi by forming a hydrogen bond with two oxygens of Pi; (iii) Arg201 directly coordinates the Pi; and (iv) through three water-mediated interactions, Asp168 contributes to the positioning and stabilization of MgA2+, MgB2+ and Pi. Our computational study at the empirical valence bond level allowed us also to elucidate the detailed reaction mechanisms. We found that the dehydrogenase activity features a proton-coupled electron transfer with charge redistribution connected to the reorganization of the surrounding water molecules which further facilitates the subsequent cyclohydrolase activity. The cyclohydrolase activity then drives the hydration of the imidazoline ring and the ring opening in a concerted way. Furthermore, we have uncovered that two key residues, Ser197/Arg233, are important factors in determining the cofactor (NADP+/NAD+) preference of the dehydrogenase activity. Our work sheds new light on the structural and kinetic framework of MTHFD2, which will be helpful to design small molecule inhibitors that can be used for cancer treatment.
Collapse
|
17
|
Zhao LN, Kaldis P. Pairing structural reconstruction with catalytic competence to evaluate the mechanisms of key enzymes in the folate-mediated one-carbon pathway. FEBS J 2022; 290:2279-2291. [PMID: 35303396 DOI: 10.1111/febs.16439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/05/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023]
Abstract
Mammalian metabolism comprises a series of interlinking pathways that include two major cycles: the folate and methionine cycles. The folate-mediated metabolic cycle uses several oxidation states of tetrahydrofolate to carry activated one-carbon units to be readily used and interconverted within the cell. They are required for nucleotide synthesis, methylation and metabolism, and particularly for proliferation of cancer cells. Based on the latest progress in genome-wide CRISPR loss-of-function viability screening of 789 cell lines, we focus on the most cancer-dependent enzymes in this pathway, especially those that are hyperactivated in cancer, to provide new insight into the chemical basis for cancer drug development. Since the complete 3D structure of several of these enzymes of the one-carbon pathway in their active form are not available, we used homology modelling integrated with the interpretation of the reaction mechanism. In addition, have reconstructed the most likely scenario for the reactions taking place paired with their catalytic competence that provides a testable framework for this pathway.
Collapse
Affiliation(s)
- Li Na Zhao
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
18
|
Sugiura A, Andrejeva G, Voss K, Heintzman DR, Xu X, Madden MZ, Ye X, Beier KL, Chowdhury NU, Wolf MM, Young AC, Greenwood DL, Sewell AE, Shahi SK, Freedman SN, Cameron AM, Foerch P, Bourne T, Garcia-Canaveras JC, Karijolich J, Newcomb DC, Mangalam AK, Rabinowitz JD, Rathmell JC. MTHFD2 is a metabolic checkpoint controlling effector and regulatory T cell fate and function. Immunity 2022; 55:65-81.e9. [PMID: 34767747 PMCID: PMC8755618 DOI: 10.1016/j.immuni.2021.10.011] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 07/23/2021] [Accepted: 10/13/2021] [Indexed: 01/13/2023]
Abstract
Antigenic stimulation promotes T cell metabolic reprogramming to meet increased biosynthetic, bioenergetic, and signaling demands. We show that the one-carbon (1C) metabolism enzyme methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) regulates de novo purine synthesis and signaling in activated T cells to promote proliferation and inflammatory cytokine production. In pathogenic T helper-17 (Th17) cells, MTHFD2 prevented aberrant upregulation of the transcription factor FoxP3 along with inappropriate gain of suppressive capacity. MTHFD2 deficiency also promoted regulatory T (Treg) cell differentiation. Mechanistically, MTHFD2 inhibition led to depletion of purine pools, accumulation of purine biosynthetic intermediates, and decreased nutrient sensor mTORC1 signaling. MTHFD2 was also critical to regulate DNA and histone methylation in Th17 cells. Importantly, MTHFD2 deficiency reduced disease severity in multiple in vivo inflammatory disease models. MTHFD2 is thus a metabolic checkpoint to integrate purine metabolism with pathogenic effector cell signaling and is a potential therapeutic target within 1C metabolism pathways.
Collapse
Affiliation(s)
- Ayaka Sugiura
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Gabriela Andrejeva
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kelsey Voss
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Darren R Heintzman
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xincheng Xu
- Department of Chemistry, Ludwig Cancer Research Institute Princeton Branch, Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Matthew Z Madden
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xiang Ye
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Katherine L Beier
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nowrin U Chowdhury
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Melissa M Wolf
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Arissa C Young
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dalton L Greenwood
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Allison E Sewell
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Shailesh K Shahi
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Alanna M Cameron
- Sitryx Therapeutics Limited, Magdalen Centre, Oxford Science Park, Oxford, UK
| | - Patrik Foerch
- Sitryx Therapeutics Limited, Magdalen Centre, Oxford Science Park, Oxford, UK
| | - Tim Bourne
- Sitryx Therapeutics Limited, Magdalen Centre, Oxford Science Park, Oxford, UK
| | - Juan C Garcia-Canaveras
- Department of Chemistry, Ludwig Cancer Research Institute Princeton Branch, Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - John Karijolich
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dawn C Newcomb
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Joshua D Rabinowitz
- Department of Chemistry, Ludwig Cancer Research Institute Princeton Branch, Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Jeffrey C Rathmell
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
19
|
Quevedo-Ocampo J, Escobedo-Calvario A, Souza-Arroyo V, Miranda-Labra RU, Bucio-Ortiz L, Gutiérrez-Ruiz MC, Chávez-Rodríguez L, Gomez-Quiroz LE. Folate Metabolism in Hepatocellular Carcinoma. What Do We Know So Far? Technol Cancer Res Treat 2022; 21:15330338221144446. [PMID: 36503290 DOI: 10.1177/15330338221144446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancer cells are characterized by accelerated proliferation and an outstanding adaptation of their metabolic pathways to meet energy demands. The folate cycle, also known as folate metabolism or one-carbon metabolism, through enzymatic interconversions, provides metabolites necessary for nucleotide synthesis, methylation, and reduction power, helping to maintain the high rate of proliferation; therefore, the study of this metabolic pathway is of great importance in the study of cancer. Moreover, multiple enzymes involved in this cycle have been implicated in different types of cancer, corroborating the cell's adaptations under this pathology. During the last decade, nonalcoholic fatty liver disease has emerged as the leading etiology related to the rise in the incidence and deaths of hepatocellular carcinoma. Specifically, cholesterol accumulation has been a determinant promoter of tumor formation, with solid evidence that an enriched-cholesterol diet plays a crucial role in accelerating the development of an aggressive subtype of hepatocellular carcinoma compared to other models. In this review, we will discuss the most recent findings to understand the contribution of folate metabolism to cancer cells and tumor microenvironment while creating a link between the dynamics given by cholesterol and methylenetetrahydrofolate dehydrogenase 1-like, a key enzyme of the cycle located in the mitochondrial compartment.
Collapse
Affiliation(s)
- Jaqueline Quevedo-Ocampo
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metrolitana-Iztapalapa, Mexico City, Mexico
| | - Alejandro Escobedo-Calvario
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metrolitana-Iztapalapa, Mexico City, Mexico
| | - Verónica Souza-Arroyo
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Roxana U Miranda-Labra
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Leticia Bucio-Ortiz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - María C Gutiérrez-Ruiz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Lisette Chávez-Rodríguez
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metrolitana-Iztapalapa, Mexico City, Mexico
| | - Luis E Gomez-Quiroz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
20
|
Folate Transport and One-Carbon Metabolism in Targeted Therapies of Epithelial Ovarian Cancer. Cancers (Basel) 2021; 14:cancers14010191. [PMID: 35008360 PMCID: PMC8750473 DOI: 10.3390/cancers14010191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022] Open
Abstract
New therapies are urgently needed for epithelial ovarian cancer (EOC), the most lethal gynecologic malignancy. To identify new approaches for targeting EOC, metabolic vulnerabilities must be discovered and strategies for the selective delivery of therapeutic agents must be established. Folate receptor (FR) α and the proton-coupled folate transporter (PCFT) are expressed in the majority of EOCs. FRβ is expressed on tumor-associated macrophages, a major infiltrating immune population in EOC. One-carbon (C1) metabolism is partitioned between the cytosol and mitochondria and is important for the synthesis of nucleotides, amino acids, glutathione, and other critical metabolites. Novel inhibitors are being developed with the potential for therapeutic targeting of tumors via FRs and the PCFT, as well as for inhibiting C1 metabolism. In this review, we summarize these exciting new developments in targeted therapies for both tumors and the tumor microenvironment in EOC.
Collapse
|
21
|
Liu X, Liu S, Piao C, Zhang Z, Zhang X, Jiang Y, Kong C. Non-metabolic function of MTHFD2 activates CDK2 in bladder cancer. Cancer Sci 2021; 112:4909-4919. [PMID: 34632667 PMCID: PMC8645701 DOI: 10.1111/cas.15159] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/30/2021] [Accepted: 10/03/2021] [Indexed: 12/24/2022] Open
Abstract
Bladder cancer is a common tumor with a high recurrence rate and high fatality rate, and its mechanism of occurrence and development remains unclear. Many proteins and metabolites reprogram at different stages of tumor development to support tumor cell growth. The moonlighting effect happens when a protein performs multiple functions simultaneously in a cell. In this study, we identified a metabolic protein, MTHFD2, which participates in the cell cycle by binding to CDK2 in bladder cancer. MTHFD2 has been shown to affect bladder cancer cell growth, which is independent of its metabolic function. We found that MTHFD2 was involved in cell cycle regulation and could encourage cell cycle progression by activating CDK2 and sequentially affecting E2F1 activation. In addition, moonlighting MTHFD2 might be regulated by the dynamics of the mitochondria. In conclusion, MTHFD2 localizes in the nucleus to perform a distinct function of catalyzing metabolic reactions. Moreover, the nuclear MTHFD2 activates CDK2 and promotes bladder cancer cell growth by modulating the cell cycle.
Collapse
Affiliation(s)
- Xi Liu
- Department of UrologyThe First Hospital of China Medical UniversityShenyangChina
| | - Shuangjie Liu
- Department of UrologyThe First Hospital of China Medical UniversityShenyangChina
| | - Chiyuan Piao
- Department of UrologyThe First Hospital of China Medical UniversityShenyangChina
| | - Zhe Zhang
- Department of UrologyThe First Hospital of China Medical UniversityShenyangChina
| | - Xiaotong Zhang
- Department of UrologyThe First Hospital of China Medical UniversityShenyangChina
| | - Yuanjun Jiang
- Department of UrologyThe First Hospital of China Medical UniversityShenyangChina
| | - Chuize Kong
- Department of UrologyThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
22
|
Huang J, Qin Y, Lin C, Huang X, Zhang F. MTHFD2 facilitates breast cancer cell proliferation via the AKT signaling pathway. Exp Ther Med 2021; 22:703. [PMID: 34007312 PMCID: PMC8120508 DOI: 10.3892/etm.2021.10135] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 10/20/2020] [Indexed: 02/05/2023] Open
Abstract
MTHFD2 is a folate-coupled mitochondrial metabolic enzyme which has been extensively studied in breast cancer; however, its molecular functions in this cancer remain unclear. The current study aimed to reveal the underlying mechanism of breast cancer. MTHFD2 expression status and prognostic value were determined using the Gene Expression Profiling Interactive Analysis database. To determine the function of MTHFD2 in breast cancer, MCF-7 cells with stable overexpression of Flag-MTHFD2 or depletion of MTHFD2 were generated. Cell Counting Kit-8 and colony formation assays were used to examine the effect of MTHFD2 overexpression or knockout on MCF-7 cell proliferation and clonogenicity, respectively. Luciferase reporter and an AKT inhibitor (GSK6906) analysis were carried out to investigate the effect of MTHFD2 on the AKT signaling pathway. The results demonstrated that MTHFD2 expression level was higher in breast cancer tissues compared with adjacent normal tissues. Furthermore, patients with high MTHFD2 expression had significantly poorer overall survival compared with patients with low MTHFD2 expression. In addition, ectopic expression of MTHFD2 promoted the tumorigenic properties of MCF-7 cells, including proliferation and clonogenicity. Conversely, depletion of MTHFD2 had the opposite effect on the malignant properties of MCF-7 cells. Luciferase reporter demonstrated that MTHFD2 can significantly increase the ATK luciferase density. Furthermore, the Akt inhibitor GSK690693 significantly decreased the increased clonogenicity caused by MTHFD2 overexpression in MCF-7 cells. Taken together, the findings from the present study suggested that MTHFD2 may serve a protumor role in the malignancy of breast cancer by activating the AKT signaling pathway. These results provide an alternative theoretical foundation that could help the development of MTHFD2-targeted breast cancer treatment.
Collapse
Affiliation(s)
- Jun Huang
- Department of General Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Jinping, Shantou, Guangzhou 515000, P.R. China
| | - Yinyin Qin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Jinping, Shantou, Guangzhou 515000, P.R. China
- Pulmonary and Critical Care Medicine (PCCM), Shunde Affiliated Hospital of Guangzhou Medical University, Jinping, Shantou, Guangzhou 515000, P.R. China
| | - Canfeng Lin
- Department of Oncology, Shantou Central Hospital, Jinping, Shantou, Guangzhou 515000, P.R. China
| | - Xiaoguang Huang
- Department of Oncology, Shantou Central Hospital, Jinping, Shantou, Guangzhou 515000, P.R. China
| | - Feiran Zhang
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Jinping, Shantou, Guangdong 515041, P.R. China
- Correspondence to: Professor Feiran Zhang, Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Jinping, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
23
|
Pällmann N, Deng K, Livgård M, Tesikova M, Jin Y, Frengen NS, Kahraman N, Mokhlis HM, Ozpolat B, Kildal W, Danielsen HE, Fazli L, Rennie PS, Banerjee PP, Üren A, Jin Y, Kuzu OF, Saatcioglu F. Stress-Mediated Reprogramming of Prostate Cancer One-Carbon Cycle Drives Disease Progression. Cancer Res 2021; 81:4066-4078. [PMID: 34183356 DOI: 10.1158/0008-5472.can-20-3956] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/01/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022]
Abstract
One-carbon (1C) metabolism has a key role in metabolic programming with both mitochondrial (m1C) and cytoplasmic (c1C) components. Here we show that activating transcription factor 4 (ATF4) exclusively activates gene expression involved in m1C, but not the c1C cycle in prostate cancer cells. This includes activation of methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) expression, the central player in the m1C cycle. Consistent with the key role of m1C cycle in prostate cancer, MTHFD2 knockdown inhibited prostate cancer cell growth, prostatosphere formation, and growth of patient-derived xenograft organoids. In addition, therapeutic silencing of MTHFD2 by systemically administered nanoliposomal siRNA profoundly inhibited tumor growth in preclinical prostate cancer mouse models. Consistently, MTHFD2 expression is significantly increased in human prostate cancer, and a gene expression signature based on the m1C cycle has significant prognostic value. Furthermore, MTHFD2 expression is coordinately regulated by ATF4 and the oncoprotein c-MYC, which has been implicated in prostate cancer. These data suggest that the m1C cycle is essential for prostate cancer progression and may serve as a novel biomarker and therapeutic target. SIGNIFICANCE: These findings demonstrate that the mitochondrial, but not cytoplasmic, one-carbon cycle has a key role in prostate cancer cell growth and survival and may serve as a biomarker and/or therapeutic target.
Collapse
Affiliation(s)
- Nora Pällmann
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ke Deng
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Marte Livgård
- Department of Biosciences, University of Oslo, Oslo, Norway.,Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Martina Tesikova
- Department of Mathematics and Science, University of South-Eastern Norway, Borre, Norway
| | - Yixin Jin
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | - Nermin Kahraman
- Gynecological Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Hamada M Mokhlis
- Gynecological Oncology, MD Anderson Cancer Center, Houston, Texas.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Bulent Ozpolat
- Gynecological Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Wanja Kildal
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Havard Emil Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway.,Center for Cancer Biomedicine, University of Oslo, Oslo, Norway.,Department of Informatics, University of Oslo, Oslo, Norway.,Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK
| | - Ladan Fazli
- The Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Paul S Rennie
- The Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Partha P Banerjee
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Aykut Üren
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Yang Jin
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Omer F Kuzu
- Department of Biosciences, University of Oslo, Oslo, Norway.
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway. .,Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
24
|
Ababneh E, Saad AM, Crane GM. The role of EBV in haematolymphoid proliferations: emerging concepts relevant to diagnosis and treatment. Histopathology 2021; 79:451-464. [PMID: 33829526 DOI: 10.1111/his.14379] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/22/2021] [Accepted: 04/04/2021] [Indexed: 12/18/2022]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous gammaherpesvirus with >90% of the adult population worldwide harbouring latent infection. A small subset of those infected develop EBV-associated neoplasms, including a range of lymphoproliferative disorders (LPD). The diagnostic distinction of these entities appears increasingly relevant as our understanding of EBV-host interactions and mechanisms of EBV-driven lymphomagenesis improves. EBV may lower the mutational threshold for malignant transformation, create potential vulnerabilities related to viral alteration of cell metabolism and allow for improved immune targeting. However, these tumours may escape immune surveillance by affecting their immune microenvironment, limiting viral gene expression or potential loss of the viral episome. Methods to manipulate the latency state of the virus to enhance immunogenicity are emerging as well as the potential to detect so-called 'hit and run' cases where EBV has been lost. Finally, measurement of EBV DNA remains an important biomarker for screening and monitoring of LPD. Methods to distinguish EBV DNA derived from virions during lytic activation from latent, methylated EBV DNA present in EBV-associated neoplasms may broaden the utility of this testing, particularly in patients with compromised immune function. We highlight some of these emerging areas relevant to the diagnosis and treatment of EBV-associated LPD with potential applicability to other EBV-associated neoplasms.
Collapse
Affiliation(s)
- Emad Ababneh
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland, OH, USA
| | - Anas M Saad
- Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Genevieve M Crane
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland, OH, USA
| |
Collapse
|
25
|
Zhao LN, Björklund M, Caldez MJ, Zheng J, Kaldis P. Therapeutic targeting of the mitochondrial one-carbon pathway: perspectives, pitfalls, and potential. Oncogene 2021; 40:2339-2354. [PMID: 33664451 DOI: 10.1038/s41388-021-01695-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023]
Abstract
Most of the drugs currently prescribed for cancer treatment are riddled with substantial side effects. In order to develop more effective and specific strategies to treat cancer, it is of importance to understand the biology of drug targets, particularly the newly emerging ones. A comprehensive evaluation of these targets will benefit drug development with increased likelihood for success in clinical trials. The folate-mediated one-carbon (1C) metabolism pathway has drawn renewed attention as it is often hyperactivated in cancer and inhibition of this pathway displays promise in developing anticancer treatment with fewer side effects. Here, we systematically review individual enzymes in the 1C pathway and their compartmentalization to mitochondria and cytosol. Based on these insight, we conclude that (1) except the known 1C targets (DHFR, GART, and TYMS), MTHFD2 emerges as good drug target, especially for treating hematopoietic cancers such as CLL, AML, and T-cell lymphoma; (2) SHMT2 and MTHFD1L are potential drug targets; and (3) MTHFD2L and ALDH1L2 should not be considered as drug targets. We highlight MTHFD2 as an excellent therapeutic target and SHMT2 as a complementary target based on structural/biochemical considerations and up-to-date inhibitor development, which underscores the perspectives of their therapeutic potential.
Collapse
Affiliation(s)
- Li Na Zhao
- Department of Clinical Sciences, Lund University, Malmö, Sweden.
| | - Mikael Björklund
- Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, Haining, Zhejiang, PR China.,2nd Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China.,Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Matias J Caldez
- Laboratory of Host Defense, The World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Jie Zheng
- School of Information Science and Technology, Shanghai Tech University, Shanghai, PR China
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Malmö, Sweden.
| |
Collapse
|
26
|
Cuthbertson CR, Arabzada Z, Bankhead A, Kyani A, Neamati N. A Review of Small-Molecule Inhibitors of One-Carbon Enzymes: SHMT2 and MTHFD2 in the Spotlight. ACS Pharmacol Transl Sci 2021; 4:624-646. [PMID: 33860190 DOI: 10.1021/acsptsci.0c00223] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Indexed: 02/06/2023]
Abstract
Metabolic reprogramming is a key hallmark of cancer and shifts cellular metabolism to meet the demands of biomass production necessary for abnormal cell reproduction. One-carbon metabolism (1CM) contributes to many biosynthetic pathways that fuel growth and is comprised of a complex network of enzymes. Methotrexate and 5-fluorouracil were pioneering drugs in this field and are still widely used today as anticancer agents as well as for other diseases such as arthritis. Besides dihydrofolate reductase and thymidylate synthase, two other enzymes of the folate cycle arm of 1CM have not been targeted clinically: serine hydroxymethyltransferase (SHMT) and methylenetetrahydrofolate dehydrogenase (MTHFD). An increasing body of literature suggests that the mitochondrial isoforms of these enzymes (SHMT2 and MTHFD2) are clinically relevant in the context of cancer. In this review, we focused on the 1CM pathway as a target for cancer therapy and, in particular, SHMT2 and MTHFD2. The function, regulation, and clinical relevance of SHMT2 and MTHFD2 are all discussed. We expand on previous clinical studies and evaluate the prognostic significance of these critical enzymes by performing a pan-cancer analysis of patient data from the The Cancer Genome Atlas and a transcriptional coexpression network enrichment analysis. We also provide an overview of preclinical and clinical inhibitors targeting the folate pathway, the methionine cycle, and folate-dependent purine biosynthesis enzymes.
Collapse
Affiliation(s)
- Christine R Cuthbertson
- Department of Medicinal Chemistry, College of Pharmacy and the Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Zahra Arabzada
- Department of Medicinal Chemistry, College of Pharmacy and the Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Armand Bankhead
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States.,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Armita Kyani
- Department of Medicinal Chemistry, College of Pharmacy and the Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy and the Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
27
|
One-carbon metabolism in cancer cells: a critical review based on a core model of central metabolism. Biochem Soc Trans 2021; 49:1-15. [PMID: 33616629 DOI: 10.1042/bst20190008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 12/25/2022]
Abstract
One-carbon metabolism (1C-metabolism), also called folate metabolism because the carbon group is attached to folate-derived tetrahydrofolate, is crucial in metabolism. It is at the heart of several essential syntheses, particularly those of purine and thymidylate. After a short reminder of the organization of 1C-metabolism, I list its salient features as reported in the literature. Then, using flux balance analysis, a core model of central metabolism and the flux constraints for an 'average cancer cell metabolism', I explore the fundamentals underlying 1C-metabolism and its relationships with the rest of metabolism. Some unreported properties of 1C-metabolism emerge, such as its potential roles in mitochondrial NADH exchange with cytosolic NADPH, participation in NADH recycling, and optimization of cell proliferation.
Collapse
|
28
|
Pan S, Fan M, Liu Z, Li X, Wang H. Serine, glycine and one‑carbon metabolism in cancer (Review). Int J Oncol 2021; 58:158-170. [PMID: 33491748 PMCID: PMC7864012 DOI: 10.3892/ijo.2020.5158] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Serine/glycine biosynthesis and one‑carbon metabolism are crucial in sustaining cancer cell survival and rapid proliferation, and of high clinical relevance. Excessive activation of serine/glycine biosynthesis drives tumorigenesis and provides a single carbon unit for one‑carbon metabolism. One‑carbon metabolism, which is a complex cyclic metabolic network based on the chemical reaction of folate compounds, provides the necessary proteins, nucleic acids, lipids and other biological macromolecules to support tumor growth. Moreover, one‑carbon metabolism also maintains the redox homeostasis of the tumor microenvironment and provides substrates for the methylation reaction. The present study reviews the role of key enzymes with tumor‑promoting functions and important intermediates that are physiologically relevant to tumorigenesis in serine/glycine/one‑carbon metabolism pathways. The related regulatory mechanisms of action of the key enzymes and important intermediates in tumors are also discussed. It is hoped that investigations into these pathways will provide new translational opportunities for human cancer drug development, dietary interventions, and biomarker identification.
Collapse
Affiliation(s)
- Sijing Pan
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, Institute of Translational Medicine, School of Basic Medicine, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Ming Fan
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, Institute of Translational Medicine, School of Basic Medicine, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Zhangnan Liu
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, Institute of Translational Medicine, School of Basic Medicine, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Xia Li
- Correspondence to: Dr Huijuan Wang or Dr Xia Li, Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, Institute of Translational Medicine, School of Basic Medicine, Henan University, Jinming Road, Kaifeng, Henan 475004, P.R. China, E-mail: , E-mail:
| | - Huijuan Wang
- Correspondence to: Dr Huijuan Wang or Dr Xia Li, Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, Institute of Translational Medicine, School of Basic Medicine, Henan University, Jinming Road, Kaifeng, Henan 475004, P.R. China, E-mail: , E-mail:
| |
Collapse
|
29
|
Lim EW, Parker SJ, Metallo CM. Deuterium Tracing to Interrogate Compartment-Specific NAD(P)H Metabolism in Cultured Mammalian Cells. Methods Mol Biol 2020; 2088:51-71. [PMID: 31893370 DOI: 10.1007/978-1-0716-0159-4_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Oxidation-reduction (redox) reactions are ubiquitous in biology and typically occur in specific subcellular compartments. In cells, the electron transfer between molecules and organelles is commonly facilitated by pyridine nucleotides such as nicotinamide adenine dinucleotide phosphate (NADPH) and nicotinamide adenine dinucleotide (NADH). While often taken for granted, these metabolic reactions are critically important for maintaining redox homeostasis and biochemical potentials across membranes. While 13C tracing and metabolic flux analysis (MFA) have emerged as powerful tools to study intracellular metabolism, this approach is limited when applied to pathways catalyzed in multiple cellular compartments. To address this issue, we and others have applied 2H (deuterium) tracers to observe transfer of labeled hydride anions, which accompanies electron transfer. Furthermore, we have developed a reporter system for indirectly quantifying NADPH enrichment in specific subcellular compartments. Here, we provide a detailed description of 2H tracing applications and the interrogation of mitochondrial versus cytosolic NAD(P)H metabolism in cultured mammalian cells. Specifically, we describe the generation of reporter cell lines that express epitope-tagged R132H-IDH1 or R172K-IDH2 and produce (D)2-hydroxyglutarate in a doxycycline-dependent manner. These tools and methods allow for quantitation of reducing equivalent turnover rates, the directionality of pathways present in multiple compartments, and the estimation of pathway contributions to NADPH pools.
Collapse
Affiliation(s)
- Esther W Lim
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Seth J Parker
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Christian M Metallo
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
- Diabetes and Endocrinology Research Center, University of California San Diego, La Jolla, CA, USA.
- Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
30
|
Shukla K, Singh N, Lewis JE, Tsang AW, Boothman DA, Kemp ML, Furdui CM. MTHFD2 Blockade Enhances the Efficacy of β-Lapachone Chemotherapy With Ionizing Radiation in Head and Neck Squamous Cell Cancer. Front Oncol 2020; 10:536377. [PMID: 33262939 PMCID: PMC7685994 DOI: 10.3389/fonc.2020.536377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
Head and Neck Squamous Cell Cancer (HNSCC) presents with multiple treatment challenges limiting overall survival rates and affecting patients' quality of life. Amongst these, resistance to radiation therapy constitutes a major clinical problem in HNSCC patients compounded by origin, location, and tumor grade that limit tumor control. While cisplatin is considered the standard radiosensitizing agent for definitive or adjuvant radiotherapy, in recurrent tumors or for palliative care other chemotherapeutics such as the antifolates methotrexate or pemetrexed are also being utilized as radiosensitizers. These drugs inhibit the enzyme dihydrofolate reductase, which is essential for DNA synthesis and connects the 1-C/folate metabolism to NAD(P)H and NAD(P)+ balance in cells. In previous studies, we identified MTHFD2, a mitochondrial enzyme involved in folate metabolism, as a key contributor to NAD(P)H levels in the radiation-resistant cells and HNSCC tumors. In the study presented here, we investigated the role of MTHFD2 in the response to radiation alone and in combination with β-lapachone, a NQO1 bioactivatable drug, which generates reactive oxygen species concomitant with NAD(P)H oxidation to NAD(P)+. These studies are performed in a matched HNSCC cell model of response to radiation: the radiation resistant rSCC-61 and radiation sensitive SCC-61 cells reported earlier by our group. Radiation resistant rSCC-61 cells had increased sensitivity to β-lapachone compared to SCC-61 and knockdown of MTHFD2 in rSCC-61 cells further potentiated the cytotoxicity of β-lapachone with radiation in a dose and time-dependent manner. rSCC-61 MTHFD2 knockdown cells irradiated and treated with β-lapachone showed increased PARP1 activation, inhibition of mitochondrial respiration, decreased respiration-linked ATP production, and increased mitochondrial superoxide and protein oxidation as compared to control rSCC-61 scrambled shRNA. Thus, these studies point to MTHFD2 as a potential target for development of radiosensitizing chemotherapeutics and potentiator of β-lapachone cytotoxicity.
Collapse
Affiliation(s)
- Kirtikar Shukla
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Naveen Singh
- Department of Biochemistry and Molecular Biology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Joshua E. Lewis
- The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, United States
| | - Allen W. Tsang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - David A. Boothman
- Department of Biochemistry and Molecular Biology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Melissa L. Kemp
- The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, United States
| | - Cristina M. Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States,*Correspondence: Cristina M. Furdui
| |
Collapse
|
31
|
Wang J, Luo J, Sun Z, Sun F, Kong Z, Yu J. Identification of MTHFD2 as a novel prognosis biomarker in esophageal carcinoma patients based on transcriptomic data and methylation profiling. Medicine (Baltimore) 2020; 99:e22194. [PMID: 32925794 PMCID: PMC7489726 DOI: 10.1097/md.0000000000022194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
DNA methylation is an important epigenetic regulatory mechanism in esophageal carcinoma (EC) and is associated with genomic instability and carcinogenesis. In the present study, we aimed to identify tumor biomarkers for predicting prognosis of EC patients.We downloaded mRNA expression profiles and DNA methylation profiles associated with EC from the Gene Expression Omnibus database. Differentially expressed and differentially methylated genes between tumor tissues and adjacent normal tissue samples were identified. Functional enrichment analyses were performed, followed by the construction of protein-protein interaction networks. Data were validated based on methylation profiles from The Cancer Genome Atlas. Candidate genes were further verified according to survival analysis and Cox regression analysis.We uncovered multiple genes with differential expression or methylation in tumor samples compared with normal samples. After taking the intersection of 3 differential gene sets, we obtained a total of 232 overlapping genes. Functional enrichment analysis revealed that these genes are related to pathways such as "glutathione metabolism," "p53 signaling pathway," and "focal adhesion." Furthermore, 8 hub genes with inversed expression and methylation correlation were identified as candidate genes. The abnormal expression levels of MSN, PELI1, and MTHFD2 were correlated with overall survival times in EC patients (P < .05). Only MTHFD2 was significantly associated with a pathologic stage according to univariate analysis (P = .037) and multivariate analysis (P = .043).Our study identified several novel EC biomarkers with prognostic value by integrated analysis of transcriptomic data and methylation profiles. MTHFD2 could serve as an independent biomarker for predicting prognosis and pathological stages of EC.
Collapse
Affiliation(s)
- Jianlin Wang
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
- Center for Medical Physics, Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Judong Luo
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
| | - Zhiqiang Sun
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
| | - Fei Sun
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
| | - Ze Kong
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
| | - Jingping Yu
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University
- Center for Medical Physics, Nanjing Medical University, Changzhou, Jiangsu Province, China
| |
Collapse
|
32
|
Benzarti M, Delbrouck C, Neises L, Kiweler N, Meiser J. Metabolic Potential of Cancer Cells in Context of the Metastatic Cascade. Cells 2020; 9:E2035. [PMID: 32899554 PMCID: PMC7563895 DOI: 10.3390/cells9092035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
The metastatic cascade is a highly plastic and dynamic process dominated by cellular heterogeneity and varying metabolic requirements. During this cascade, the three major metabolic pillars, namely biosynthesis, RedOx balance, and bioenergetics, have variable importance. Biosynthesis has superior significance during the proliferation-dominated steps of primary tumour growth and secondary macrometastasis formation and only minor relevance during the growth-independent processes of invasion and dissemination. Consequently, RedOx homeostasis and bioenergetics emerge as conceivable metabolic key determinants in cancer cells that disseminate from the primary tumour. Within this review, we summarise our current understanding on how cancer cells adjust their metabolism in the context of different microenvironments along the metastatic cascade. With the example of one-carbon metabolism, we establish a conceptual view on how the same metabolic pathway can be exploited in different ways depending on the current cellular needs during metastatic progression.
Collapse
Affiliation(s)
- Mohaned Benzarti
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2 Avenue de l’Université, L-4365 Esch-sur-Alzette, Luxembourg
| | - Catherine Delbrouck
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2 Avenue de l’Université, L-4365 Esch-sur-Alzette, Luxembourg
| | - Laura Neises
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
| | - Nicole Kiweler
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
| | - Johannes Meiser
- Cancer Metabolism Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg; (M.B.); (C.D.); (L.N.); (N.K.)
| |
Collapse
|
33
|
Dekhne AS, Hou Z, Gangjee A, Matherly LH. Therapeutic Targeting of Mitochondrial One-Carbon Metabolism in Cancer. Mol Cancer Ther 2020; 19:2245-2255. [PMID: 32879053 DOI: 10.1158/1535-7163.mct-20-0423] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/06/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
One-carbon (1C) metabolism encompasses folate-mediated 1C transfer reactions and related processes, including nucleotide and amino acid biosynthesis, antioxidant regeneration, and epigenetic regulation. 1C pathways are compartmentalized in the cytosol, mitochondria, and nucleus. 1C metabolism in the cytosol has been an important therapeutic target for cancer since the inception of modern chemotherapy, and "antifolates" targeting cytosolic 1C pathways continue to be a mainstay of the chemotherapy armamentarium for cancer. Recent insights into the complexities of 1C metabolism in cancer cells, including the critical role of the mitochondrial 1C pathway as a source of 1C units, glycine, reducing equivalents, and ATP, have spurred the discovery of novel compounds that target these reactions, with particular focus on 5,10-methylene tetrahydrofolate dehydrogenase 2 and serine hydroxymethyltransferase 2. In this review, we discuss key aspects of 1C metabolism, with emphasis on the importance of mitochondrial 1C metabolism to metabolic homeostasis, its relationship with the oncogenic phenotype, and its therapeutic potential for cancer.
Collapse
Affiliation(s)
- Aamod S Dekhne
- Department of Oncology, Wayne State University School of Medicine, and the Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Zhanjun Hou
- Department of Oncology, Wayne State University School of Medicine, and the Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Aleem Gangjee
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania
| | - Larry H Matherly
- Department of Oncology, Wayne State University School of Medicine, and the Barbara Ann Karmanos Cancer Institute, Detroit, Michigan.
| |
Collapse
|
34
|
Reina-Campos M, Diaz-Meco MT, Moscat J. The complexity of the serine glycine one-carbon pathway in cancer. J Cell Biol 2020; 219:jcb.201907022. [PMID: 31690618 PMCID: PMC7039202 DOI: 10.1083/jcb.201907022] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/09/2019] [Accepted: 09/19/2019] [Indexed: 12/21/2022] Open
Abstract
Perturbations in cellular metabolism are ubiquitous in cancer. Here Reina-Campos et al. review the role of one-carbon metabolism in tumorigenesis. The serine glycine and one-carbon pathway (SGOCP) is a crucially important metabolic network for tumorigenesis, of unanticipated complexity, and with implications in the clinic. Solving how this network is regulated is key to understanding the underlying mechanisms of tumor heterogeneity and therapy resistance. Here, we review its role in cancer by focusing on key enzymes with tumor-promoting functions and important products of the SGOCP that are of physiological relevance for tumorigenesis. We discuss the regulatory mechanisms that coordinate the metabolic flux through the SGOCP and their deregulation, as well as how the actions of this metabolic network affect other cells in the tumor microenvironment, including endothelial and immune cells.
Collapse
Affiliation(s)
- Miguel Reina-Campos
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Maria T Diaz-Meco
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Jorge Moscat
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| |
Collapse
|
35
|
Li AM, Ye J. Reprogramming of serine, glycine and one-carbon metabolism in cancer. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165841. [PMID: 32439610 DOI: 10.1016/j.bbadis.2020.165841] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 04/28/2020] [Accepted: 05/10/2020] [Indexed: 02/06/2023]
Abstract
Metabolic pathways leading to the synthesis, uptake, and usage of the nonessential amino acid serine are frequently amplified in cancer. Serine encounters diverse fates in cancer cells, including being charged onto tRNAs for protein synthesis, providing head groups for sphingolipid and phospholipid synthesis, and serving as a precursor for cellular glycine and one-carbon units, which are necessary for nucleotide synthesis and methionine cycle reloading. This review will focus on the participation of serine and glycine in the mitochondrial one-carbon (SGOC) pathway during cancer progression, with an emphasis on the genetic and epigenetic determinants that drive SGOC gene expression. We will discuss recently elucidated roles for SGOC metabolism in nucleotide synthesis, redox balance, mitochondrial function, and epigenetic modifications. Finally, therapeutic considerations for targeting SGOC metabolism in the clinic will be discussed.
Collapse
Affiliation(s)
- Albert M Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Cancer Biology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Cancer Biology Program, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
36
|
Zhu Z, Leung GKK. More Than a Metabolic Enzyme: MTHFD2 as a Novel Target for Anticancer Therapy? Front Oncol 2020; 10:658. [PMID: 32411609 PMCID: PMC7199629 DOI: 10.3389/fonc.2020.00658] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/08/2020] [Indexed: 01/22/2023] Open
Abstract
The bifunctional methylenetetrahydrofolate dehydrogenase/cyclohydrolase (MTHFD2) is a mitochondrial one-carbon folate metabolic enzyme whose role in cancer was not known until recently. MTHFD2 is highly expressed in embryos and a wide range of tumors but has low or absent expression in most adult differentiated tissues. Elevated MTHFD2 expression is associated with poor prognosis in both hematological and solid malignancy. Its depletion leads to suppression of multiple malignant phenotypes including proliferation, invasion, migration, and induction of cancer cell death. The non-metabolic functions of this enzyme, especially in cancers, have thus generated considerable research interests. This review summarizes current knowledge on both the metabolic functions and non-enzymatic roles of MTHFD2. Its expression, potential functions, and regulatory mechanism in cancers are highlighted. The development of MTHFD2 inhibitors and their implications in pre-clinical models are also discussed.
Collapse
Affiliation(s)
- Zhiyuan Zhu
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Gilberto Ka Kit Leung
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
37
|
Abstract
In this issue of Cell Metabolism, Yang et al., 2020 report that serine is a source of mitochondrial NADH derived from one-carbon metabolism. Serine becomes a major source of NADH when cellular respiration is inhibited, and the un-utilized, accumulated NADH inhibits the TCA cycle and slows proliferation.
Collapse
Affiliation(s)
- Adam G Maynard
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Naama Kanarek
- Department of Pathology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, 415 Main Street, Cambridge, MA 02142, USA.
| |
Collapse
|
38
|
Abstract
BACKGROUND Formate is a one-carbon molecule at the crossroad between cellular and whole body metabolism, between host and microbiome metabolism, and between nutrition and toxicology. This centrality confers formate with a key role in human physiology and disease that is currently unappreciated. SCOPE OF REVIEW Here we review the scientific literature on formate metabolism, highlighting cellular pathways, whole body metabolism, and interactions with the diet and the gut microbiome. We will discuss the relevance of formate metabolism in the context of embryonic development, cancer, obesity, immunometabolism, and neurodegeneration. MAJOR CONCLUSIONS We will conclude with an outlook of some open questions bringing formate metabolism into the spotlight.
Collapse
Affiliation(s)
| | - Johannes Meiser
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Alexei Vazquez
- Cancer Research UK Beatson Institute, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
39
|
Pramono AA, Rather GM, Herman H, Lestari K, Bertino JR. NAD- and NADPH-Contributing Enzymes as Therapeutic Targets in Cancer: An Overview. Biomolecules 2020; 10:biom10030358. [PMID: 32111066 PMCID: PMC7175141 DOI: 10.3390/biom10030358] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/14/2022] Open
Abstract
Actively proliferating cancer cells require sufficient amount of NADH and NADPH for biogenesis and to protect cells from the detrimental effect of reactive oxygen species. As both normal and cancer cells share the same NAD biosynthetic and metabolic pathways, selectively lowering levels of NAD(H) and NADPH would be a promising strategy for cancer treatment. Targeting nicotinamide phosphoribosyltransferase (NAMPT), a rate limiting enzyme of the NAD salvage pathway, affects the NAD and NADPH pool. Similarly, lowering NADPH by mutant isocitrate dehydrogenase 1/2 (IDH1/2) which produces D-2-hydroxyglutarate (D-2HG), an oncometabolite that downregulates nicotinate phosphoribosyltransferase (NAPRT) via hypermethylation on the promoter region, results in epigenetic regulation. NADPH is used to generate D-2HG, and is also needed to protect dihydrofolate reductase, the target for methotrexate, from degradation. NAD and NADPH pools in various cancer types are regulated by several metabolic enzymes, including methylenetetrahydrofolate dehydrogenase, serine hydroxymethyltransferase, and aldehyde dehydrogenase. Thus, targeting NAD and NADPH synthesis under special circumstances is a novel approach to treat some cancers. This article provides the rationale for targeting the key enzymes that maintain the NAD/NADPH pool, and reviews preclinical studies of targeting these enzymes in cancers.
Collapse
Affiliation(s)
- Alvinsyah Adhityo Pramono
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; (A.A.P.); (G.M.R.)
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Gulam M. Rather
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; (A.A.P.); (G.M.R.)
| | - Herry Herman
- Division of Oncology, Department of Orthopaedic Surgery, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia;
| | - Keri Lestari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Joseph R. Bertino
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; (A.A.P.); (G.M.R.)
- Department of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Correspondence: ; Tel.: +1-(732)-235-8510
| |
Collapse
|
40
|
Kushwaha PP, Gupta S, Singh AK, Prajapati KS, Shuaib M, Kumar S. MicroRNA Targeting Nicotinamide Adenine Dinucleotide Phosphate Oxidases in Cancer. Antioxid Redox Signal 2020; 32:267-284. [PMID: 31656079 DOI: 10.1089/ars.2019.7918] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Significance: Reactive oxygen species (ROS) production occurs primarily in the mitochondria as a by-product of cellular metabolism. ROS are also produced by nicotinamide adenine dinucleotide phosphate (NADPH) oxidases in response to growth factors and cytokines by normal physiological signaling pathways. NADPH oxidase, a member of NADPH oxidase (NOX) family, utilizes molecular oxygen (O2) to generate ROS such as hydrogen peroxide and superoxide. Imbalance between ROS production and its elimination is known to be the major cause of various human diseases. NOX family proteins are exclusively involved in ROS production, which makes them attractive target(s) for the treatment of ROS-mediated diseases including cancer. Recent Advances: Molecules such as Keap1/nuclear factor erythroid 2-related factor 2 (Nrf2), N-methyl-d-aspartic acid (NMDA) receptors, nuclear factor-kappaB, KRAS, kallistatin, gene associated with retinoic-interferon-induced mortality-19, and deregulated metabolic pathways are involved in ROS production in association with NADPH oxidase. Critical Issues: Therapeutic strategies targeting NADPH oxidases in ROS-driven cancers are not very effective due to its complex regulatory circuit. Tumor suppressor microRNAs (miRNAs) viz. miR-34a, miR-137, miR-99a, and miR-21a-3p targeting NADPH oxidases are predominantly downregulated in ROS-driven cancers. miRNAs also regulate other cellular machineries such as Keap1/Nrf2 pathway and NMDA receptors involved in ROS production and consequently drug resistance. Here, we discuss the structure, function, and metabolic role of NADPH oxidase, NOX family protein-protein interaction, their association with other pathways, and NADPH oxidase alteration by miRNAs. Moreover, we also discuss and summarize studies on NADPH oxidase associated with various malignancies and their therapeutic implications. Future Directions: Targeting NADPH oxidases through miRNAs appears to be a promising strategy for the treatment of ROS-driven cancer.
Collapse
Affiliation(s)
- Prem Prakash Kushwaha
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Sanjay Gupta
- The James and Eilleen Dicke Laboratory, Department of Urology, Case Western Reserve University, Cleveland, Ohio
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio
- Divison of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio
| | - Atul Kumar Singh
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Kumari Sunita Prajapati
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Mohd Shuaib
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Shashank Kumar
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW The aim of this report is to examine critical relationships between amino acid and formate metabolism with particular reference to the production of formate, and to review novel functions of formate. RECENT FINDINGS In addition to well established mechanisms in one-carbon metabolism, formate may play an important role in early pregnancy by preventing the onset of neural tube defects in sensitive strains of mice, including mice with deficiencies in MTHFD1L, the glycine cleavage system and the mitochondrial folate transporter. Markedly elevated, circulating levels of formate are found in late pregnancy, including in cord blood, as well as elevated levels of amino acid precursors. These are consistent with specific roles for formate in late pregnancy. Serine metabolism may reduce NADP to NADPH and permit the use of NADPH in reductive reactions. Novel, noncanonical functions of formate include high rates of formate production from serine in cells and in cancers. SUMMARY Novel, noncanonical functions of formate continue to be discovered. Integrating their functions with well established elements of one-carbon metabolism remains an important future objective.
Collapse
Affiliation(s)
- John T Brosnan
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | | |
Collapse
|
42
|
Wang LW, Shen H, Nobre L, Ersing I, Paulo JA, Trudeau S, Wang Z, Smith NA, Ma Y, Reinstadler B, Nomburg J, Sommermann T, Cahir-McFarland E, Gygi SP, Mootha VK, Weekes MP, Gewurz BE. Epstein-Barr-Virus-Induced One-Carbon Metabolism Drives B Cell Transformation. Cell Metab 2019; 30:539-555.e11. [PMID: 31257153 PMCID: PMC6720460 DOI: 10.1016/j.cmet.2019.06.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 03/14/2019] [Accepted: 06/05/2019] [Indexed: 02/05/2023]
Abstract
Epstein-Barr virus (EBV) causes Burkitt, Hodgkin, and post-transplant B cell lymphomas. How EBV remodels metabolic pathways to support rapid B cell outgrowth remains largely unknown. To gain insights, primary human B cells were profiled by tandem-mass-tag-based proteomics at rest and at nine time points after infection; >8,000 host and 29 viral proteins were quantified, revealing mitochondrial remodeling and induction of one-carbon (1C) metabolism. EBV-encoded EBNA2 and its target MYC were required for upregulation of the central mitochondrial 1C enzyme MTHFD2, which played key roles in EBV-driven B cell growth and survival. MTHFD2 was critical for maintaining elevated NADPH levels in infected cells, and oxidation of mitochondrial NADPH diminished B cell proliferation. Tracing studies underscored contributions of 1C to nucleotide synthesis, NADPH production, and redox defense. EBV upregulated import and synthesis of serine to augment 1C flux. Our results highlight EBV-induced 1C as a potential therapeutic target and provide a new paradigm for viral onco-metabolism.
Collapse
Affiliation(s)
- Liang Wei Wang
- Graduate Program in Virology, Division of Medical Sciences, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Hongying Shen
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Luis Nobre
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Ina Ersing
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen Trudeau
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Zhonghao Wang
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Nicholas A Smith
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Yijie Ma
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Bryn Reinstadler
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Jason Nomburg
- Graduate Program in Virology, Division of Medical Sciences, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Thomas Sommermann
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Ellen Cahir-McFarland
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Vamsi K Mootha
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK.
| | - Benjamin E Gewurz
- Graduate Program in Virology, Division of Medical Sciences, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Wei Y, Liu P, Li Q, Du J, Chen Y, Wang Y, Shi H, Wang Y, Zhang H, Xue W, Gao Y, Li D, Feng Y, Yan J, Han J, Zhang J. The effect of MTHFD2 on the proliferation and migration of colorectal cancer cell lines. Onco Targets Ther 2019; 12:6361-6370. [PMID: 31496738 PMCID: PMC6697661 DOI: 10.2147/ott.s210800] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/22/2019] [Indexed: 01/11/2023] Open
Abstract
Purpose Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) is a tetramethylfolate dehydrogenase enzyme involved in folate metabolism. The aim of this study is to determine the effect of MTHFD2 on the proliferation and metastasis of colorectal cancer (CRC). Patients and methods MTHFD2 was silenced or overexpressed in CRC cells. qRT-PCR and Western blotting were used to analyze mRNA and protein expression, respectively. The MTT assay and colony forming assay were used to detect cell proliferation and colony formation ability. The cycle and apoptosis changes were detected by flow cytometry. Transwell experiments were used to analyze the migration ability of CRC cells. Results The expression of MTHFD2 in 31 kinds of cancers was analyzed by bioinformatics, and MTHFD2 was found highly expressed in various cancer cells including CRC cells. Silencing the expression of MTHFD2 resulted in inhibition of the proliferation of CRC cells, weakening of the migration ability, blocking of the cell cycle in G0/G1-S phase, and promotion of the apoptosis of CRC cells. On the contrary, overexpression of MTHFD2 in CRC cells resulted in enhancement of the proliferation and migration ability, promotion of cell cycle progression and inhibition of cell apoptosis. Conclusion MTHFD2 is positively related with colorectal cancer and the MTHFD2 gene is a tumor promoting gene in CRC cells.
Collapse
Affiliation(s)
- Yameng Wei
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Pengfei Liu
- Department of Gastroenterology, Yan'an University Affiliated Hospital, Yan'an 716000, People's Republic of China
| | - Qian Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, People's Republic of China
| | - Juan Du
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Yani Chen
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Yu Wang
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Haiyan Shi
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Yanfeng Wang
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Huahua Zhang
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Wanjuan Xue
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Yi Gao
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Dan Li
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Yun Feng
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Jing Yan
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Jiming Han
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| | - Jing Zhang
- Department of Clinical Medicine, Medical College of Yan'an University, Yan'an 716000, People's Republic of China
| |
Collapse
|
44
|
Nilsson R, Nicolaidou V, Koufaris C. Mitochondrial MTHFD isozymes display distinct expression, regulation, and association with cancer. Gene 2019; 716:144032. [PMID: 31377316 DOI: 10.1016/j.gene.2019.144032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023]
Abstract
Mitochondrial folate metabolism is central to the generation of nucleotides, fuelling methylation reactions, and redox homeostasis. Uniquely among the reactions of the mitochondrial folate pathway, the key step of the oxidation of 5,10-methylene-tetrahydrofolate (CH2-THF) can be catalysed by two isozymes, MTHFD2 and MTHFD2L. The MTHFD2 enzyme has recently received considerable attention as an oncogenic enzyme upregulated in several tumour types, which is additionally required by cancer cells in vitro and in vivo. However, much less is currently known about MTHFD2L and its expression in cancer. In this study, we examine and compare the expression and regulation of the two mitochondrial MTHFD isozymes in normal human and cancer cells. We found that normal and cancer cells express both enzymes, although MTHFD2 has a much higher baseline expression. Unlike MTHFD2, the MTHFD2L isozyme does not show an association with proliferation and growth factor stimulation. In addition, we did not find evidence of a compensatory increase of MTHFD2L following suppression of its isozyme. This study supports that MTHFD2L is unlikely to have an important function in increased proliferation or cancer. Furthermore, therapeutic strategies aiming to block the mitochondrial folate pathway in cancer should focus on MTHFD2, with MTHFD2L being unlikely to be involved in the development of chemoresistance to targeting of its mitochondrial isozyme.
Collapse
Affiliation(s)
- R Nilsson
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; Division of Cardiovascular Medicine, Karolinska University Hospital, SE-171 76 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - V Nicolaidou
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | - C Koufaris
- Department of Biological Sciences, University of Cyprus, 2109 Nicosia, Cyprus.
| |
Collapse
|
45
|
Gerner MC, Niederstaetter L, Ziegler L, Bileck A, Slany A, Janker L, Schmidt RLJ, Gerner C, Del Favero G, Schmetterer KG. Proteome Analysis Reveals Distinct Mitochondrial Functions Linked to Interferon Response Patterns in Activated CD4+ and CD8+ T Cells. Front Pharmacol 2019; 10:727. [PMID: 31354474 PMCID: PMC6635586 DOI: 10.3389/fphar.2019.00727] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 06/05/2019] [Indexed: 01/08/2023] Open
Abstract
While genetic traits and epigenetic modifications mainly encode cell type-specific effector functions, the eventual outcome is also prone to modulation by post-transcriptional regulation mechanisms. T cells are a powerful model for the investigation of such modulatory effects, as common precursor cells may differentiate either to helper CD4+ T cells or cytotoxic CD8+ cells, which elicit distinct functionalities upon TCR-stimulation. Human primary CD4+ and CD8+ T cells were purified from three individual donors and activated with anti-CD3/CD28 antibodies. Associated proteome alterations were analyzed by high-resolution mass spectrometry using a label-free shotgun approach. Metabolic activation was indicated by upregulation of enzymes related to glycolysis, NADH production, fatty acid synthesis, and uptake as well as amino acid and iron uptake. Besides various inflammatory effector molecules, the mitochondrial proteins CLUH, TFAM, and TOMM34 were found specifically induced in CD4+ T cells. Investigation of overrepresented conserved transcription binding sites by the oPOSSUM software suggested interferon type I inducer IRF1 to cause many of the observed proteome alterations in CD4+ T cells. RT qPCR demonstrated the specific induction of IRF1 in CD4+ T cells only. While the interferon regulatory factor IRF4 was found induced in both T cell subtypes at protein and mRNA level, IRF9 and the type I interferon-induced proteins IFIT1, IFIT3, and MX1 were only found induced in CD4+ T cells. As oxidative stress enhances mitochondrial DNA-dependent type I interferon responses, the present data suggested that mitochondrial activities regulate those cell type-specific signaling pathways. Indeed, we detected mitochondrial superoxide formation predominantly in CD4+ T cells via FACS analysis with MitoSOX™ and confirmed this observation by live cell imaging with confocal microscopy. As interferon signaling regulates important features such as resistance regarding immune checkpoint blockade therapy, the present data may identify potential new targets for the efficient control of highly relevant immune cell properties.
Collapse
Affiliation(s)
- Marlene C Gerner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Laura Niederstaetter
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Liesa Ziegler
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Astrid Slany
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Ralf L J Schmidt
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria.,Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Klaus G Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
46
|
Haque MR, Higashiura A, Nakagawa A, Hirowatari A, Furuya S, Yamamoto K. Molecular structure of a 5,10-methylenetetrahydrofolate dehydrogenase from the silkworm Bombyx mori. FEBS Open Bio 2019; 9:618-628. [PMID: 30984537 PMCID: PMC6443876 DOI: 10.1002/2211-5463.12595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 12/27/2018] [Accepted: 01/14/2019] [Indexed: 11/12/2022] Open
Abstract
The enzyme 5,10‐methylenetetrahydrofolate dehydrogenase (MTHFD) is essential for the production of certain amino acids (glycine, serine, and methionine) and nucleic acids (thymidylate and purine). Here, we identified a cDNA encoding this enzyme from the silkworm Bombyx mori. The recombinant B. mori MTHFD (bmMTHFD) expressed in Escherichia coli recognized 5,10‐methylenetetrahydrofolate and 5,10‐methenyltetrahydrofolate as substrate in the presence of NADP+ as well as NAD+. The bmMTHFD structure was determined at a resolution of 1.75 Å by X‐ray crystallography. Site‐directed mutagenesis indicated that the amino acid residue Tyr49 contributed to its catalytic activity. Our findings provide insight into the mechanism underlying the activity of MTHFD from B. mori and potentially other insects and may therefore facilitate the development of inhibitors specific to MTHFD as insecticides.
Collapse
Affiliation(s)
- Mohammad R Haque
- Department of Bioscience and Biotechnology Kyushu University Graduate School Fukuoka Japan
| | - Akifumi Higashiura
- Institute for Protein Research Osaka University Suita Japan.,Present address: Department of Virology Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | | | - Aiko Hirowatari
- Department of Bioscience and Biotechnology Kyushu University Graduate School Fukuoka Japan
| | - Shigeki Furuya
- Department of Bioscience and Biotechnology Kyushu University Graduate School Fukuoka Japan
| | - Kohji Yamamoto
- Department of Bioscience and Biotechnology Kyushu University Graduate School Fukuoka Japan
| |
Collapse
|
47
|
Zhu Y, Dean AE, Horikoshi N, Heer C, Spitz DR, Gius D. Emerging evidence for targeting mitochondrial metabolic dysfunction in cancer therapy. J Clin Invest 2018; 128:3682-3691. [PMID: 30168803 DOI: 10.1172/jci120844] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mammalian cells use a complex network of redox-dependent processes necessary to maintain cellular integrity during oxidative metabolism, as well as to protect against and/or adapt to stress. The disruption of these redox-dependent processes, including those in the mitochondria, creates a cellular environment permissive for progression to a malignant phenotype and the development of resistance to commonly used anticancer agents. An extension of this paradigm is that when these mitochondrial functions are altered by the events leading to transformation and ensuing downstream metabolic processes, they can be used as molecular biomarkers or targets in the development of new therapeutic interventions to selectively kill and/or sensitize cancer versus normal cells. In this Review we propose that mitochondrial oxidative metabolism is altered in tumor cells, and the central theme of this dysregulation is electron transport chain activity, folate metabolism, NADH/NADPH metabolism, thiol-mediated detoxification pathways, and redox-active metal ion metabolism. It is proposed that specific subgroups of human malignancies display distinct mitochondrial transformative and/or tumor signatures that may benefit from agents that target these pathways.
Collapse
Affiliation(s)
- Yueming Zhu
- Department of Radiation Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Angela Elizabeth Dean
- Department of Radiation Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Nobuo Horikoshi
- Department of Radiation Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Collin Heer
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - David Gius
- Department of Radiation Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|