51
|
Keenan T, Spears RJ, Akkad S, Mahon CS, Hatton NE, Walton J, Noble A, Yates ND, Baumann CG, Parkin A, Signoret N, Fascione MA. A Tale of Two Bioconjugations: pH Controlled Divergent Reactivity of Protein α-oxo-Aldehydes in Competing α-oxo-Mannich and Catalyst-Free Aldol Ligations. ACS Chem Biol 2021; 16:2387-2400. [PMID: 34751550 DOI: 10.1021/acschembio.1c00531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Site-selective chemical methods for protein bioconjugation have revolutionized the fields of cell and chemical biology through the development of novel protein/enzyme probes bearing fluorescent, spectroscopic, or even toxic cargos. Herein, we report two new methods for the bioconjugation of α-oxo aldehyde handles within proteins using small molecule aniline and/or phenol probes. The "α-oxo-Mannich" and "catalyst-free aldol" ligations both compete for the electrophilic α-oxo aldehyde, which displays pH divergent reactivity proceeding through the "Mannich" pathway at acidic pH to afford bifunctionalized bioconjugates, and the "catalyst-free aldol" pathway at neutral pH to afford monofunctionalized bioconjugates. We explore the substrate scope and utility of both of these bioconjugations in the construction of neoglycoproteins, in the process formulating a mechanistic rationale for how both pathways intersect with each other at different reaction pH's.
Collapse
Affiliation(s)
- Tessa Keenan
- Department of Chemistry, University of York, York, United Kingdom YO10 5DD
| | - Richard J. Spears
- Department of Chemistry, University of York, York, United Kingdom YO10 5DD
| | - Saeed Akkad
- Department of Chemistry, University of York, York, United Kingdom YO10 5DD
| | - Clare S. Mahon
- Department of Chemistry, Durham University, Durham, United Kingdom DH1 3LE
| | - Natasha E. Hatton
- Department of Chemistry, University of York, York, United Kingdom YO10 5DD
| | - Julia Walton
- Department of Chemistry, University of York, York, United Kingdom YO10 5DD
| | - Amanda Noble
- Department of Chemistry, University of York, York, United Kingdom YO10 5DD
| | - Nicholas D. Yates
- Department of Chemistry, University of York, York, United Kingdom YO10 5DD
| | | | - Alison Parkin
- Department of Chemistry, University of York, York, United Kingdom YO10 5DD
| | - Nathalie Signoret
- Hull York Medical School, University of York, York, United Kingdom YO10 5DD
| | - Martin A. Fascione
- Department of Chemistry, University of York, York, United Kingdom YO10 5DD
| |
Collapse
|
52
|
Inaishi T, Shibata M, Ichikawa T, Kanda M, Hayashi M, Soeda I, Takeuchi D, Takano Y, Tsunoda N, Kodera Y, Kikumori T. Platelet isoform of phosphofructokinase accelerates malignant features in breast cancer. Oncol Rep 2021; 47:9. [PMID: 34751415 DOI: 10.3892/or.2021.8220] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/06/2021] [Indexed: 11/06/2022] Open
Abstract
The platelet isoform of phosphofructokinase (PFKP) is one of the key enzymes in the glycolytic pathway. PFKP is highly expressed in several cancers, and it has been reported to be involved in the progression of cancer cells. However, its oncological role in breast cancer (BC) remains unclear. The present study aimed to evaluate the function of PFKP in BC cells and its expression level in patients with BC. Firstly, the mRNA and protein expression of PFKP was evaluated in BC and non‑cancerous mammary cell lines. Polymerase chain reaction (PCR) array analysis was conducted to evaluate the correlation between PFKP and 84 cancer‑related genes. Then, PFKP knockdown was conducted using small interfering RNA, and cell proliferation, invasiveness and migration were analyzed. Furthermore, the association between PFKP mRNA expression and clinicopathological factors was investigated in 167 patients with BC. PFKP was highly expressed in estrogen receptor‑negative and human epidermal growth factor receptor 2‑negative BC cell lines. PCR array analysis demonstrated that the expression level of PFKP was significantly correlated with that of transforming growth factor‑β1 and MYC proto‑oncogene. PFKP knockdown significantly decreased the proliferation and invasiveness of MCF7, SK‑BR‑3, and MDA‑MB‑231 cells. Furthermore, cell migration was inhibited in SK‑BR‑3 and MDA‑MB‑231 cells. In the clinical specimens, patients with T2/T3/T4, lymph node metastasis, or stage II/III/IV exhibited higher expression of PFKP mRNA than patients with less severe disease. In conclusion, the present findings indicated that PFKP is involved in promoting tumor‑progressive oncological roles in BC cells across different subtypes and is considered a possible novel therapeutic target for BC.
Collapse
Affiliation(s)
- Takahiro Inaishi
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Masahiro Shibata
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Takahiro Ichikawa
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Ikumi Soeda
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Dai Takeuchi
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Yuko Takano
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Nobuyuki Tsunoda
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| | - Toyone Kikumori
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya 466‑8550, Japan
| |
Collapse
|
53
|
Parmar KM, Thummala Y, Kumar S, Singh P. Utility of FDG-PET CT scan to detect an incidental rare recurrence of carcinoma urinary bladder following radical cystectomy. BMJ Case Rep 2021; 14:e245393. [PMID: 34706917 PMCID: PMC8552151 DOI: 10.1136/bcr-2021-245393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 11/03/2022] Open
|
54
|
The Dog as a Model to Study the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:123-152. [PMID: 34664237 DOI: 10.1007/978-3-030-73119-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Cancer is a complex and dynamic disease with an outcome that depends on a strict crosstalk between tumor cells and other components in tumor microenvironment, namely, tumor-infiltrating immune cells, fibroblasts, cancer stem cells, adipocytes, and endothelial cells. Within the tumor microenvironment, macrophages and T-lymphocytes appear to be key effectors during the several steps of tumor initiation and progression. Tumor cells, through the release of a plethora of signaling molecules, can induce immune tolerance, by avoiding immune surveillance, and inhibit immune cells cytotoxic functions. Furthermore, as the tumor grows, tumor microenvironment reveals a series of dysfunctional conditions that potentiate a polarization of harmful humoral Th2 and Th17, an upregulation of Treg cells, and a differentiation of macrophages into the M2 subtype, which contribute to the activation of several signaling pathways involving important tissue biomarkers (COX-2, EGFR, VEGF) implicated in cancer aggressiveness and poor clinical outcomes. In order to maintain the tumor growth, cancer cells acquire several adaptations such as neovascularization and metabolic reprogramming. An extensive intracellular production of lactate and protons is observed in tumor cells as a result of their high glycolytic metabolism. This contributes not only for the microenvironment pH alteration but also to shape the immune response that ultimately impairs immune cells capabilities and effector functions.In this chapter, the complexity of tumor microenvironment, with special focus on macrophages, T-lymphocytes, and the impact of lactate efflux, was reviewed, always trying to demonstrate the strong similarities between data from studies of humans and dogs, a widely proposed model for comparative oncology studies.
Collapse
|
55
|
Satyanarayana G, Turaga RC, Sharma M, Wang S, Mishra F, Peng G, Deng X, Yang J, Liu ZR. Pyruvate kinase M2 regulates fibrosis development and progression by controlling glycine auxotrophy in myofibroblasts. Theranostics 2021; 11:9331-9341. [PMID: 34646373 PMCID: PMC8490528 DOI: 10.7150/thno.60385] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/29/2021] [Indexed: 12/13/2022] Open
Abstract
Rationale: Fibrosis is a pathologic condition of abnormal accumulation of collagen fibrils. Collagen is a major extracellular matrix (ECM) protein synthesized and secreted by myofibroblasts, composing mainly (Gly-X-Y)n triplet repeats with >30% Gly residue. During fibrosis progression, myofibroblasts must upregulate glycine metabolism to meet the high demands of amino acids for collagen synthesis. Method: Expression of PKM2 in myofibroblasts was analyzed in cultured fibroblasts and fibrosis disease tissues. Functional roles of PKM2 and PKM2 activator in biosynthesis of serine → glycine and production of collagen from glycolysis intermediates were assayed in cultured activated LX-2 and human primary lung fibroblast cells. Mouse models of Liver, lung, and pancreas fibrosis were employed to analyze treatment effects of PKM2 activator in organ tissue fibrosis. Results: We report here that myofibroblast differentiation upregulates pyruvate kinase M2 (PKM2) and promotes dimerization of PKM2. Dimer PKM2 slows the flow rate of glycolysis and channels glycolytic intermediates to de novo glycine synthesis, which facilitates collagen synthesis and secretion in myofibroblasts. Our results show that PKM2 activator that converts PKM2 dimer to tetramer, inhibits fibrosis progression in mouse models of liver, lung, and pancreatic fibrosis. Furthermore, metabolism alteration by dimer PKM2 increases NADPH production, which consequently protects myofibroblasts from apoptosis. Conclusion: Our study uncovers a novel role of PKM2 in tissue/organ fibrosis, suggesting a possible strategy for treatment of fibrotic diseases using PKM2 activator.
Collapse
|
56
|
Carpentieri G, Leoni C, Pietraforte D, Cecchetti S, Iorio E, Belardo A, Pietrucci D, Di Nottia M, Pajalunga D, Megiorni F, Mercurio L, Tatti M, Camero S, Marchese C, Rizza T, Tirelli V, Onesimo R, Carrozzo R, Rinalducci S, Chillemi G, Zampino G, Tartaglia M, Flex E. Hyperactive HRAS dysregulates energetic metabolism in fibroblasts from patients with Costello syndrome via enhanced production of reactive oxidizing species. Hum Mol Genet 2021; 31:561-575. [PMID: 34508588 DOI: 10.1093/hmg/ddab270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/10/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023] Open
Abstract
Germline activating mutations in HRAS cause Costello Syndrome (CS), a cancer prone multisystem disorder characterized by reduced postnatal growth. In CS, poor weight gain and growth are not caused by low caloric intake. Here we show that constitutive plasma membrane translocation and activation of the GLUT4 glucose transporter, via ROS-dependent AMPKα and p38 hyperactivation, occurs in CS, resulting in accelerated glycolysis, and increased fatty acid synthesis and storage as lipid droplets in primary fibroblasts. An accelerated autophagic flux was also identified as contributing to the increased energetic expenditure in CS. Concomitant inhibition of p38 and PI3K signaling by wortmannin was able to rescue both the dysregulated glucose intake and accelerated autophagic flux. Our findings provide a mechanistic link between upregulated HRAS function, defective growth and increased resting energetic expenditure in CS, and document that targeting p38 and PI3K signaling is able to revert this metabolic dysfunction.
Collapse
Affiliation(s)
- Giovanna Carpentieri
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy.,Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | | | - Serena Cecchetti
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Egidio Iorio
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Antonio Belardo
- Department of Ecological and Biological Sciences, Università della Tuscia, 01100 Viterbo, Italy
| | - Daniele Pietrucci
- Department for Innovation in Biological, Agro-food and Forest systems, Università della Tuscia, 01100 Viterbo, Italy
| | - Michela Di Nottia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Deborah Pajalunga
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy
| | - Laura Mercurio
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata, IRCCS, 00167 Rome, Italy
| | - Massimo Tatti
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Simona Camero
- Department Maternal Infantile and Urological Sciences, SAPIENZA University, 00161 Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy
| | - Teresa Rizza
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | | | - Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Rosalba Carrozzo
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Sara Rinalducci
- Department of Ecological and Biological Sciences, Università della Tuscia, 01100 Viterbo, Italy
| | - Giovanni Chillemi
- Department for Innovation in Biological, Agro-food and Forest systems, Università della Tuscia, 01100 Viterbo, Italy
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
57
|
Guan Z, Lan H, Cai X, Zhang Y, Liang A, Li J. Blood-Brain Barrier, Cell Junctions, and Tumor Microenvironment in Brain Metastases, the Biological Prospects and Dilemma in Therapies. Front Cell Dev Biol 2021; 9:722917. [PMID: 34504845 PMCID: PMC8421648 DOI: 10.3389/fcell.2021.722917] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/16/2021] [Indexed: 12/25/2022] Open
Abstract
Brain metastasis is the most commonly seen brain malignancy, frequently originating from lung cancer, breast cancer, and melanoma. Brain tumor has its unique cell types, anatomical structures, metabolic constraints, and immune environment, which namely the tumor microenvironment (TME). It has been discovered that the tumor microenvironment can regulate the progression, metastasis of primary tumors, and response to the treatment through the particular cellular and non-cellular components. Brain metastasis tumor cells that penetrate the brain–blood barrier and blood–cerebrospinal fluid barrier to alter the function of cell junctions would lead to different tumor microenvironments. Emerging evidence implies that these tumor microenvironment components would be involved in mechanisms of immune activation, tumor hypoxia, antiangiogenesis, etc. Researchers have applied various therapeutic strategies to inhibit brain metastasis, such as the combination of brain radiotherapy, immune checkpoint inhibitors, and monoclonal antibodies. Unfortunately, they hardly access effective treatment. Meanwhile, most clinical trials of target therapy patients with brain metastasis are always excluded. In this review, we summarized the clinical treatment of brain metastasis in recent years, as well as their influence and mechanisms underlying the differences between the composition of tumor microenvironments in the primary tumor and brain metastasis. We also look forward into the feasibility and superiority of tumor microenvironment-targeted therapies in the future, which may help to improve the strategy of brain metastasis treatment.
Collapse
Affiliation(s)
- Zhiyuan Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongyu Lan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xin Cai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yichi Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Annan Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
58
|
Can the Mitochondrial Metabolic Theory Explain Better the Origin and Management of Cancer than Can the Somatic Mutation Theory? Metabolites 2021; 11:metabo11090572. [PMID: 34564387 PMCID: PMC8467939 DOI: 10.3390/metabo11090572] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022] Open
Abstract
A theory that can best explain the facts of a phenomenon is more likely to advance knowledge than a theory that is less able to explain the facts. Cancer is generally considered a genetic disease based on the somatic mutation theory (SMT) where mutations in proto-oncogenes and tumor suppressor genes cause dysregulated cell growth. Evidence is reviewed showing that the mitochondrial metabolic theory (MMT) can better account for the hallmarks of cancer than can the SMT. Proliferating cancer cells cannot survive or grow without carbons and nitrogen for the synthesis of metabolites and ATP (Adenosine Triphosphate). Glucose carbons are essential for metabolite synthesis through the glycolysis and pentose phosphate pathways while glutamine nitrogen and carbons are essential for the synthesis of nitrogen-containing metabolites and ATP through the glutaminolysis pathway. Glutamine-dependent mitochondrial substrate level phosphorylation becomes essential for ATP synthesis in cancer cells that over-express the glycolytic pyruvate kinase M2 isoform (PKM2), that have deficient OxPhos, and that can grow in either hypoxia (0.1% oxygen) or in cyanide. The simultaneous targeting of glucose and glutamine, while elevating levels of non-fermentable ketone bodies, offers a simple and parsimonious therapeutic strategy for managing most cancers.
Collapse
|
59
|
Huang S, Wang Z, Zhao L. The Crucial Roles of Intermediate Metabolites in Cancer. Cancer Manag Res 2021; 13:6291-6307. [PMID: 34408491 PMCID: PMC8364365 DOI: 10.2147/cmar.s321433] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Metabolic alteration, one of the hallmarks of cancer cells, is important for cancer initiation and development. To support their rapid growth, cancer cells alter their metabolism so as to obtain the necessary energy and building blocks for biosynthetic pathways, as well as to adjust their redox balance. Once thought to be merely byproducts of metabolic pathways, intermediate metabolites are now known to mediate epigenetic modifications and protein post-transcriptional modifications (PTM), as well as connect cellular metabolism with signal transduction. Consequently, they can affect a myriad of processes, including proliferation, apoptosis, and immunity. In this review, we summarize multiple representative metabolites involved in glycolysis, the pentose phosphate pathway (PPP), the tricarboxylic acid (TCA) cycle, lipid synthesis, ketogenesis, methionine metabolism, glutamine metabolism, and tryptophan metabolism, focusing on their roles in chromatin and protein modifications and as signal-transducing messengers.
Collapse
Affiliation(s)
- Sisi Huang
- Hengyang School of Medicine, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Zhiqin Wang
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Liang Zhao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| |
Collapse
|
60
|
Miranda-Gonçalves V, Gonçalves CS, Granja S, Vieira de Castro J, Reis RM, Costa BM, Baltazar F. MCT1 Is a New Prognostic Biomarker and Its Therapeutic Inhibition Boosts Response to Temozolomide in Human Glioblastoma. Cancers (Basel) 2021; 13:cancers13143468. [PMID: 34298681 PMCID: PMC8306807 DOI: 10.3390/cancers13143468] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Glioblastoma, the brain tumour with highest prevalence and lethality, exhibits a characteristic glycolytic phenotype with increased lactate production. Recently, we reported a MCT1 overexpression in GBMs tumours, being associated to tumour growth and aggressiveness. Thus, we aimed to disclose the role of MCT1 in GBM prognosis and in vivo therapy response. Importantly, MCT1 overexpression is associated with poor prognosis of GBM. Moreover, MCT1 inhibition retards GBM tumour growth and boosts response to temozolomide treatment. Abstract Background: Glioblastomas (GBMs) present remarkable metabolism reprograming, in which many cells display the “Warburg effect”, with the production of high levels of lactate that are extruded to the tumour microenvironment by monocarboxylate transporters (MCTs). We described previously that MCT1 is up-regulated in human GBM samples, and MCT1 inhibition decreases glioma cell viability and aggressiveness. In the present study, we aimed to unveil the role of MCT1 in GBM prognosis and to explore it as a target for GBM therapy in vivo. Methods: MCT1 activity and protein expression were inhibited by AR-C155858 and CHC compounds or stable knockdown with shRNA, respectively, to assess in vitro and in vivo the effects of MCT1 inhibition and on response of GBM to temozolomide. Survival analyses on GBM patient cohorts were performed using Cox regression and Log-rank tests. Results: High levels of MCT1 expression were revealed to be a predictor of poor prognosis in multiple cohorts of GBM patients. Functionally, in U251 GBM cells, MCT1 stable knockdown decreased glucose consumption and lactate efflux, compromising the response to the MCT1 inhibitors CHC and AR-C155858. MCT1 knockdown significantly increased the survival of orthotopic GBM intracranial mice models when compared to their control counterparts. Furthermore, MCT1 downregulation increased the sensitivity to temozolomide in vitro and in vivo, resulting in significantly longer mice survival. Conclusions: This work provides first evidence for MCT1 as a new prognostic biomarker of GBM survival and further supports MCT1 targeting, alone or in combination with classical chemotherapy, for the treatment of GBM.
Collapse
Affiliation(s)
- Vera Miranda-Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (V.M.-G.); (C.S.G.); (S.G.); (J.V.d.C.); (R.M.R.); (B.M.C.)
- ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Céline S. Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (V.M.-G.); (C.S.G.); (S.G.); (J.V.d.C.); (R.M.R.); (B.M.C.)
- ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Sara Granja
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (V.M.-G.); (C.S.G.); (S.G.); (J.V.d.C.); (R.M.R.); (B.M.C.)
- ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
- Research Centre in Health and Environment (CISA), School of Health (ESS), Polytechnic Institute of Porto (P.PORTO), 4200-072 Porto, Portugal
- Department of Pathological, Cytological and Thanatological Anatomy, School of Health (ESS), Polytechnic Institute of Porto (P.PORTO), 4200-072 Porto, Portugal
| | - Joana Vieira de Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (V.M.-G.); (C.S.G.); (S.G.); (J.V.d.C.); (R.M.R.); (B.M.C.)
- ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Rui M. Reis
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (V.M.-G.); (C.S.G.); (S.G.); (J.V.d.C.); (R.M.R.); (B.M.C.)
- ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, SP, Brazil
| | - Bruno M. Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (V.M.-G.); (C.S.G.); (S.G.); (J.V.d.C.); (R.M.R.); (B.M.C.)
- ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (V.M.-G.); (C.S.G.); (S.G.); (J.V.d.C.); (R.M.R.); (B.M.C.)
- ICVS/3Bs-PT Government Associate Laboratory, 4805-017 Guimarães, Portugal
- Correspondence: ; Tel.: +351-253-604828
| |
Collapse
|
61
|
Talib WH, Mahmod AI, Kamal A, Rashid HM, Alashqar AMD, Khater S, Jamal D, Waly M. Ketogenic Diet in Cancer Prevention and Therapy: Molecular Targets and Therapeutic Opportunities. Curr Issues Mol Biol 2021; 43:558-589. [PMID: 34287243 PMCID: PMC8928964 DOI: 10.3390/cimb43020042] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022] Open
Abstract
Although cancer is still one of the most significant global challenges facing public health, the world still lacks complementary approaches that would significantly enhance the efficacy of standard anticancer therapies. One of the essential strategies during cancer treatment is following a healthy diet program. The ketogenic diet (KD) has recently emerged as a metabolic therapy in cancer treatment, targeting cancer cell metabolism rather than a conventional dietary approach. The ketogenic diet (KD), a high-fat and very-low-carbohydrate with adequate amounts of protein, has shown antitumor effects by reducing energy supplies to cells. This low energy supply inhibits tumor growth, explaining the ketogenic diet's therapeutic mechanisms in cancer treatment. This review highlights the crucial mechanisms that explain the ketogenic diet's potential antitumor effects, which probably produces an unfavorable metabolic environment for cancer cells and can be used as a promising adjuvant in cancer therapy. Studies discussed in this review provide a solid background for researchers and physicians to design new combination therapies based on KD and conventional therapies.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Ayah Kamal
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Hasan M. Rashid
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Aya M. D. Alashqar
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Samar Khater
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Duaa Jamal
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Mostafa Waly
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khoud 34-123, Oman;
| |
Collapse
|
62
|
Chen AN, Luo Y, Yang YH, Fu JT, Geng XM, Shi JP, Yang J. Lactylation, a Novel Metabolic Reprogramming Code: Current Status and Prospects. Front Immunol 2021; 12:688910. [PMID: 34177945 PMCID: PMC8222712 DOI: 10.3389/fimmu.2021.688910] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022] Open
Abstract
Lactate is an end product of glycolysis. As a critical energy source for mitochondrial respiration, lactate also acts as a precursor of gluconeogenesis and a signaling molecule. We briefly summarize emerging concepts regarding lactate metabolism, such as the lactate shuttle, lactate homeostasis, and lactate-microenvironment interaction. Accumulating evidence indicates that lactate-mediated reprogramming of immune cells and enhancement of cellular plasticity contribute to establishing disease-specific immunity status. However, the mechanisms by which changes in lactate states influence the establishment of diverse functional adaptive states are largely uncharacterized. Posttranslational histone modifications create a code that functions as a key sensor of metabolism and are responsible for transducing metabolic changes into stable gene expression patterns. In this review, we describe the recent advances in a novel lactate-induced histone modification, histone lysine lactylation. These observations support the idea that epigenetic reprogramming-linked lactate input is related to disease state outputs, such as cancer progression and drug resistance.
Collapse
Affiliation(s)
- An-Na Chen
- Department of Translational Medicine Center, Affiliated Hospital of Hangzhou Normal University, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| | - Yan Luo
- Department of Translational Medicine Center, Affiliated Hospital of Hangzhou Normal University, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| | - Yu-Han Yang
- Department of Translational Medicine Center, Affiliated Hospital of Hangzhou Normal University, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| | - Jian-Tao Fu
- Department of Translational Medicine Center, Affiliated Hospital of Hangzhou Normal University, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| | - Xiu-Mei Geng
- Department of Translational Medicine Center, Affiliated Hospital of Hangzhou Normal University, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| | - Jun-Ping Shi
- Department of Translational Medicine Center, Affiliated Hospital of Hangzhou Normal University, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| | - Jin Yang
- Department of Translational Medicine Center, Affiliated Hospital of Hangzhou Normal University, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
63
|
Zhang Y, Zhang T, Yang W, Chen H, Geng X, Li G, Chen H, Wang Y, Li L, Sun B. Beneficial Diets and Pancreatic Cancer: Molecular Mechanisms and Clinical Practice. Front Oncol 2021; 11:630972. [PMID: 34123787 PMCID: PMC8193730 DOI: 10.3389/fonc.2021.630972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/19/2021] [Indexed: 01/02/2023] Open
Abstract
Pancreatic cancer (PC) is a malignant tumor with high invasiveness, easy metastatic ability, and chemoresistance. Patients with PC have an extremely low survival rate due to the difficulty in early diagnosis. It is estimated that nearly 90% of PC cases are caused by environmental risk factors. Approximately 50% of PC cases are induced by an unhealthy diet, which can be avoided. Given this large attribution to diet, numerous studies have assessed the relationship between various dietary factors and PC. This article reviews three beneficial diets: a ketogenic diet (KD), a Mediterranean diet (MD), and a low-sugar diet. Their composition and impact mechanism are summarized and discussed. The associations between these three diets and PC were analyzed, and we aimed to provide more help and new insights for the prevention and treatment of PC.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tao Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbo Yang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongze Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinglong Geng
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guanqun Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hua Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongwei Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| |
Collapse
|
64
|
Abstract
Glucose is converted to energy through “fermentation” or “oxidation.” Generally, if oxygen is available, cells will oxidize glucose to CO2 because it is more efficient than fermentation, which produces lactic acid. But Warburg noted that cancers ferment glucose at a “remarkable” rate even if O2 is available! This “Warburg Effect” is still misunderstood because it doesn’t make sense that a cell would ferment glucose when it could get much more energy by oxidizing it. The current paper goes to the heart of this problem by defining the microenvironmental conditions that exist in early cancers that would select for a Warburg Effect. This is important because such cells are much more aggressive and like to lead to cancers that are lethal. The harsh microenvironment of ductal carcinoma in situ (DCIS) exerts strong evolutionary selection pressures on cancer cells. We hypothesize that the poor metabolic conditions near the ductal center foment the emergence of a Warburg Effect (WE) phenotype, wherein cells rapidly ferment glucose to lactic acid, even in normoxia. To test this hypothesis, we subjected low-glycolytic breast cancer cells to different microenvironmental selection pressures using combinations of hypoxia, acidosis, low glucose, and starvation for many months and isolated single clones for metabolic and transcriptomic profiling. The two harshest conditions selected for constitutively expressed WE phenotypes. RNA sequencing analysis of WE clones identified the transcription factor KLF4 as potential inducer of the WE phenotype. In stained DCIS samples, KLF4 expression was enriched in the area with the harshest microenvironmental conditions. We simulated in vivo DCIS phenotypic evolution using a mathematical model calibrated from the in vitro results. The WE phenotype emerged in the poor metabolic conditions near the necrotic core. We propose that harsh microenvironments within DCIS select for a WE phenotype through constitutive transcriptional reprogramming, thus conferring a survival advantage and facilitating further growth and invasion.
Collapse
|
65
|
Wu DJ. Oversupply of Limiting Cell Resources and the Evolution of Cancer Cells: A Review. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.653622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cancer prevention is superior to cancer treatment—indeed, understanding and controlling cancer risk is a key question in the fields of applied ecology and evolutionary oncology. Ecological cancer risk models offer the dual benefit of being generalizable across cancer types, and unveiling common mechanisms underlying cancer development and spread. Understanding the biological mechanisms of cancer risk may also guide the design of interventions to prevent cancer. Ecological considerations are central to many of these mechanisms; as one example, the ecologically-based hypothesis of metabolic cancer suppression posits that restricted vascular supply of limiting resources to somatic tissues normally suppresses the evolution of somatic cells toward cancer. Here we present a critical review of published evidence relevant to this hypothesis, and we conclude that there is substantial evidence that cancer risk does increase with an abnormal excess of limiting cell resources, including both dietary macronutrients as well as certain micronutrients.
Collapse
|
66
|
Benito I, Encío IJ, Milagro FI, Alfaro M, Martínez-Peñuela A, Barajas M, Marzo F. Microencapsulated Bifidobacterium bifidum and Lactobacillus gasseri in Combination with Quercetin Inhibit Colorectal Cancer Development in Apc Min/+ Mice. Int J Mol Sci 2021; 22:4906. [PMID: 34063173 PMCID: PMC8124226 DOI: 10.3390/ijms22094906] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/28/2021] [Accepted: 05/01/2021] [Indexed: 12/12/2022] Open
Abstract
Recent studies have suggested that flavonoids such as quercetin and probiotics such as Bifidobacterium bifidum (Bf) and Lactobacillus gasseri (Lg) could play a relevant role in inhibiting colon cancer cell growth. Our study investigated the role of dietary supplementation with microencapsulated probiotics (Bf and Lg) along with quercetin in the development of mouse colorectal cancer (CRC). Methods: Adenomatous polyposis coli/multiple intestinal neoplasia (ApcMin/+) mice were fed a standard diet or the same diet supplemented with microencapsulated probiotics (Bf and Lg strains, 107 CFU/100 g food) or both probiotics strains plus microencapsulated quercetin (15 mg/100 g food) for 73 days. Changes in body and organ weights, energy metabolism, intestinal microbiota, and colon tissue were determined. The expression of genes related to the Wnt pathway was also analyzed in colon samples. Results: Dietary supplementation with microencapsulated probiotics or microencapsulated probiotics plus quercetin reduced body weight loss and intestinal bleeding in ApcMin/+ mice. An improvement in energy expenditure was observed after 8 weeks but not after 10 weeks of treatment. A supplemented diet with microencapsulated Bf and Lg reduced the number of aberrant crypt foci (ACF) and adenomas by 45% and 60%, respectively, whereas the supplementation with Bf, Lg and quercetin decreased the number of ACF and adenomas by 57% and 80%, respectively. Microencapsulated Bf and Lg in combination with quercetin could exert inhibition of the canonical Wnt/β-catenin signaling pathway in the colon of ApcMin/+ mice Conclusions: The administration of microencapsulated Bf and Lg, individually or in combination with quercetin, inhibits the CRC development in ApcMin/+ mice.
Collapse
Affiliation(s)
- Iván Benito
- Laboratory of Animal Physiology and Nutrition, School of Agronomy, Public University of Navarre, Campus Arrosadia, 31006 Pamplona, Spain; (I.B.); (M.A.)
| | - Ignacio J. Encío
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain;
| | - Fermín I. Milagro
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Alfaro
- Laboratory of Animal Physiology and Nutrition, School of Agronomy, Public University of Navarre, Campus Arrosadia, 31006 Pamplona, Spain; (I.B.); (M.A.)
| | | | - Miguel Barajas
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain;
| | - Florencio Marzo
- Laboratory of Animal Physiology and Nutrition, School of Agronomy, Public University of Navarre, Campus Arrosadia, 31006 Pamplona, Spain; (I.B.); (M.A.)
| |
Collapse
|
67
|
Singh SV, Chaube B, Mayengbam SS, Singh A, Malvi P, Mohammad N, Deb A, Bhat MK. Metformin induced lactic acidosis impaired response of cancer cells towards paclitaxel and doxorubicin: Role of monocarboxylate transporter. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166011. [PMID: 33212188 DOI: 10.1016/j.bbadis.2020.166011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/30/2022]
Abstract
Abnormal glucose metabolism in cancer cells causes generation and secretion of excess lactate, which results in acidification of the extracellular microenvironment. This altered metabolism aids not only in survival and proliferation but also in suppressing immune-mediated destruction of cancer cells. However, how it influences the response of cancer cells to chemotherapeutic drugs is not clearly understood. We employed appropriate in vitro approaches to explore the role of mono-carboxylate transporter 4 (MCT4) mediated altered intra and extracellular pH on the outcome of the therapeutic efficacy of chemotherapeutic drugs in breast and lung cancer models. We demonstrate by in vitro experiments that inhibition of complex I enhances glycolysis and increases expression as well as membrane translocation of MCT4. It causes a decrease in extracellular pH (pHe) and impairs doxorubicin and paclitaxel's therapeutic efficacy. Acidic pHe inhibits doxorubicin's uptake, while acidic intracellular pH (pH i) impairs the efficacy of paclitaxel. Under in vivo experimental settings, the modulation of pHe with phloretin or alkalizer (NaHCO3) enhances cytotoxicity of drugs and inhibits the growth of MCF-7 xenografts in mice. In a nutshell, this study indicates that MCT4 mediated extracellular acidosis is involved in impairing chemotherapeutic drugs' efficacy on cancer cells. Therefore, the use of pH neutralizing agents or MCT inhibitors may be beneficial towards circumventing impairment in the efficacy of certain drugs that are sensitive to pH changes.
Collapse
Affiliation(s)
- Shivendra Vikram Singh
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune 411 007, India
| | - Balkrishna Chaube
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune 411 007, India
| | | | - Abhijeet Singh
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune 411 007, India
| | - Parmanand Malvi
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune 411 007, India
| | - Naoshad Mohammad
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune 411 007, India
| | - Ankita Deb
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune 411 007, India
| | - Manoj Kumar Bhat
- National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind, Pune 411 007, India.
| |
Collapse
|
68
|
Mboge MY, Coombs J, Singh S, Andring J, Wolff A, Tu C, Zhang Z, McKenna R, Frost SC. Inhibition of Carbonic Anhydrase Using SLC-149: Support for a Noncatalytic Function of CAIX in Breast Cancer. J Med Chem 2021; 64:1713-1724. [PMID: 33523653 PMCID: PMC9945910 DOI: 10.1021/acs.jmedchem.0c02077] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Carbonic anhydrase IX (CAIX) is considered a target for therapeutic intervention in solid tumors. In this study, the efficacy of the inhibitor, 4-(3-(2,4-difluorophenyl)-oxoimidazolidin-1-yl)benzenesulfonamide (SLC-149), is evaluated on CAIX and a CAIX-mimic. We show that SLC-149 is a better inhibitor than acetazolamide against CAIX. Binding of SLC-149 thermally stabilizes CAIX-mimic at lower concentrations compared to that of CAII. Structural examinations of SLC-149 bound to CAIX-mimic and CAII explain binding preferences. In cell culture, SLC-149 is a more effective inhibitor of CAIX activity in a triple-negative breast cancer cell line than previously studied sulfonamide inhibitors. SLC-149 is also a better inhibitor of activity in cells expressing CAIX versus CAXII. However, SLC-149 has little effect on cytotoxicity, and high concentrations are required to inhibit cell growth, migration, and invasion. These data support the hypothesis that CAIX activity, shown to be important in regulating extracellular pH, does not underlie its ability to control cell growth.
Collapse
Affiliation(s)
- Mam Y. Mboge
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Jacob Coombs
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Srishti Singh
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Jacob Andring
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Alyssa Wolff
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Chingkuang Tu
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Zaihui Zhang
- SignalChem Lifesciences Corp 13120 Vanier Place, Richmond, British Columbia V6V 2J2
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Susan C. Frost
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| |
Collapse
|
69
|
Wu XT, Wang YH, Cai XY, Dong Y, Cui Q, Zhou YN, Yang XW, Lu WF, Zhang M. RNF115 promotes lung adenocarcinoma through Wnt/β-catenin pathway activation by mediating APC ubiquitination. Cancer Metab 2021; 9:7. [PMID: 33509267 PMCID: PMC7842072 DOI: 10.1186/s40170-021-00243-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Background Patients with lung adenocarcinoma (LUAD) have high mortality rate and poor prognosis. The LUAD cells display increased aerobic glycolysis, which generates energy required for their survival and proliferation. Deregulation of Wnt/β-catenin signaling pathway induces the metabolism switching and oncogenesis in tumor cells. RING finger protein 115 (RNF115) is an E3 ligase for ubiquitin-mediated degradation. Although the oncogenic functions of RNF115 have been revealed in breast tumor cells, the effect of RNF115 on lung cancer is still not clear. Methods RNF115 expression and its correlation with the features of LUAD patients were analyzed by using public database and our own cohort. The functions of RNF115 in proliferation and energy metabolism in LUAD cells were explored by downregulating or upregulating RNF115 expression. Results We demonstrated that RNF115 was overexpressed in LUAD tissues and its expression was positively correlated with the poor overall survival of LUAD patients. Moreover, RNF115 overexpression inhibited LUAD cell apoptosis and promoted cellular proliferation and metabolism in LUAD cells. On the contrary, RNF115 knockdown displayed reverse effects. Furthermore, the underlying mechanism of the biological function of RNF115 in LUAD was through regulating Wnt/β-catenin pathway via ubiquitination of adenomatous polyposis coli (APC). Conclusion The current study reveals a close association between RNF115 expression and prognostic conditions in LUAD patients and the oncogenic roles of RNF115 in LUAD at the first time. These findings may help establish the foundation for the development of therapeutics strategies and clinical management for lung cancer in future. Supplementary Information The online version contains supplementary material available at 10.1186/s40170-021-00243-y.
Collapse
Affiliation(s)
- Xiao-Ting Wu
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Xuhui District, Shanghai, 200030, China
| | - Yu-Han Wang
- Department of Integrated Traditional Chinese and Western Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Yue Cai
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Xuhui District, Shanghai, 200030, China
| | - Yun Dong
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Xuhui District, Shanghai, 200030, China
| | - Qing Cui
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Xuhui District, Shanghai, 200030, China
| | - Ya-Ning Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Xuhui District, Shanghai, 200030, China
| | - Xi-Wen Yang
- LongHua Hospital Shanghai University of Traditional Chinese Medicine/Oncology Division 2, Shanghai, China
| | - Wen-Feng Lu
- Department of Integrated Traditional Chinese and Western Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Xuhui District, Shanghai, 200030, China.
| |
Collapse
|
70
|
The hypoxia-sensor carbonic anhydrase IX affects macrophage metabolism, but is not a suitable biomarker for human cardiovascular disease. Sci Rep 2021; 11:425. [PMID: 33432108 PMCID: PMC7801702 DOI: 10.1038/s41598-020-79978-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/11/2020] [Indexed: 01/18/2023] Open
Abstract
Hypoxia is prevalent in atherosclerotic plaques, promoting plaque aggravation and subsequent cardiovascular disease (CVD). Transmembrane protein carbonic anhydrase IX (CAIX) is hypoxia-induced and can be shed into the circulation as soluble CAIX (sCAIX). As plaque macrophages are hypoxic, we hypothesized a role for CAIX in macrophage function, and as biomarker of hypoxic plaque burden and CVD. As tumor patients with probable CVD are treated with CAIX inhibitors, this study will shed light on their safety profile. CAIX co-localized with macrophages (CD68) and hypoxia (pimonidazole), and correlated with lipid core size and pro-inflammatory iNOS+ macrophages in unstable human carotid artery plaques. Although elevated pH and reduced lactate levels in culture medium of CAIX knock-out (CAIXko) macrophages confirmed its role as pH-regulator, only spare respiratory capacity of CAIXko macrophages was reduced. Proliferation, apoptosis, lipid uptake and expression of pro- and anti-inflammatory genes were not altered. Plasma sCAIX levels and plaque-resident CAIX were below the detection threshold in 50 and 90% of asymptomatic and symptomatic cases, respectively, while detectable levels did not associate with primary or secondary events, or intraplaque hemorrhage. Initial findings show that CAIX deficiency interferes with macrophage metabolism. Despite a correlation with inflammatory macrophages, plaque-resident and sCAIX expression levels are too low to serve as biomarkers of future CVD.
Collapse
|
71
|
Anemone A, Consolino L, Conti L, Irrera P, Hsu MY, Villano D, Dastrù W, Porporato PE, Cavallo F, Longo DL. Tumour acidosis evaluated in vivo by MRI-CEST pH imaging reveals breast cancer metastatic potential. Br J Cancer 2021; 124:207-216. [PMID: 33257841 PMCID: PMC7782702 DOI: 10.1038/s41416-020-01173-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/07/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Tumour acidosis is considered to play a central role in promoting cancer invasion and migration, but few studies have investigated in vivo how tumour pH correlates with cancer invasion. This study aims to determine in vivo whether tumour acidity is associated with cancer metastatic potential. METHODS Breast cancer cell lines with different metastatic potentials have been characterised for several markers of aggressiveness and invasiveness. Murine tumour models have been developed and assessed for lung metastases and tumour acidosis has been assessed in vivo by a magnetic resonance imaging-based chemical exchange saturation transfer (CEST) pH imaging approach. RESULTS The higher metastatic potential of 4T1 and TS/A primary tumours, in comparison to the less aggressive TUBO and BALB-neuT ones, was confirmed by the highest expression of cancer cell stem markers (CD44+CD24-), highlighting their propensity to migrate and invade, coinciding with the measurement obtained by in vitro assays. MRI-CEST pH imaging successfully discriminated the more aggressive 4T1 and TS/A tumours that displayed a more acidic pH. Moreover, the observed higher tumour acidity was significantly correlated with an increased number of lung metastases. CONCLUSIONS The findings of this study indicate that the extracellular acidification is associated with the metastatic potential.
Collapse
Affiliation(s)
- Annasofia Anemone
- Department of Molecular Biotechnology and Health Sciences, Molecular Imaging Center, University of Torino, Via Nizza 52, Torino, Italy
| | - Lorena Consolino
- Department of Molecular Biotechnology and Health Sciences, Molecular Imaging Center, University of Torino, Via Nizza 52, Torino, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, Torino, Italy
| | - Pietro Irrera
- University of Campania "Luigi Vanvitelli", Viale Abramo Lincoln, 5, Caserta, Italy
| | - Myriam Y Hsu
- Department of Molecular Biotechnology and Health Sciences, Molecular Imaging Center, University of Torino, Via Nizza 52, Torino, Italy
| | - Daisy Villano
- Department of Molecular Biotechnology and Health Sciences, Molecular Imaging Center, University of Torino, Via Nizza 52, Torino, Italy
| | - Walter Dastrù
- Department of Molecular Biotechnology and Health Sciences, Molecular Imaging Center, University of Torino, Via Nizza 52, Torino, Italy
| | - Paolo E Porporato
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, Torino, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), Italian National Research Council (CNR), Via Nizza 52, Torino, Italy.
| |
Collapse
|
72
|
Flavylium Dye as pH-Tunable Fluorescent and CD Probe for Double-Stranded DNA and RNA. CHEMOSENSORS 2020. [DOI: 10.3390/chemosensors8040129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The interaction of 4′-(N,N-dimethylamino)-6-hydroxyflavylium cation with double stranded (ds-) DNA/RNA was studied by UV/Vis spectrophotometry, circular dichroism (CD), and also steady-state and time-resolved emission spectroscopies at neutral and weakly acidic conditions. At pH 5, the studied molecule, in its flavylium cationic form, showed considerable binding affinities (5 < logKs < 6) for all ds-DNA/RNA, contrary to chalcones forms (dominant at pH 7), which did not show binding to polynucleotides. Flavylium cation intercalated into ds-DNAs at variance to dominant groove aggregation within ds-RNA, which was reported by RNA-specific bisignate induced CD spectrum (ICD) bands. The intrinsically negligible fluorescence of flavylium was strongly increased upon the addition of DNA or RNA, whereby both the fluorescence intensity and emission lifetimes of complexes differed considerably: the strongest emission increase was observed for AU-RNA (detection limit estimated to 10 nM) followed by AT-DNAs and the much weaker effect of GC-DNAs. Both fluorescence sensitivity on the ds-DNA/RNA secondary structure and sequence-selective ICD bands make the flavylium–chalcones system an intriguing pH-switchable new probe for distinguishing between various polynucleotide sequences.
Collapse
|
73
|
Sreeja SR, Seo SS, Kim MK. Associations of Dietary Glycemic Index, Glycemic Load and Carbohydrate with the Risk of Cervical Intraepithelial Neoplasia and Cervical Cancer: A Case-Control Study. Nutrients 2020; 12:E3742. [PMID: 33291721 PMCID: PMC7761966 DOI: 10.3390/nu12123742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/27/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The association of dietary glycemic index (GI) and glycemic load (GL) with the risk of cervical cancer has never been investigated. Thus, we aimed to find evidence of any association of GI and GL with the risk of cervical intraepithelial neoplasia (CIN) and cervical cancer. METHODS In this hospital-based case-control study, we included 1340 women (670 controls and 262, 187 and 221 patients with CIN1, CIN2/3, and cervical cancer, respectively) from the Korean human papillomavirus cohort study. Completed demographic questionnaires and semi-quantitative food-frequency questionnaires were collected. The association of dietary GI and GL with CIN and cervical cancer was estimated using a logistic regression model. RESULTS The multivariate odds ratios (OR) of the highest compared with the lowest quintile of GL for CIN1 were 2.8 (95% confidence interval (CI) = 1.33-5.88). Dietary GI and GL were not associated with CIN2/3 and cervical cancer. Stratified analyses by body mass index (BMI) indicated a positive association between GI and GL and CIN 1 risk among women with a BMI (in kg/m2) <23 (OR = 2.94; 95% CI = 1.32-6.53; p for trend = 0.031 for GI and OR = 3.15; 95% CI = 1.53-6.52; p for trend = 0.013 for GL), but not among those with a BMI of ≥23. A stratification analysis by menopausal status showed that the highest quintile of GI and GL was significantly associated with the risk of CIN1 (OR = 2.91; 95% CI = 1.43-5.96; p for trend = 0.005) (OR = 2.96; 95% CI = 1.53-5.69; p for trend = 0.023) among premenopausal women. Also, in HPV positive women, dietary GL showed significant CIN1 risk (OR = 2.61; 95% CI = 1.09-6.24; p for trend = 0.087). CONCLUSION Our case-control study supports the hypothesized associations of dietary GI and GL with increased risk of CIN1. Thus, the consumption of low GI and GL foods plays a significant role in the prevention of cervical carcinogenesis.
Collapse
Affiliation(s)
- Sundara Raj Sreeja
- Division of Cancer Epidemiology and Prevention, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Korea;
| | - Sang Soo Seo
- Center for Uterine Cancer, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Korea;
| | - Mi Kyung Kim
- Division of Cancer Epidemiology and Prevention, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Korea;
| |
Collapse
|
74
|
Damaghi M, Mori H, Byrne S, Xu L, Chen T, Johnson J, Gallant ND, Marusyk A, Borowsky AD, Gillies RJ. Collagen production and niche engineering: A novel strategy for cancer cells to survive acidosis in DCIS and evolve. Evol Appl 2020; 13:2689-2703. [PMID: 33294017 PMCID: PMC7691473 DOI: 10.1111/eva.13075] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/09/2020] [Accepted: 07/12/2020] [Indexed: 12/31/2022] Open
Abstract
Growing tumors are dynamic and nonlinear ecosystems, wherein cancer cells adapt to their local microenvironment, and these adaptations further modify the environment, inducing more changes. From nascent intraductal neoplasms to disseminated metastatic disease, several levels of evolutionary adaptations and selections occur. Here, we focus on one example of such an adaptation mechanism, namely, "niche construction" promoted by adaptation to acidosis, which is a metabolic adaptation to the early harsh environment in intraductal neoplasms. The avascular characteristics of ductal carcinoma in situ (DCIS) make the periluminal volume profoundly acidic, and cancer cells must adapt to this to survive. Based on discovery proteomics, we hypothesized that a component of acid adaptation involves production of collagen by pre-cancer cells that remodels the extracellular matrix (ECM) and stabilizes cells under acid stress. The proteomic data were surprising as collagen production and deposition are commonly believed to be the responsibility of mesenchymally derived fibroblasts, and not cells of epithelial origin. Subsequent experiments in 3D culture, spinning disk and second harmonic generation microscopy of DCIS lesions in patients' samples are concordant. Collagen production assay by acid-adapted cells in vitro demonstrated that the mechanism of induction involves the RAS and SMAD pathways. Secretome analyses show upregulation of ECM remodeling enzymes such as TGM2 and LOXL2 that are collagen crosslinkers. These data strongly indicate that acidosis in incipient cancers induces collagen production by cancer cells and support the hypothesis that this adaptation initiates a tumor-permissive microenvironment promoting survival and growth of nascent cancers.
Collapse
Affiliation(s)
- Mehdi Damaghi
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
- Department of Oncologic SciencesMorsani College of MedicineUniversity of South FloridaTampaFLUSA
| | - Hidetoshi Mori
- Center for Immunology and Infectious DiseasesComprehensive Cancer CenterDepartment of Pathology and Laboratory MedicineSchool of MedicineUniversity of California, DavisSacramentoCAUSA
| | - Samantha Byrne
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Liping Xu
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Tingan Chen
- Analytic Microscopy CoreMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Joseph Johnson
- Analytic Microscopy CoreMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Nathan D. Gallant
- Department of Mechanical EngineeringUniversity of South FloridaTampaFLUSA
| | - Andriy Marusyk
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Alexander D. Borowsky
- Center for Immunology and Infectious DiseasesComprehensive Cancer CenterDepartment of Pathology and Laboratory MedicineSchool of MedicineUniversity of California, DavisSacramentoCAUSA
| | - Robert J. Gillies
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
| |
Collapse
|
75
|
Shaterabadi Z, Nabiyouni G, Soleymani M. Correlation between effects of the particle size and magnetic field strength on the magnetic hyperthermia efficiency of dextran-coated magnetite nanoparticles. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 117:111274. [DOI: 10.1016/j.msec.2020.111274] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/27/2020] [Accepted: 07/05/2020] [Indexed: 12/15/2022]
|
76
|
Persi E, Wolf YI, Horn D, Ruppin E, Demichelis F, Gatenby RA, Gillies RJ, Koonin EV. Mutation-selection balance and compensatory mechanisms in tumour evolution. Nat Rev Genet 2020; 22:251-262. [PMID: 33257848 DOI: 10.1038/s41576-020-00299-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 12/11/2022]
Abstract
Intratumour heterogeneity and phenotypic plasticity, sustained by a range of somatic aberrations, as well as epigenetic and metabolic adaptations, are the principal mechanisms that enable cancers to resist treatment and survive under environmental stress. A comprehensive picture of the interplay between different somatic aberrations, from point mutations to whole-genome duplications, in tumour initiation and progression is lacking. We posit that different genomic aberrations generally exhibit a temporal order, shaped by a balance between the levels of mutations and selective pressures. Repeat instability emerges first, followed by larger aberrations, with compensatory effects leading to robust tumour fitness maintained throughout the tumour progression. A better understanding of the interplay between genetic aberrations, the microenvironment, and epigenetic and metabolic cellular states is essential for early detection and prevention of cancer as well as development of efficient therapeutic strategies.
Collapse
Affiliation(s)
- Erez Persi
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA.
| | - Yuri I Wolf
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - David Horn
- School of Physics and Astronomy, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Eytan Ruppin
- Cancer Data Science Lab, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Francesca Demichelis
- Department for Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.,Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Robert A Gatenby
- Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Eugene V Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
77
|
Platelet isoform of phosphofructokinase promotes aerobic glycolysis and the progression of non‑small cell lung cancer. Mol Med Rep 2020; 23:74. [PMID: 33236133 PMCID: PMC7716410 DOI: 10.3892/mmr.2020.11712] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
The platelet isoform of phosphofructokinase (PFKP) is a rate-limiting enzyme involved in glycolysis that serves an important role in various types of cancer. The aim of the present study was to explore the specific regulatory relationship between PFKP and non-small cell lung cancer (NSCLC) progression. PFKP expression in NSCLC tissues and corresponding adjacent tissues was detected using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemical analysis. PFKP expression in human bronchial epithelial cells (16HBE) and NSCLC cells (H1299, H23 and A549) was also detected using RT-qPCR. Cell proliferation was detected by Cell Counting Kit-8 and colony formation assays. Transwell invasion and wound healing assays, and flow cytometry were used to detect cell invasion, migration and apoptosis, respectively. The expression levels of glycolysis-associated enzymes (hexokinase-2, lactate dehydrogenase A and glucose transporter-1), epithelial-mesenchymal transition-related proteins (N-cadherin, vimentin and E-cadherin) and apoptosis-related proteins (caspase-3 and B-cell lymphoma-2) were detected by western blotting. Glucose uptake, lactate production and the adenosine trisphosphate/adenosine diphosphate ratio were measured using the corresponding kits. The results of the present study demonstrated that PFKP expression was upregulated in NSCLC tissues and cells, and PFKP expression was related to lymph node metastasis and histological grade. In addition, overexpression of PFKP inhibited cell apoptosis, and promoted proliferation, migration, invasion and glycolysis of H1299 cells, whereas knockdown of PFKP had the opposite effects. In conclusion, PFKP inhibited cell apoptosis, and promoted proliferation, migration, invasion and glycolysis of NSCLC cells; these findings may lay the foundation for novel treatments of NSCLC.
Collapse
|
78
|
Xu M, Wang P, Sun S, Gao L, Sun L, Zhang L, Zhang J, Wang S, Liang X. Smart strategies to overcome tumor hypoxia toward the enhancement of cancer therapy. NANOSCALE 2020; 12:21519-21533. [PMID: 33095224 DOI: 10.1039/d0nr05501h] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Hypoxia, as a typical factor in a tumor microenvironment, plays a vital role in tumor treatment resistance, tumor invasion and migration. Hypoxia inducible factor (HIF), as the vital response element of hypoxia, mediates these untoward effects through a series of downstream reactions. Cancer treatments such as photodynamic therapy (PDT), radiotherapy (RT) and chemotherapy are severely hindered by hypoxia and HIF, back, however, could be intelligently manipulated through nanocomposite materials for their great potentiality to combine different functions. Herein, we reviewed the smart strategies in emerging research studies to overcome hypoxia toward the enhancement of tumor therapy.
Collapse
Affiliation(s)
- Menghong Xu
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Hu Y, Xu W, Zeng H, He Z, Lu X, Zuo D, Qin G, Chen W. OXPHOS-dependent metabolic reprogramming prompts metastatic potential of breast cancer cells under osteogenic differentiation. Br J Cancer 2020; 123:1644-1655. [PMID: 32934344 PMCID: PMC7686370 DOI: 10.1038/s41416-020-01040-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 06/22/2020] [Accepted: 08/06/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Microcalcification is one of the most reliable clinical features of the malignancy risk of breast cancer, and it is associated with enhanced tumour aggressiveness and poor prognosis. However, its underlying molecular mechanism remains unclear. METHODS Clinical data were retrieved to analyse the association between calcification and bone metastasis in patients with breast cancer. Using multiple human breast cancer cell lines, the osteogenic cocktail model was established in vitro to demonstrate calcification-exacerbated metastasis. Migration and invasion characteristics were determined by wound healing and transwell migration. mRNA and protein expression were identified by quantitative PCR and western blotting. Metabolic alterations in breast cancer cells were evaluated using Seahorse Analyser. RESULTS The osteogenic differentiation of human breast cancer cells activated the classical TGF-β/Smad signalling pathway and the non-canonical MAPK pathway, which, in turn, exacerbated the progression of epithelial-mesenchymal transition (EMT). The metabolic programme switched to enhancing mitochondrial oxidative phosphorylation (OXPHOS) upon osteogenic differentiation. Rotenone was used to inhibit the OXPHOS complex during osteogenesis to block mitochondrial function, consequently reversing the EMT phenotype. CONCLUSIONS This study provides important insights into the mechanisms involved in breast cancer bone metastasis, and outlines a possible strategy to intervene in OXPHOS for the treatment of breast tumours.
Collapse
Affiliation(s)
- Yangling Hu
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Weimin Xu
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Hui Zeng
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Zilong He
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Xiao Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Daming Zuo
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, 510515, Guangzhou, China
| | - Genggeng Qin
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| | - Weiguo Chen
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
80
|
Riess JW, Frankel P, Shackelford D, Dunphy M, Badawi RD, Nardo L, Cherry SR, Lanza I, Reid J, Gonsalves WI, Kunos C, Gandara DR, Lara PN, Newman E, Paik PK. Phase 1 Trial of MLN0128 (Sapanisertib) and CB-839 HCl (Telaglenastat) in Patients With Advanced NSCLC (NCI 10327): Rationale and Study Design. Clin Lung Cancer 2020; 22:67-70. [PMID: 33229301 DOI: 10.1016/j.cllc.2020.10.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/12/2020] [Indexed: 01/24/2023]
Abstract
INTRODUCTION There are currently no approved targeted therapies for lung squamous-cell carcinoma (LSCC) and KRAS-mutant lung adenocarcinoma (LUAD). About 30% of LSCC and 25% of KRAS-mutant LUAD exhibit hyperactive NRF2 pathway activation through mutations in NFE2L2 (the gene encoding NRF2) or its negative regulator, KEAP1. Preclinical data demonstrate that these tumors are uniquely sensitive to dual inhibition of glycolysis and glutaminolysis via mammalian target of rapamycin (mTOR) and glutaminase inhibitors. This phase 1 study was designed to assess safety and preliminary activity of the mTOR inhibitor MLN0128 (sapanisertib) in combination with the glutaminase inhibitor CB-839 HCl. METHODS Phase 1 dose finding will use the queue-based variation of the 3 + 3 dose escalation scheme with the primary endpoint of identifying the recommended expansion dose. To confirm the acceptable tolerability of the recommended expansion dose, patients will subsequently enroll onto 1 of 4 expansion cohorts (n = 14 per cohort): (1) LSCC harboring NFE2L2 or (2) KEAP1 mutations, or (3) LUAD harboring KRAS/(KEAP1 or NFE2L2) coalterations, or (4) LSCC wild type for NFE2L2 and KEAP1. The primary endpoint of the dose expansion is to determine the preliminary efficacy of MLN0128/CB-839 combination therapy. CONCLUSION This phase 1 study will determine the recommended expansion dose and preliminary efficacy of MLN0128 and CB-839 in advanced non-small-cell lung cancer with a focus on subsets of LSCC and KRAS-mutant LUAD harboring NFE2L2 or KEAP1 mutations.
Collapse
Affiliation(s)
- Jonathan W Riess
- Division of Hematology/Oncology, Department of Internal Medicine, UC Davis Medical Center, UC Davis Comprehensive Cancer Center, Sacramento, CA.
| | - Paul Frankel
- City of Hope Department of Biostatistics, Duarte, CA
| | - David Shackelford
- Department of Molecular and Medical Pharmacology, UCLA, Rochester, MN
| | - Mark Dunphy
- Division of Nuclear Medicine, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ramsey D Badawi
- Division of Nuclear Medicine, Department of Radiology, UC Davis Medical Center, Sacramento, CA; Department of Biomedical Engineering, UC Davis School of Medicine, Sacramento, CA
| | - Lorenzo Nardo
- Division of Nuclear Medicine, Department of Radiology, UC Davis Medical Center, Sacramento, CA
| | - Simon R Cherry
- Department of Biomedical Engineering, UC Davis School of Medicine, Sacramento, CA; Division of Nuclear Medicine, Department of Radiology, UC Davis Medical Center, Sacramento, CA
| | - Ian Lanza
- Division of Nuclear Medicine, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Joel Reid
- Division of Nuclear Medicine, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wilson I Gonsalves
- Division of Nuclear Medicine, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Charles Kunos
- National Cancer Institute, Cancer Therapy Evaluation Program, Rockville, MD
| | - David R Gandara
- Division of Hematology/Oncology, Department of Internal Medicine, UC Davis Medical Center, UC Davis Comprehensive Cancer Center, Sacramento, CA
| | - Primo N Lara
- Division of Hematology/Oncology, Department of Internal Medicine, UC Davis Medical Center, UC Davis Comprehensive Cancer Center, Sacramento, CA
| | - Edward Newman
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Paul K Paik
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY
| |
Collapse
|
81
|
Fu X, Zhang P, Song H, Wu C, Li S, Li S, Yan C. LTBP1 plays a potential bridge between depressive disorder and glioblastoma. J Transl Med 2020; 18:391. [PMID: 33059753 PMCID: PMC7566028 DOI: 10.1186/s12967-020-02509-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/26/2020] [Indexed: 12/27/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most malignant tumor in human brain. Diagnosis and treatment of GBM may lead to psychological disorders such as depressive and anxiety disorders. There was no research focusing on the correlation between depressive/anxiety disorder and the outcome of GBM. Thus, the aim of this study was to investigate the possibility of depressive/anxiety disorder correlated with the outcome of GBM patients, as well as the overlapped mechanism bridge which could link depressive/anxiety disorders and GBM. Methods Patient Health Questionnaire (PHQ-9) and Generalized Anxiety Disorder (GAD-7) were used to investigate the psychological condition of GBM patients in our department. To further explore the potential mechanism, bioinformatic methods were used to screen out genes that could be indicators of outcome in GBM, followed by gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and protein–protein interaction (PPI) analysis. Further, cellular experiments were conducted to evaluate the proliferation, migration capacity of primary GBM cells from the patients. Results It was revealed that patients with higher PHQ-9 and GAD-7 scores had significantly worse prognosis than their lower-scored counterparts. Bioinformatic mining revealed that LTBP1 could be a potential genetic mechanism in both depressive/anxiety disorder and GBM. Primary GBM cells with different expression level of LTBP1 should significantly different proliferation and migration capacity. GO, KEGG analysis confirmed that extracellular matrix (ECM) was the most enriched function of LTBP1. PPI network showed the interaction of proteins altered by LTBP1. Hub genes COL1A2, COL5A1 and COL10A1, as well as mesenchymal marker CD44 and Vimentin were statistically higher expressed in LTBP1 high group; while proneural marker E-cadherin was significantly higher expressed in low LTBP1 group. Conclusion There is closely correlation between depressive/anxiety disorders and GBM. LTBP1 could be a potential bridge linking the two diseases through the regulation of ECM.
Collapse
Affiliation(s)
- Xiaojun Fu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China.,Capital Medical University, Beijing, People's Republic of China
| | - Pei Zhang
- Beijing Institute of Technology, Beijing, China
| | - Hongwang Song
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Chenxing Wu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China
| | | | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China.
| | - Changxiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China.
| |
Collapse
|
82
|
Brustolin L, Pettenuzzo N, Nardon C, Quarta S, Montagner I, Pontisso P, Rosato A, Conte P, Merigliano S, Fregona D. Labelled micelles for the delivery of cytotoxic Cu(II) and Ru(III) compounds in the treatment of aggressive orphan cancers: Design and biological in vitro data. J Inorg Biochem 2020; 213:111259. [PMID: 33039747 DOI: 10.1016/j.jinorgbio.2020.111259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/06/2020] [Accepted: 09/20/2020] [Indexed: 12/16/2022]
Abstract
A recent study on our metal-dithiocarbamato complexes pointed out the antiproliferative properties and the druglikeness of some new patented derivatives. In this work, the best compounds have been encapsulated in micellar nanocarriers, being also carbohydrate-functionalized on their hydrophilic surface to investigate the possibility of a cancer-selective delivery. In particular, the nonionic block copolymer Pluronic® F127 (PF127) has been chemically modified with sugars and the derivatives characterized by means of NMR spectroscopy and FT-IR spectrophotometry. Then, the two selected complexes (β-[Ru2(PipeDTC)5]Cl (PipeDTC = piperidine dithiocarbamate) and [Cu(ProOMeDTC)2] (ProOMeDTC = L-proline methyl ester dithiocarbamate)), have been loaded into the hydrophobic core of PF127 micelles and cancer-targeting counterparts. These nanoformulations have been studied for their dimensions (DLS, TEM) and stability, and tested for their cytotoxicity against aggressive human cancer cell lines. The in vitro results were paralleled with mechanistic studies through Confocal Laser Scanning Microscopy and xCELLigence analysis.
Collapse
Affiliation(s)
- Leonardo Brustolin
- Department of Surgical, Oncologic and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128 Padova, Italy; Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy
| | - Nicolò Pettenuzzo
- Department of Surgical, Oncologic and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128 Padova, Italy; Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy
| | - Chiara Nardon
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy
| | - Santina Quarta
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Isabella Montagner
- Department of Surgical, Oncologic and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128 Padova, Italy; Venetian Oncological Institute, Via Gattamelata 64, 35128 Padova, Italy
| | - Patrizia Pontisso
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Antonio Rosato
- Department of Surgical, Oncologic and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128 Padova, Italy; Venetian Oncological Institute, Via Gattamelata 64, 35128 Padova, Italy
| | - Pierfranco Conte
- Department of Surgical, Oncologic and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128 Padova, Italy; Venetian Oncological Institute, Via Gattamelata 64, 35128 Padova, Italy
| | - Stefano Merigliano
- Department of Surgical, Oncologic and Gastroenterological Sciences, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Dolores Fregona
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35131 Padova, Italy.
| |
Collapse
|
83
|
Fujita M, Imadome K, Somasundaram V, Kawanishi M, Karasawa K, Wink DA. Metabolic characterization of aggressive breast cancer cells exhibiting invasive phenotype: impact of non-cytotoxic doses of 2-DG on diminishing invasiveness. BMC Cancer 2020; 20:929. [PMID: 32993545 PMCID: PMC7525976 DOI: 10.1186/s12885-020-07414-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/15/2020] [Indexed: 01/08/2023] Open
Abstract
Background Metabolic reprogramming is being recognized as a fundamental hallmark of cancer, and efforts to identify drugs that can target cancer metabolism are underway. In this study, we used human breast cancer (BC) cell lines and established their invading phenotype (INV) collected from transwell inserts to compare metabolome differences and evaluate prognostic significance of the metabolome in aggressive BC invasiveness. Methods The invasiveness of seven human BC cell lines were compared using the transwell invasion assay. Among these, INV was collected from SUM149, which exhibited the highest invasiveness. Levels of metabolites in INV were compared with those of whole cultured SUM149 cells (WCC) using CE-TOFMS. The impact of glycolysis in INV was determined by glucose uptake assay using fluorescent derivative of glucose (2-NBDG), and significance of glycolysis, or tricarboxylic acid cycle (TCA) and electron transport chain (ETC) in the invasive process were further determined in aggressive BC cell lines, SUM149, MDA-MB-231, HCC1937, using invasion assays in the presence or absence of inhibitors of glycolysis, TCA cycle or ETC. Results SUM149 INV sub-population exhibited a persistent hyperinvasive phenotype. INV were hyper-glycolytic with increased glucose (2-NBDG) uptake; diminished glucose-6-phosphate (G6P) levels but elevated pyruvate and lactate, along with higher expression of phosphorylated-pyruvate dehydrogenase (pPDH) compared to WCC. Notably, inhibiting of glycolysis with lower doses of 2-DG (1 mM), non-cytotoxic to MDA-MB-231 and HCC1937, was effective in diminishing invasiveness of aggressive BC cell lines. In contrast, 3-Nitropropionic acid (3-NA), an inhibitor of succinate dehydrogenase, the enzyme that oxidizes succinate to fumarate in TCA cycle, and functions as complex II of ETC, had no significant effect on their invasiveness, although levels of TCA metabolites or detection of mitochondrial membrane potential with JC-1 staining, indicated that INV cells originally had functional TCA cycles and membrane potential. Conclusions Hyper-glycolytic phenotype of invading cells caters to rapid energy production required for invasion while TCA cycle/ETC cater to cellular energy needs for sustenance in aggressive BC. Lower, non-cytotoxic doses of 2-DG can hamper invasion and can potentially be used as an adjuvant with other anti-cancer therapies without the usual side-effects associated with cytotoxic doses.
Collapse
Affiliation(s)
- Mayumi Fujita
- Department of Basic Medical Science for Radiation Damages, National Institute of Radiological Sciences, NIRS, National Institute for Quantum and Radiological Science and Technology, QST, 4-9-1, Anagawa, Inage-ku, Chiba-shi, Chiba-ken, Japan.
| | - Kaori Imadome
- Department of Basic Medical Science for Radiation Damages, National Institute of Radiological Sciences, NIRS, National Institute for Quantum and Radiological Science and Technology, QST, 4-9-1, Anagawa, Inage-ku, Chiba-shi, Chiba-ken, Japan
| | - Veena Somasundaram
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Miki Kawanishi
- Department of Radiation Oncology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kumiko Karasawa
- Department of Radiation Oncology, Tokyo Women's Medical University, Tokyo, Japan
| | - David A Wink
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| |
Collapse
|
84
|
Pezzuto A, D'Ascanio M, Ricci A, Pagliuca A, Carico E. Expression and role of p16 and GLUT1 in malignant diseases and lung cancer: A review. Thorac Cancer 2020; 11:3060-3070. [PMID: 32945604 PMCID: PMC7606016 DOI: 10.1111/1759-7714.13651] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
Non‐small cell lung cancer (NSCLC) is the leading cause of cancer death and in most cases it is often diagnosed at an advanced stage. Many genetic and microenvironmental factors are able to modify the cell cycle inducing carcinogenesis and tumor growth. Among the metabolic and genetic factors that come into play in carcinogenesis and tumor cell differentiation and growth there are two different proteins that should be considered which are glucose transporters (GLUTs) and p16INK4 The first are glucose transporters which are strongly involved in tumor metabolism, notably accelerating cancer cell metabolism both in aerobic and anaerobic conditions. There are different subtypes of GLUT family factors of which GLUT 1 is the most important and widely expressed. By contrast, p16 is mainly a tumor‐suppressor protein that acts on cyclin‐dependent kinase favoring cell cycle arrest in the G1 phase. Our search focused on the action of the aforementioned factors.
Collapse
Affiliation(s)
- Aldo Pezzuto
- Cardiovascular-Pulmonary Science Department, Sant' Andrea Hospital-Sapienza University, Rome, Italy
| | - Michela D'Ascanio
- Clinical and Molecular Medicine Department, Sant' Andrea Hospital- Sapienza University, Rome, Italy
| | - Alberto Ricci
- Clinical and Molecular Medicine Department, Sant' Andrea Hospital- Sapienza University, Rome, Italy
| | - Alessandra Pagliuca
- Cardiovascular-Pulmonary Science Department, Sant' Andrea Hospital-Sapienza University, Rome, Italy
| | - Elisabetta Carico
- Clinical and Molecular Medicine Department, Sant' Andrea Hospital- Sapienza University, Rome, Italy
| |
Collapse
|
85
|
The not-so-sweet side of sugar: Influence of the microenvironment on the processes that unleash cancer. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165960. [PMID: 32919034 DOI: 10.1016/j.bbadis.2020.165960] [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: 05/21/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022]
Abstract
The role of "aerobic glycolysis" in cancer has been examined often in the past. Results from those studies, most of which were performed on two dimensional conditions (2D, tissue culture plastic), demonstrate that aerobic glycolysis occurs as a consequence of oncogenic events. These oncogenic events often drive malignant cell growth and survival. Although 2D based experiments are useful in elucidating the molecular mechanisms of oncogenesis, they fail to take contributions of the extracellular microenvironment into account. Indeed we, and others, have shown that the cellular microenvironment is essential in regulating processes that induce and/or suppress the malignant phenotype/properties. This regulation between the cell and its microenvironment is both dynamic and reciprocal and involves the integration of cellular signaling networks in the right context. Therefore, given our previous demonstration of the effect of the microenvironment including tissue architecture and media composition on gene expression and the integration of signaling events observed in three-dimension (3D), we hypothesized that glucose uptake and metabolism must also be essential components of the tissue's signal "integration plan" - that is, if uptake and metabolism of glucose were hyperactivated, the canonical oncogenic pathways should also be similarly activated. This hypothesis, if proven true, suggests that direct inhibition of glucose metabolism in cancer cells should either suppress or revert the malignant phenotype in 3D. Here, we review the up-to-date progress that has been made towards understanding the role that glucose metabolism plays in oncogenesis and re-establishing basally polarized acini in malignant human breast cells.
Collapse
|
86
|
Clementino M, Xie J, Yang P, Li Y, Lin HP, Fenske WK, Tao H, Kondo K, Yang C, Wang Z. A Positive Feedback Loop Between c-Myc Upregulation, Glycolytic Shift, and Histone Acetylation Enhances Cancer Stem Cell-like Property and Tumorigenicity of Cr(VI)-transformed Cells. Toxicol Sci 2020; 177:71-83. [PMID: 32525551 PMCID: PMC7553706 DOI: 10.1093/toxsci/kfaa086] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chronic hexavalent chromium [Cr(VI)] exposure causes lung cancer and other types of cancer; however, the mechanism of Cr(VI) carcinogenesis remains to be clearly defined. Our recent study showed that chronic Cr(VI) exposure upregulates the proto oncogene c-Myc expression, which contributes significantly to Cr(VI)-induced cell transformation, cancer stem cell (CSC)-like property and tumorigenesis. c-Myc is a master regulator of cancer cell abnormal metabolism and accumulating evidence suggests that metabolism dysregulation plays an important role in both cancer development and progression. However, little is known about the role of metabolism dysregulation in Cr(VI) carcinogenesis. This study was performed to investigate the potential role and mechanism of metabolism dysregulation in Cr(VI) carcinogenesis. It was found that Cr(VI)-transformed cells display glycolytic shift, which depends on the upregulation of c-Myc. The glycolytic shift in Cr(VI)-transformed cells led to increased production of acetyl coenzyme A (acetyl-CoA) and elevation of histone acetylation. This, in turn, upregulated the expression of an acetyl-CoA producing key enzyme ATP citrate lyase and c-Myc, forming a positive feedback loop between the upregulation of c-Myc expression, glycolytic shift and increased histone acetylation. It was further determined that glucose depletion not only reverses the glycolytic shift in Cr(VI)-transformed cells, but also significantly reduces their growth, CSC-like property and tumorigenicity. These findings indicate that glycolytic shift plays an important role in maintaining malignant phenotypes of Cr(VI)-transformed cells, suggesting that metabolism dysregulation is critically involved in Cr(VI) carcinogenesis.
Collapse
Affiliation(s)
- Marco Clementino
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Jie Xie
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Ping Yang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Yunfei Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Hsuan-Pei Lin
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536
| | - William K Fenske
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Hua Tao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Kazuya Kondo
- Department of Oncological Medical Services, Graduate School of Biomedical Sciences, Tokushima University Graduate School, Tokushima City 770-8509, Japan
| | - Chengfeng Yang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Zhishan Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536
| |
Collapse
|
87
|
Chandel V, Raj S, Kumar P, Gupta S, Dhasmana A, Kesari KK, Ruokolainen J, Mehra P, Das BC, Kamal MA, Kumar D. Metabolic regulation in HPV associated head and neck squamous cell carcinoma. Life Sci 2020; 258:118236. [PMID: 32795537 DOI: 10.1016/j.lfs.2020.118236] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/25/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022]
Abstract
Cancer cells exhibit distinct energy metabolic pathways due to multiple oncogenic events. In normoxia condition, the anaerobic glycolysis (Warburg effect) is highly observed in head and neck squamous cell carcinoma (HNSCC). HNSCC is associated with smoking, chewing tobacco, consumption of alcohol or Human Papillomavirus (HPV) infection primarily HPV16. In recent years, the correlation of HPV with HNSCC has significantly expanded. Despite the recent advancement in therapeutic approaches, the rate of HPV infected HNSCC has significantly increased in the last few years, specifically, in lower middle-income countries. The oncoproteins of High-risk Human Papillomavirus (HR-HPV), E6 and E7, alter the metabolic phenotype in HNSCC, which is distinct from non-HPV associated HNSCC. These oncoproteins, modulate the cell cycle and metabolic signalling through interacting with tumor suppressor proteins, p53 and pRb. Since, metabolic alteration represents a major hallmark for tumorigenesis, HPV acts as a source of biomarker linked to cancer progression in HNSCC. The dependency of cancer cells to specific nutrients and alteration of various metabolic associated genes may provide a unique opportunity for pharmacological intervention in HPV infected HNSCC. In this review, we have discussed the molecular mechanism (s) and metabolic regulation in HNSCC depending on the HPV status. We have also discussed the possible potential therapeutic approaches for HPV associated HNSCC through targeting metabolic pathways.
Collapse
Affiliation(s)
- Vaishali Chandel
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University UttarPradesh, Sec 125, Noida 201303, India
| | - Sibi Raj
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University UttarPradesh, Sec 125, Noida 201303, India
| | - Prabhat Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University UttarPradesh, Sec 125, Noida 201303, India
| | - Shilpi Gupta
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University UttarPradesh, Sec 125, Noida 201303, India
| | - Anupam Dhasmana
- Himalayan School of Biosciences, Swami Rama Himalayan University, Swami Ram Nagar, Jolly Grant, Doiwala, Dehradun 248016, India; Department of Immunology and Microbiology, School of Medicine, University of Rio Grande Valley, McAllen, TX, USA
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo 02150, Finland
| | - Janne Ruokolainen
- Department of Applied Physics, School of Science, Aalto University, Espoo 02150, Finland
| | - Pravesh Mehra
- Department of Oral and Maxillofacial surgery, Lady Hardinge Medical College, New Delhi, India
| | - Bhudev C Das
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University UttarPradesh, Sec 125, Noida 201303, India
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Australia; Novel Global Community Educational Foundation, NSW, Australia
| | - Dhruv Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University UttarPradesh, Sec 125, Noida 201303, India.
| |
Collapse
|
88
|
Ellingson BM, Yao J, Raymond C, Nathanson DA, Chakhoyan A, Simpson J, Garner JS, Olivero AG, Mueller LU, Rodon J, Gerstner E, Cloughesy TF, Wen PY. Multiparametric MR-PET Imaging Predicts Pharmacokinetics and Clinical Response to GDC-0084 in Patients with Recurrent High-Grade Glioma. Clin Cancer Res 2020; 26:3135-3144. [PMID: 32269051 DOI: 10.1158/1078-0432.ccr-19-3817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 02/14/2020] [Accepted: 04/03/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE GDC-0084 is an oral, brain-penetrant small-molecule inhibitor of PI3K and mTOR. Because these two targets alter tumor vascularity and metabolism, respectively, we hypothesized multiparametric MR-PET could be used to quantify the response, estimate pharmacokinetic (PK) parameters, and predict progression-free survival (PFS) in patients with recurrent malignant gliomas. PATIENTS AND METHODS Multiparametric advanced MR-PET imaging was performed to evaluate physiologic response in a first-in-man, multicenter, phase I, dose-escalation study of GDC-0084 (NCT01547546) in 47 patients with recurrent malignant glioma. RESULTS Measured maximum concentration (C max) was associated with a decrease in enhancing tumor volume (P = 0.0287) and an increase in fractional anisotropy (FA; P = 0.0418). Posttreatment tumor volume, 18F-FDG uptake, Ktrans, and relative cerebral blood volume (rCBV) were all correlated with C max. A linear combination of change in 18F-FDG PET uptake, apparent diffusion coefficient (ADC), FA, Ktrans, vp, and rCBV was able to estimate both C max (R2 = 0.4113; P < 0.0001) and drug exposure (AUC; R2 = 0.3481; P < 0.0001). Using this composite multiparametric MR-PET imaging response biomarker to predict PK, patients with an estimated C max > 0.1 μmol/L and AUC > 1.25 μmol/L*hour demonstrated significantly longer PFS compared with patients with a lower estimated concentration and exposure (P = 0.0039 and P = 0.0296, respectively). CONCLUSIONS Results from this study suggest composite biomarkers created from multiparametric MR-PET imaging targeting metabolic and/or physiologic processes specific to the drug mechanism of action may be useful for subsequent evaluation of treatment efficacy for larger phase II-III studies.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, California. .,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, California.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Brain Research Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, California
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, California.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, California
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, California.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Ararat Chakhoyan
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, University of California, Los Angeles, Los Angeles, California.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Jeremy Simpson
- Kazia Therapeutics Limited, Sydney, New South Wales, Australia
| | - James S Garner
- Kazia Therapeutics Limited, Sydney, New South Wales, Australia
| | | | | | - Jordi Rodon
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Elizabeth Gerstner
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, University of California, Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
89
|
CDC20 Knockdown and Acidic Microenvironment Collaboratively Promote Tumorigenesis through Inhibiting Autophagy and Apoptosis. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:94-106. [PMID: 32322666 PMCID: PMC7163048 DOI: 10.1016/j.omto.2020.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/25/2020] [Indexed: 12/02/2022]
Abstract
The reconstitution of the tumorigenesis process would shed light on the tumor development study and further drug selection strategies. To construct a tumorigenesis model and explore potential mechanism is of great importance. In our study, we observed that CDC20-knockdown cells cultured in acidic environment exhibited chromosomal instability and better survival ability. The tumorigenic metabolism transformation was confirmed through the increase of the extracellular acidification rate (ECAR) and decrease of the oxygen consumption rate (OCR) in CDC20-knockdown cells. After a long-term culture for 3–4 months, CDC20-knockdown cells in acidic medium showed a strong tumor formation ability by subcutaneous injection into mice that is similar to that of tumor cells. Meanwhile, transcriptome analysis of cells from different stages showed that stage D cells almost resembled the phenotype of immortal cancer cells. The oncogene accumulation laid a firm foundation in the development of the tumorigenesis process by suppressing autophagy and p53-induced apoptosis. Several autophage- and apoptosis-related genes showed inhibition during this tumorigenesis process. In summary, chromosomal instability induced by CDC20 knockdown and acidic microenvironment could collaboratively promote cell tumorigenesis through the downregulation of autophagy and apoptosis.
Collapse
|
90
|
Yang W, Zheng Z, Yi P, Wang S, Zhang N, Ming J, Tan J, Guo H. LncRNA NBR2 Inhibits the Malignancy of Thyroid Cancer, Associated With Enhancing the AMPK Signaling. Front Oncol 2020; 10:956. [PMID: 32596161 PMCID: PMC7304297 DOI: 10.3389/fonc.2020.00956] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/15/2020] [Indexed: 12/29/2022] Open
Abstract
Long non-coding RNA NBR2 is a transcript of the neighbor of BRCA1 gene 2 and can regulate tumor development. However, there is little information on the role of NBR2 in the progression of thyroid cancers (TC). Here, we show that NBR2 expression is down-regulated in TC tissues and associated with histologic subtypes of TC. NBR2 expression was variably reduced in different TC cells. While NBR2 silencing significantly enhanced the malignancy of BCPAP cells by increasing cell proliferation, clonogenicity, wound healing, and invasion as well as tumor growth in vivo, and decreasing spontaneous apoptosis, NBR2 over-expression had opposite effects in BHT101 cells. Furthermore, treatment with A-769662 (a specific AMPK activator), like NBR2 over-expression, significantly attenuated the malignancy of BHT101 cells while treatment with Compound C (a specific AMPK inhibitor) significantly rescued that NBR2-reduced malignancy of BHT101 cells. In comparison with non-tumor thyroid epithelial Nthy-ori 3-1 cells, obviously increased GLUT-1 expression, but decreased AMPK and ACC phosphorylation were detected in TC cells. While NBR2 silencing further enhanced GLUT-1 expression and reduced AMPK and ACC phosphorylation as well as the EMT process in BCPAP cells. NBR2 over-expression also had opposite effects in BHT101 cells. Similar patterns of GLUT-1 expression and AMPK and ACC phosphorylation were detected in the different types of xenograft TC tumors in vivo. Therefore, such data indicated that NBR2 acted as a tumor suppressor of thyroid cancers associated with enhancing the AMPK signaling and NBR2 may be a potential biomarker and therapeutic target for thyroid cancers.
Collapse
Affiliation(s)
- Wen Yang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhikun Zheng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengfei Yi
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Ming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Tan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Guo
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
91
|
Effects of chitosan-loaded hydroxyapatite on osteoblasts and osteosarcoma for chemopreventative applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111041. [PMID: 32600681 DOI: 10.1016/j.msec.2020.111041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 12/14/2022]
Abstract
Osteosarcoma remains one of the most common malignant primary bone tumors. Post-surgical defect repair combined with tumor suppression remains a major clinical challenge. Investigations of alternative treatments for osteosarcoma, while promising, have led to multi-drug resistance. These constraints of common treatment strategies have triggered the need for new therapeutic candidates in bone cancer treatment. Chitosan, a common biopolymer utilized in bone and tissue engineering applications, has recently been studied as a pro-apoptotic agent in metastatic cell lines like breast cancer, but has not been utilized in bone cancer applications. In this study, chitosan was directly loaded onto HA disks to evaluate its in vitro release and effects on human fetal osteoblast (hFOB) and human osteosarcoma (MG-63) cell lines. It is hypothesized that the sustained release of chitosan will decrease osteosarcoma cell proliferation and enhance proliferation of osteoblast cells. Through morphological characterization and MTT assay analysis, chitosan showed no toxicity to human fetal osteoblast (hFOB) cells. Chitosan was also shown to decrease human osteosarcoma cell viability by up to 96% compared to control samples. This suggests a pro-apoptotic mechanism against osteosarcoma as well as the potential clinical application of chitosan as a drug candidate in ceramic scaffolds at tumor resected sites.
Collapse
|
92
|
Qian Y, Wu X, Wang H, Hou G, Han X, Song W. MicroRNA-4290 suppresses PDK1-mediated glycolysis to enhance the sensitivity of gastric cancer cell to cisplatin. ACTA ACUST UNITED AC 2020; 53:e9330. [PMID: 32321153 PMCID: PMC7184963 DOI: 10.1590/1414-431x20209330] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/10/2020] [Indexed: 02/06/2023]
Abstract
The development of chemotherapy resistance significantly impairs the efficiency of chemotherapy, but the underlying mechanisms of chemotherapy resistance in gastric cancer (GC) are complicated and still need to be further explored. Here, we aimed to reveal the effects of miR-4290/PDK1 (pyruvate dehydrogenase kinase 1) axis on chemotherapy resistance of GC in vitro. The expression patterns of miR-4290 in GC tissues and cell lines were determined by real-time quantitative PCR. Kaplan-Meier was used to assess the relationship between miR-4290 expression levels and patients' overall survival. CCK-8 and flow cytometry technologies were applied to detect cell proliferation and apoptosis. The luciferase gene reporter assay was used to evaluate the interaction between miR-4290 and PDK1. miR-4290 was lowly expressed in GC tissues and cell lines, which was closely associated with the shorter overall survival of GC patients. miR-4290 mimics significantly inhibited cell proliferation and induced cell apoptosis, as well as induced a significant reduction in the expression of PDK1. Moreover, miR-4290 significantly inhibited glycolysis and decreased the IC50 value to cisplatin in SGC7901 cells, whereas these effects were abolished and cell apoptosis was promoted when PDK1 was overexpressed. In conclusion, this study revealed that miR-4290 suppressed PDK1-mediated glycolysis to enhance the sensitivity of GC cells to cisplatin.
Collapse
Affiliation(s)
- Yan Qian
- Department of Gastric Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xu Wu
- Department of Gastric Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Haixiao Wang
- Department of Gastric Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Guowei Hou
- Department of Gastric Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xiao Han
- Department of Gastric Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Wei Song
- Department of Gastroenterology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| |
Collapse
|
93
|
Becker HM, Deitmer JW. Transport Metabolons and Acid/Base Balance in Tumor Cells. Cancers (Basel) 2020; 12:cancers12040899. [PMID: 32272695 PMCID: PMC7226098 DOI: 10.3390/cancers12040899] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023] Open
Abstract
Solid tumors are metabolically highly active tissues, which produce large amounts of acid. The acid/base balance in tumor cells is regulated by the concerted interplay between a variety of membrane transporters and carbonic anhydrases (CAs), which cooperate to produce an alkaline intracellular, and an acidic extracellular, environment, in which cancer cells can outcompete their adjacent host cells. Many acid/base transporters form a structural and functional complex with CAs, coined "transport metabolon". Transport metabolons with bicarbonate transporters require the binding of CA to the transporter and CA enzymatic activity. In cancer cells, these bicarbonate transport metabolons have been attributed a role in pH regulation and cell migration. Another type of transport metabolon is formed between CAs and monocarboxylate transporters, which mediate proton-coupled lactate transport across the cell membrane. In this complex, CAs function as "proton antenna" for the transporter, which mediate the rapid exchange of protons between the transporter and the surroundings. These transport metabolons do not require CA catalytic activity, and support the rapid efflux of lactate and protons from hypoxic cancer cells to allow sustained glycolytic activity and cell proliferation. Due to their prominent role in tumor acid/base regulation and metabolism, transport metabolons might be promising drug targets for new approaches in cancer therapy.
Collapse
Affiliation(s)
- Holger M. Becker
- Institute of Physiological Chemistry, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany
- Correspondence:
| | - Joachim W. Deitmer
- Department of Biology, University of Kaiserslautern, D-67653 Kaiserslautern, Germany;
| |
Collapse
|
94
|
Zhou Y, Li X, Lu Z, Zhang L, Dai T. Prognostic significance of red blood cell distribution width in gastrointestinal cancers: A meta-analysis. Medicine (Baltimore) 2020; 99:e19588. [PMID: 32311927 PMCID: PMC7220356 DOI: 10.1097/md.0000000000019588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Many publications showed red blood cell distribution width (RDW) might associate with the prognosis of gastrointestinal (GI) cancers, however, the agreement has not been reached because of controversial results. This meta-analysis aimed to explore the prognostic value of RDW in GI cancers. METHODS Four common databases were comprehensively searched to look for relevant studies. The meta-analyses for overall survival (OS) and disease-free survival were performed using hazard ratio (HR) and 95% confidence interval (CI), and the meta-analyses for clinical parameters were conducted using odd ratio and 95% CI. RESULTS A total of 13 studies involving with 3,509 patients with GI cancers were included into this study. The results showed, compared to patients with low RDW, patients with high RDW tended to have shorter OS (HR = 1.75, 95%CI = 1.57-1.94, P < .01) and disease-free survival (HR = 1.67, 95%CI = 1.39-2.00, P < .01). High RDW was associated with larger tumor size (P < .01), worse differentiation (P = .02), deeper invasion (P < .01), earlier lymph node metastasis (P < .01), more advanced clinical stage (P < .01) and higher carcinoembryonic antigen level (P < .01) when compared to low RDW. CONCLUSION High RDW was significantly associated with worse prognosis of GI cancers, which could be regarded as a prognostic biomarker for GI cancers. More prospective studies with large sample size and long follow-up period should be carried out to determine the prognostic significance of RDW in GI cancers in future.
Collapse
Affiliation(s)
| | - Xiding Li
- Department of General Surgery, Wuxi Second Hospital, Nanjing Medical University, Wuxi, China
| | | | | | - Tu Dai
- Department of Hepatobiliary
| |
Collapse
|
95
|
Zhang X, Wu L, Xu Y, Yu H, Chen Y, Zhao H, Lei J, Zhou Y, Zhang J, Wang J, Peng J, Jiang L, Sheng H, Li Y. Microbiota-derived SSL6 enhances the sensitivity of hepatocellular carcinoma to sorafenib by down-regulating glycolysis. Cancer Lett 2020; 481:32-44. [PMID: 32246956 DOI: 10.1016/j.canlet.2020.03.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 03/18/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023]
Abstract
Enhancing the sensitivity of hepatocellular carcinoma (HCC) cells to sorafenib (SFN) is an essential clinical bottleneck to be solved. Here we report that the expression of CD47 negatively correlated with HCC sensitivity to SFN. The microbiota-derived Staphylococcal superantigen-like protein 6 (SSL6) inhibited CD47 and promoted SFN-induced apoptosis of HCC cells Huh-7 and MHCC97H. Mechanistically, the sensitivity of HCC cells to SFN was inhibited by elevated Warburg effect (glycolysis), and SSL6 down-regulated PI3K/Akt-mediated glycolysis by blocking CD47. Knockdown of CD47 also dampened glycolysis and sensitized HCC cells to SFN. Moreover, SFN-resistant HCC cells exhibited enhanced glycolysis and CD47 expression. SSL6 significantly re-sensitized the resistant HCC cells to SFN. More importantly, we identified the anti-tumor effect of SSL6 in combination with SFN in HCC-bearing mice. Our results clarify the mechanism by which SSL6 enhances SFN sensitivity in HCC cells, providing a molecular basis for combination targeted therapy with microbiota-derived SSL6 to treat HCC.
Collapse
Affiliation(s)
- Xiao Zhang
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Lei Wu
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yanquan Xu
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Hua Yu
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yu Chen
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Huakan Zhao
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Juan Lei
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yu Zhou
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Jiangang Zhang
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Jingchun Wang
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Jin Peng
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Lu Jiang
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Halei Sheng
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yongsheng Li
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
96
|
Le NH, Ye G, Peng C, Chen JIL. Metabolic mapping with plasmonic nanoparticle assemblies. Analyst 2020; 145:2586-2594. [PMID: 32182299 DOI: 10.1039/c9an02262g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A rapid and simple methodology for the biomolecular analysis of single cells and microenvironments via a stick-and-peel plasmonic sensing platform is reported. Substrate-bound assemblies of plasmonic gold nanoparticles linked by reconfigurable oligonucleotides undergo disassembly upon target binding. Changes in the light scattering intensity of thousands of discrete nanoparticle assemblies are extrapolated concomitantly to yield the mapping of local target concentrations. The methodology is completely free of labelling, purification and separation steps. We quantified the intracellular ATP levels for two ovarian cancer cell lines to elucidate the differences and cellular distribution, and demonstrated the potential of the stick-and-peel platform for mapping the microenvironment of a 2D heterogeneous surface. The portable and economical analytical platform may broaden the affordability and applicability of single-cell based analyses and enable new opportunities in clinical care such as on-site molecular pathology.
Collapse
Affiliation(s)
- Nguyen H Le
- Department of Chemistry, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada.
| | | | | | | |
Collapse
|
97
|
Sadeghi M, Ordway B, Rafiei I, Borad P, Fang B, Koomen JL, Zhang C, Yoder S, Johnson J, Damaghi M. Integrative Analysis of Breast Cancer Cells Reveals an Epithelial-Mesenchymal Transition Role in Adaptation to Acidic Microenvironment. Front Oncol 2020; 10:304. [PMID: 32211331 PMCID: PMC7076123 DOI: 10.3389/fonc.2020.00304] [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: 11/03/2019] [Accepted: 02/20/2020] [Indexed: 01/06/2023] Open
Abstract
Early ducts of breast tumors are unequivocally acidic. High rates of glycolysis combined with poor perfusion lead to a congestion of acidic metabolites in the tumor microenvironment, and pre-malignant cells must adapt to this acidosis to thrive. Adaptation to acidosis selects cancer cells that can thrive in harsh conditions and are capable of outgrowing the normal or non-adapted neighbors. This selection is usually accompanied by phenotypic change. Epithelial mesenchymal transition (EMT) is one of the most important switches correlated to malignant tumor cell phenotype and has been shown to be induced by tumor acidosis. New evidence shows that the EMT switch is not a binary system and occurs on a spectrum of transition states. During confirmation of the EMT phenotype, our results demonstrated a partial EMT phenotype in our acid-adapted cell population. Using RNA sequencing and network analysis we found 10 dysregulated network motifs in acid-adapted breast cancer cells playing a role in EMT. Our further integrative analysis of RNA sequencing and SILAC proteomics resulted in recognition of S100B and S100A6 proteins at both the RNA and protein level. Higher expression of S100B and S100A6 was validated in vitro by Immunocytochemistry. We further validated our finding both in vitro and in patients' samples by IHC analysis of Tissue Microarray (TMA). Correlation analysis of S100A6 and LAMP2b as marker of acidosis in each patient from Moffitt TMA approved the acid related role of S100A6 in breast cancer patients. Also, DCIS patients with higher expression of S100A6 showed lower survival compared to lower expression. We propose essential roles of acid adaptation in cancer cells EMT process through S100 proteins such as S100A6 that can be used as therapeutic strategy targeting both acid-adapted and malignant phenotypes.
Collapse
Affiliation(s)
- Mehdi Sadeghi
- Department of Cell and Molecular Biology, Faculty of Science, Semnan University, Semnan, Iran
| | - Bryce Ordway
- Department of Cancer Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Ilyia Rafiei
- Department of Cell and Molecular Biology, Faculty of Science, Semnan University, Semnan, Iran
| | - Punit Borad
- Department of Cancer Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Bin Fang
- Proteomics Core, Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - John L Koomen
- Proteomics Core, Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Chaomei Zhang
- Molecular Biology Core, Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Sean Yoder
- Molecular Biology Core, Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Joseph Johnson
- Microscopy Core, Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Mehdi Damaghi
- Department of Cancer Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, United States.,Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
98
|
Shin MH, Kim J, Lim SA, Kim J, Kim SJ, Lee KM. NK Cell-Based Immunotherapies in Cancer. Immune Netw 2020; 20:e14. [PMID: 32395366 PMCID: PMC7192832 DOI: 10.4110/in.2020.20.e14] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/01/2020] [Accepted: 03/01/2020] [Indexed: 12/11/2022] Open
Abstract
With the development of technologies that can transform immune cells into therapeutic modalities, immunotherapy has remarkably changed the current paradigm of cancer treatment in recent years. NK cells are components of the innate immune system that act as key regulators and exhibit a potent tumor cytolytic function. Unlike T cells, NK cells exhibit tumor cytotoxicity by recognizing non-self, without deliberate immunization or activation. Currently, researchers have developed various approaches to improve the number and anti-tumor function of NK cells. These approaches include the use of cytokines and Abs to stimulate the efficacy of NK cell function, adoptive transfer of autologous or allogeneic ex vivo expanded NK cells, establishment of homogeneous NK cell lines using the NK cells of patients with cancer or healthy donors, derivation of NK cells from induced pluripotent stem cells (iPSCs), and modification of NK cells with cutting-edge genetic engineering technologies to generate chimeric Ag receptor (CAR)-NK cells. Such NK cell-based immunotherapies are currently reported as being promising anti-tumor strategies that have shown enhanced functional specificity in several clinical trials investigating malignant tumors. Here, we summarize the recent advances in NK cell-based cancer immunotherapies that have focused on providing improved function through the use of the latest genetic engineering technologies. We also discuss the different types of NK cells developed for cancer immunotherapy and present the clinical trials being conducted to test their safety and efficacy.
Collapse
Affiliation(s)
- Min Hwa Shin
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea
| | - Junghee Kim
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea
| | - Siyoung A Lim
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea
| | - Jungwon Kim
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea
| | - Seong-Jin Kim
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Korea
| | - Kyung-Mi Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea
| |
Collapse
|
99
|
Chen ZX, Liu MD, Guo DK, Zou MZ, Wang SB, Cheng H, Zhong Z, Zhang XZ. A MSN-based tumor-targeted nanoplatform to interfere with lactate metabolism to induce tumor cell acidosis for tumor suppression and anti-metastasis. NANOSCALE 2020; 12:2966-2972. [PMID: 31971210 DOI: 10.1039/c9nr10344a] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Lactate, the main contributor to the acidic tumor microenvironment, not only promotes the proliferation of tumor cells, but also closely relates to tumor invasion and metastasis. Here, a tumor targeting nanoplatform, designated as Me&Flu@MSN@MnO2-FA, was fabricated for effective tumor suppression and anti-metastasis by interfering with lactate metabolism of tumor cells. Metformin (Me) and fluvastatin sodium (Flu) were incorporated into MnO2-coated mesoporous silicon nanoparticles (MSNs), the synergism between Me and Flu can modulate the pyruvate metabolic pathway to produce more lactate, and concurrently inhibit lactate efflux to induce intracellular acidosis to kill tumor cells. As a result of the restricted lactate efflux, the extracellular lactate concentration is reduced, and the ability of the tumor cells to migrate is also weakened. This ingenious strategy based on Me&Flu@MSN@MnO2-FA showed an obvious inhibitory effect on tumor growth and resistance to metastasis.
Collapse
Affiliation(s)
- Zhao-Xia Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Miao-Deng Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Deng-Ke Guo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Mei-Zhen Zou
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Shi-Bo Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Han Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Zhenlin Zhong
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| |
Collapse
|
100
|
Strobl MAR, Krause AL, Damaghi M, Gillies R, Anderson ARA, Maini PK. Mix and Match: Phenotypic Coexistence as a Key Facilitator of Cancer Invasion. Bull Math Biol 2020; 82:15. [PMID: 31953602 PMCID: PMC6968991 DOI: 10.1007/s11538-019-00675-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/03/2019] [Indexed: 01/10/2023]
Abstract
Invasion of healthy tissue is a defining feature of malignant tumours. Traditionally, invasion is thought to be driven by cells that have acquired all the necessary traits to overcome the range of biological and physical defences employed by the body. However, in light of the ever-increasing evidence for geno- and phenotypic intra-tumour heterogeneity, an alternative hypothesis presents itself: could invasion be driven by a collection of cells with distinct traits that together facilitate the invasion process? In this paper, we use a mathematical model to assess the feasibility of this hypothesis in the context of acid-mediated invasion. We assume tumour expansion is obstructed by stroma which inhibits growth and extra-cellular matrix (ECM) which blocks cancer cell movement. Further, we assume that there are two types of cancer cells: (i) a glycolytic phenotype which produces acid that kills stromal cells and (ii) a matrix-degrading phenotype that locally remodels the ECM. We extend the Gatenby-Gawlinski reaction-diffusion model to derive a system of five coupled reaction-diffusion equations to describe the resulting invasion process. We characterise the spatially homogeneous steady states and carry out a simulation study in one spatial dimension to determine how the tumour develops as we vary the strength of competition between the two phenotypes. We find that overall tumour growth is most extensive when both cell types can stably coexist, since this allows the cells to locally mix and benefit most from the combination of traits. In contrast, when inter-species competition exceeds intra-species competition the populations spatially separate and invasion arrests either: (i) rapidly (matrix-degraders dominate) or (ii) slowly (acid-producers dominate). Overall, our work demonstrates that the spatial and ecological relationship between a heterogeneous population of tumour cells is a key factor in determining their ability to cooperate. Specifically, we predict that tumours in which different phenotypes coexist stably are more invasive than tumours in which phenotypes are spatially separated.
Collapse
Affiliation(s)
- Maximilian A. R. Strobl
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Radcliffe Observatory Quarter, OX2 6GG Oxford, UK
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Magnolia Drive, Tampa, 12902 USA
| | - Andrew L. Krause
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Radcliffe Observatory Quarter, OX2 6GG Oxford, UK
| | - Mehdi Damaghi
- Department of Cancer Physiology, Moffitt Cancer Center, Magnolia Drive, Tampa, 12902 USA
| | - Robert Gillies
- Department of Cancer Physiology, Moffitt Cancer Center, Magnolia Drive, Tampa, 12902 USA
| | - Alexander R. A. Anderson
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Magnolia Drive, Tampa, 12902 USA
| | - Philip K. Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Radcliffe Observatory Quarter, OX2 6GG Oxford, UK
| |
Collapse
|