301
|
Mirabile A, Rivoltini L, Daveri E, Vernieri C, Mele R, Porcu L, Lazzari C, Bulotta A, Viganò MG, Cascinu S, Gregorc V. Metabolism and Immune Modulation in Patients with Solid Tumors: Systematic Review of Preclinical and Clinical Evidence. Cancers (Basel) 2020; 12:E1153. [PMID: 32375310 PMCID: PMC7281426 DOI: 10.3390/cancers12051153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Several immunotherapy agents are the standard of care of many solid malignancies. Nevertheless, the majority of patients do not benefit from the currently available immunotherapies. It is therefore of paramount importance to identify the prognostic and predictive factors of tumor response/resistance and to design effective therapeutic strategies to overcome primary resistance and improve the efficacy of immunotherapy. The aim of this review is to underline the influence of the tumor and host metabolism on the antitumor immune response and to discuss possible strategies to improve the efficacy of available treatments by targeting the specific metabolic pathways in tumors or immune cells and by modifying patients' nutritional statuses. A systematic search of the Medline and EMBASE databases was carried out to identify scientific papers published until February 2020, which reported original research articles on the influence of tumor or host metabolism on antitumor immune response. The literature data showed the key role of glycolysis and mitochondrial oxidative phosphorylation, arginine, tryptophan, glutamine, lipid metabolism and microbiome on immune cell function. Moreover, specific nutritional behaviors, such as a low dietary intake of vitamin C, low glycemic index and alpha-linolenic acid, eicosapentenoic acid, docosahexaenoic acid, ornithine ketoglutarate, tryptophan and probiotic supplementation were associated with the potential clinical benefits from the currently available immunotherapies.
Collapse
Affiliation(s)
- Aurora Mirabile
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Licia Rivoltini
- Immunotherapy of Human Tumors, IRCCS National Cancer Institute (INT) and University of Milan, Via Venezian 1, 20133 Milan, Italy; (L.R.); (E.D.)
| | - Elena Daveri
- Immunotherapy of Human Tumors, IRCCS National Cancer Institute (INT) and University of Milan, Via Venezian 1, 20133 Milan, Italy; (L.R.); (E.D.)
| | - Claudio Vernieri
- Medical Oncology Department, IRCCS IRCCS National Cancer Institute (INT) and University of Milan, Via Venezian 1, 20133 Milan, Italy;
- IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Roberto Mele
- Nutritionist biologist, Hospital Health Direction, Scientific Institute San Raffaele Hospital, Via Olgettina 60, 20132 Milan, Italy;
| | - Luca Porcu
- Methodological Research Unit, Institute of Pharmacological Research Mario Negri, Via Mario Negri 2, 20156 Milan, Italy;
| | - Chiara Lazzari
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Alessandra Bulotta
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Maria Grazia Viganò
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Stefano Cascinu
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Vanesa Gregorc
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| |
Collapse
|
302
|
Hopkins BD, Goncalves MD, Cantley LC. Insulin-PI3K signalling: an evolutionarily insulated metabolic driver of cancer. Nat Rev Endocrinol 2020; 16:276-283. [PMID: 32127696 PMCID: PMC7286536 DOI: 10.1038/s41574-020-0329-9] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2020] [Indexed: 12/17/2022]
Abstract
Cancer is driven by incremental changes that accumulate, eventually leading to oncogenic transformation. Although genetic alterations dominate the way cancer biologists think about oncogenesis, growing evidence suggests that systemic factors (for example, insulin, oestrogen and inflammatory cytokines) and their intracellular pathways activate oncogenic signals and contribute to targetable phenotypes. Systemic factors can have a critical role in both tumour initiation and therapeutic responses as increasingly targeted and personalized therapeutic regimens are used to treat patients with cancer. The endocrine system controls cell growth and metabolism by providing extracellular cues that integrate systemic nutrient status with cellular activities such as proliferation and survival via the production of metabolites and hormones such as insulin. When insulin binds to its receptor, it initiates a sequence of phosphorylation events that lead to activation of the catalytic activity of phosphoinositide 3-kinase (PI3K), a lipid kinase that coordinates the intake and utilization of glucose, and mTOR, a kinase downstream of PI3K that stimulates transcription and translation. When chronically activated, the PI3K pathway can drive malignant transformation. Here, we discuss the insulin-PI3K signalling cascade and emphasize its roles in normal cells (including coordinating cell metabolism and growth), highlighting the features of this network that make it ideal for co-option by cancer cells. Furthermore, we discuss how this signalling network can affect therapeutic responses and how novel metabolic-based strategies might enhance treatment efficacy for cancer.
Collapse
Affiliation(s)
- Benjamin D Hopkins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Marcus D Goncalves
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
303
|
Affiliation(s)
- Ali Tootee
- 1Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- 2Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
304
|
Chiodi I, Mondello C. Life style factors, tumor cell plasticity and cancer stem cells. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 784:108308. [PMID: 32430096 DOI: 10.1016/j.mrrev.2020.108308] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022]
Abstract
Cancers are heterogeneous tissues and a layer of heterogeneity is determined by the presence of cells showing stemness traits, known as cancer stem cells (CSCs). Evidence indicates that CSCs are important players in tumor development, progression and relapse. Oncogenic transformation of normal stem cells can give rise to CSCs, but CSCs can also originate from de-differentiation of bulk tumor cells. Thus, factors promoting the increase of normal stem cell pools or stimulating the acquisition of stemness features by tumor cells can have serious consequences on cancer origin and progression. In this review, we will first give an overview of the CSC model of cancer development and we will then discuss the role of life style factors, such as high caloric diet, alcohol drinking and smoking, on the widening of stem cell pools and the induction of CSC features in tumors. Finally, we will discuss some healthy life style factors that can help to prevent cancer.
Collapse
Affiliation(s)
- Ilaria Chiodi
- Istituto di Genetica Molecolare L. L. Cavalli-Sforza, CNR, via Abbiategrasso 207, 27100, Pavia, Italy
| | - Chiara Mondello
- Istituto di Genetica Molecolare L. L. Cavalli-Sforza, CNR, via Abbiategrasso 207, 27100, Pavia, Italy.
| |
Collapse
|
305
|
Weng ML, Chen WK, Chen XY, Lu H, Sun ZR, Yu Q, Sun PF, Xu YJ, Zhu MM, Jiang N, Zhang J, Zhang JP, Song YL, Ma D, Zhang XP, Miao CH. Fasting inhibits aerobic glycolysis and proliferation in colorectal cancer via the Fdft1-mediated AKT/mTOR/HIF1α pathway suppression. Nat Commun 2020; 11:1869. [PMID: 32313017 PMCID: PMC7170903 DOI: 10.1038/s41467-020-15795-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Evidence suggests that fasting exerts extensive antitumor effects in various cancers, including colorectal cancer (CRC). However, the mechanism behind this response is unclear. We investigate the effect of fasting on glucose metabolism and malignancy in CRC. We find that fasting upregulates the expression of a cholesterogenic gene, Farnesyl-Diphosphate Farnesyltransferase 1 (FDFT1), during the inhibition of CRC cell aerobic glycolysis and proliferation. In addition, the downregulation of FDFT1 is correlated with malignant progression and poor prognosis in CRC. Moreover, FDFT1 acts as a critical tumor suppressor in CRC. Mechanistically, FDFT1 performs its tumor-inhibitory function by negatively regulating AKT/mTOR/HIF1α signaling. Furthermore, mTOR inhibitor can synergize with fasting in inhibiting the proliferation of CRC. These results indicate that FDFT1 is a key downstream target of the fasting response and may be involved in CRC cell glucose metabolism. Our results suggest therapeutic implications in CRC and potential crosstalk between a cholesterogenic gene and glycolysis. The molecular mechanisms underpinning how fasting inhibits tumourigenesis are not completely elucidated. Here, the authors show that fasting upregulates the cholesterogenic gene FDFT1 which leads to decreased AKT/mTOR/HIF1a signalling and glycolysis reduction in colorectal cancer.
Collapse
Affiliation(s)
- Mei-Lin Weng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wan-Kun Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiang-Yuan Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hong Lu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Rong Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qi Yu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peng-Fei Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ya-Jun Xu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Min-Min Zhu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Nan Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Science, School of Basic Medical Science, Fudan University, Shanghai, 200032, China.,Institute of Biomedical Science, Fudan University, Shanghai, 200032, China
| | - Jin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Science, School of Basic Medical Science, Fudan University, Shanghai, 200032, China.,Institute of Biomedical Science, Fudan University, Shanghai, 200032, China
| | - Jian-Ping Zhang
- Institute of Modern Physics, Fudan University; Department of Nuclear Medicine, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuan-Lin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institutes of Biomedical Science, School of Basic Medical Science, Fudan University, Shanghai, 200032, China. .,Institute of Biomedical Science, Fudan University, Shanghai, 200032, China. .,Children's Hospital, Fudan University, Shanghai, 200032, China.
| | - Xiao-Ping Zhang
- The Institute of Intervention Vessel, Tongji University School of Medicine, Shanghai, 200092, China. .,Shanghai Center of Thyroid Diseases, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Chang-Hong Miao
- Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
306
|
Gerisch B, Tharyan RG, Mak J, Denzel SI, Popkes-van Oepen T, Henn N, Antebi A. HLH-30/TFEB Is a Master Regulator of Reproductive Quiescence. Dev Cell 2020; 53:316-329.e5. [PMID: 32302543 DOI: 10.1016/j.devcel.2020.03.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 01/28/2020] [Accepted: 03/15/2020] [Indexed: 12/20/2022]
Abstract
All animals have evolved the ability to survive nutrient deprivation, and nutrient signaling pathways are conserved modulators of health and disease. In C. elegans, late-larval starvation provokes the adult reproductive diapause (ARD), a long-lived quiescent state that enables survival for months without food, yet underlying molecular mechanisms remain unknown. Here, we show that ARD is distinct from other forms of diapause, showing little requirement for canonical longevity pathways, autophagy, and fat metabolism. Instead it requires the HLH-30/TFEB transcription factor to promote the morphological and physiological remodeling involved in ARD entry, survival, and recovery, suggesting that HLH-30 is a master regulator of reproductive quiescence. HLH-30 transcriptome and genetic analyses reveal that Max-like HLH factors, AMP-kinase, mTOR, protein synthesis, and mitochondrial fusion are target processes that promote ARD longevity. ARD thus rewires metabolism to ensure long-term survival and may illuminate similar mechanisms acting in stem cell quiescence and long-term fasting.
Collapse
Affiliation(s)
- Birgit Gerisch
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Rebecca George Tharyan
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Jennifer Mak
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Sarah I Denzel
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne 50931, Germany
| | - Till Popkes-van Oepen
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne 50931, Germany
| | - Nadine Henn
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Adam Antebi
- Department of Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne 50931, Germany.
| |
Collapse
|
307
|
Krstic J, Pieber TR, Prokesch A. Stratifying nutritional restriction in cancer therapy: Next stop, personalized medicine. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 354:231-259. [PMID: 32475475 DOI: 10.1016/bs.ircmb.2020.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dietary interventions combined with cancer drugs represent a clinically valid polytherapy. In particular nutrient restriction (NR) in the form of varied fasting or caloric restriction regimens holds great clinical promise, conceptually due to the voracious anabolic appetite of cancer cells. This metabolic dependency is driven by a strong selective pressure to increasingly acquire biomass of a proliferating tumor and can be therapeutically exploited as vulnerability. A host of preclinical data suggest that NR can potentiate the efficacy of, or alleviate resistance to, cancer drugs. However, complicating clinical implementation are the many variables involved, such as host biology, cancer stage and type, oncogenic mutation landscape, tumor heterogeneity, variations in treatment modalities, and patient compliance to NR protocols. This calls for systematic preclinical screens and co-clinical studies to predict effective combinations of NR with cancer drugs and to allow for patient stratification regarding responsiveness to polytherapy. Such screen-and-stratify pipelines should consider tumor heterogeneity as well as the role of immune effectors in the tumor microenvironment and may lead to biomarker discovery advancing the oncology field toward personalized options with improved translatability to clinical settings. This opinion-based review provides a critical overview of recent literature investigating NR for cancer treatment, pinpoints limitations of current studies, and suggests standardizations and refinements for future studies and trials. The proposed measures aim to increase the translational value of preclinical data and effectively harness the vast potential of NR as adjuvant for cancer therapy.
Collapse
Affiliation(s)
- Jelena Krstic
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism & Aging, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Thomas R Pieber
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria; Health Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Andreas Prokesch
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism & Aging, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
308
|
Goudarzi A, Hosseinmardi N, Salami S, Mehdikhani F, Derakhshan S, Aminishakib P. Starvation promotes histone lysine butyrylation in the liver of male but not female mice. Gene 2020; 745:144647. [PMID: 32247738 DOI: 10.1016/j.gene.2020.144647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/18/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023]
Abstract
AIMS Post-translational modifications (PTMs) of histones are regulated by the availability of their respective acyl-CoAs. Among these histone PTMs, the metabolic origin of histone butyrylation (Kbu) is still poorly understood. MATERIAL AND METHODS The impact of starvation on the levels of Kbu was determined by western blotting on histones extracted from the liver of fed and fasted C57BL/6 mice and immunohistochemistry on liver paraffin sections. KEY FINDINGS Using animal model we provide evidence that the stimulation of ketogenesis following starvation, in addition to histone beta-hydroxybutyrylation (Kbhb), also leads to an increase in histone butyrylation (Kbu). Using an immunohistochemistry (IHC) approach we report first that hepatocytes contained butyrylated histones with important cell-to-cell heterogeneity. More importantly, our investigations based on western blotting and IHC also proposed that the basal levels of Kbu differ between male and female mice, with female mouse hepatocytes containing higher levels of butyrylated histones. Starvation enhanced solely histone Kbu levels in the liver of males but not females. SIGNIFICANCE This is the first demonstration of a sex-dependent large-scale stimulation of histone acylation. Our data also point to different basal metabolic conditions of the male and female liver cells with a sex-dependent impact on the hepatocytes' epigenome.
Collapse
Affiliation(s)
- Afsaneh Goudarzi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Narges Hosseinmardi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siamak Salami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mehdikhani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Derakhshan
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Pouyan Aminishakib
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
309
|
Yang B, Chen Y, Shi J. Tumor‐Specific Chemotherapy by Nanomedicine‐Enabled Differential Stress Sensitization. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Bowen Yang
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of CeramicsChinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of CeramicsChinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of CeramicsChinese Academy of Sciences Shanghai 200050 P. R. China
| |
Collapse
|
310
|
Yang B, Chen Y, Shi J. Tumor-Specific Chemotherapy by Nanomedicine-Enabled Differential Stress Sensitization. Angew Chem Int Ed Engl 2020; 59:9693-9701. [PMID: 32162453 DOI: 10.1002/anie.202002306] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/09/2020] [Indexed: 12/31/2022]
Abstract
Most of current nanomedicines are administrated intravenously to favour tumor accumulation through enhanced permeability and retention (EPR) effect, which, however, suffers from several drawbacks such as low drug bioavailability and severe side effect. In this work, we have constructed a doxorubicin(Dox)-based liposomal nanosystem for tumor-specific chemotherapy, by enabling differential stress sensitization between cancer and normal cells for restricting the chemodrug toxicity exclusively in tumor regions. 2-Deoxy-D-glucose (2DG) was loaded in the nanoliposome to inhibit glycolysis of cancer cells, which works in synergy with the co-loaded chemodrug Dox to promote mitochondrial depolarization and subsequent apoptosis. In addition, the starvation effect of 2DG can counteract the toxicity of Dox in normal cells and thus mitigates the harmful side effect of chemotherapy. It is expected that such a differential stress sensitization strategy may greatly benefit future nanomedicine design.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
311
|
Dietary modifications for enhanced cancer therapy. Nature 2020; 579:507-517. [DOI: 10.1038/s41586-020-2124-0] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 01/27/2020] [Indexed: 02/07/2023]
|
312
|
Effects of fasting on the expression pattern of FGFs in different skeletal muscle fibre types and sexes in mice. Biol Sex Differ 2020; 11:9. [PMID: 32156311 PMCID: PMC7063800 DOI: 10.1186/s13293-020-00287-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
Fibroblast growth factors (FGFs) belong to a large family comprising 22 FGF polypeptides that are widely expressed in tissues. Most of the FGFs can be secreted and involved in the regulation of skeletal muscle function and structure. However, the role of fasting on FGF expression pattern in skeletal muscles remains unknown. In this study, we combined bioinformatics analysis and in vivo studies to explore the effect of 24-h fasting on the expression of Fgfs in slow-twitch soleus and fast-twitch tibialis anterior (TA) muscle from male and female C57BL/6 mice. We found that fasting significantly affected the expression of many Fgfs in mouse skeletal muscle. Furthermore, skeletal muscle fibre type and sex also influenced Fgf expression and response to fasting. We observed that in both male and female mice fasting reduced Fgf6 and Fgf11 in the TA muscle rather than the soleus. Moreover, fasting reduced Fgf8 expression in the soleus and TA muscles in female mice rather than in male mice. Fasting also increased Fgf21 expression in female soleus muscle and female and male plasma. Fasting reduced Fgf2 and Fgf18 expression levels without fibre-type and sex-dependent effects in mice. We further found that fasting decreased the expression of an FGF activation marker gene-Flrt2 in the TA muscle but not in the soleus muscle in both male and female mice. This study revealed the expression profile of Fgfs in different skeletal muscle fibre types and different sexes and provides clues to the interaction between the skeletal muscle and other organs, which deserves future investigations.
Collapse
|
313
|
Dart A. Starving tumours. Nat Rev Cancer 2020; 20:141. [PMID: 32029923 DOI: 10.1038/s41568-020-0244-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
314
|
Lanser L, Kink P, Egger EM, Willenbacher W, Fuchs D, Weiss G, Kurz K. Inflammation-Induced Tryptophan Breakdown is Related With Anemia, Fatigue, and Depression in Cancer. Front Immunol 2020; 11:249. [PMID: 32153576 PMCID: PMC7047328 DOI: 10.3389/fimmu.2020.00249] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
Many patients with cancer suffer from anemia, depression, and an impaired quality of life (QoL). These patients often also show decreased plasma tryptophan levels and increased kynurenine concentrations in parallel with elevated concentrations of Th1 type immune activation marker neopterin. In the course of anti-tumor immune response, the pro-inflammatory cytokine interferon gamma (IFN-γ) induces both, the enzyme indoleamine 2,3-dioxygenase (IDO) to degrade tryptophan and the enzyme GTP-cyclohydrolase I to form neopterin. High neopterin concentrations as well as an increased kynurenine to tryptophan ratio (Kyn/Trp) in the blood of cancer patients are predictive for a worse outcome. Inflammation-mediated tryptophan catabolism along the kynurenine pathway is related to fatigue and anemia as well as to depression and a decreased QoL in patients with solid tumors. In fact, enhanced tryptophan breakdown might greatly contribute to the development of anemia, fatigue, and depression in cancer patients. IDO activation and stimulation of the kynurenine pathway exert immune regulatory mechanisms, which may impair anti-tumor immune responses. In addition, tumor cells can degrade tryptophan to weaken immune responses directed against them. High IDO expression in the tumor tissue is associated with a poor prognosis of patients. The efficiency of IDO-inhibitors to inhibit cancer progression is currently tested in combination with established chemotherapies and with immune checkpoint inhibitors. Inflammation-mediated tryptophan catabolism and its possible influence on the development and persistence of anemia, fatigue, and depression in cancer patients are discussed.
Collapse
Affiliation(s)
- Lukas Lanser
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Patricia Kink
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Eva Maria Egger
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Wolfgang Willenbacher
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
- Oncotyrol Centre for Personalized Cancer Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Katharina Kurz
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
315
|
Hersi F, Omar HA, Al-Qawasmeh RA, Ahmad Z, Jaber AM, Zaher DM, Al-Tel TH. Design and synthesis of new energy restriction mimetic agents: Potent anti-tumor activities of hybrid motifs of aminothiazoles and coumarins. Sci Rep 2020; 10:2893. [PMID: 32076009 PMCID: PMC7031302 DOI: 10.1038/s41598-020-59685-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 02/03/2020] [Indexed: 11/09/2022] Open
Abstract
The incidence of obesity-related diseases like diabetes, cardiovascular diseases, and different types of cancers shed light on the importance of dietary control as preventive and treatment measures. However, long-term dietary control is challenging to achieve in most individuals. The use of energy restriction mimetic agents (ERMAs) as an alternative approach to affect the energy machinery of cancer cells has emerged as a promising approach for cancer therapy. ERMAs limit the high need for energy in rapidly growing tumor cells, with their survival rate strongly dependent on the robust availability of energy. In this context, initial phenotypic screening of an in-house pilot compound library identified a new class of aminothiazole anchored on coumarin scaffold as potent anticancer lead drug candidates with potential activity as ERMA. The identified chemotypes were able to inhibit glucose uptake and increase ROS content in cancer cells. Compounds 9b, 9c, 9i, 11b, and 11c were highly active against colorectal cancer cell lines, HCT116 and HT-29, with half-maximal inhibitory concertation (IC50) range from 0.25 to 0.38 µM. Further biological evaluations of 9b and 9f using Western blotting, caspase activity, glucose uptake, ROS production, and NADPH/NADP levels revealed the ability of these lead drug candidates to induce cancer cell death via, at least in part, energy restriction. Moreover, the assessment of 9b and 9f synergistic activity with cisplatin showed promising outcomes. The current work highlights the significant potential of the lead compounds, 9b, and 9f as potential anticancer agents via targeting the cellular energy machinery in cancer cells.
Collapse
Affiliation(s)
- Fatema Hersi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates.,College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates. .,College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates. .,Department of Pharmacology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Raed A Al-Qawasmeh
- Department of Chemistry, Faculty of Science, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Zainab Ahmad
- Department of Chemistry, Faculty of Science, The University of Jordan, Amman, 11942, Jordan
| | - Areej M Jaber
- Department of Chemistry, Faculty of Science, The University of Jordan, Amman, 11942, Jordan
| | - Dana M Zaher
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates.,College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Taleb H Al-Tel
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates. .,College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
316
|
Holma R, Laatikainen R, Orell H, Joensuu H, Peuhkuri K, Poussa T, Korpela R, Österlund P. Consumption of Lactose, Other FODMAPs and Diarrhoea during Adjuvant 5-Fluorouracil Chemotherapy for Colorectal Cancer. Nutrients 2020; 12:nu12020407. [PMID: 32033152 PMCID: PMC7071323 DOI: 10.3390/nu12020407] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy-induced mucosal injury of the small intestine may interfere with the enzymes and transporters responsible for the hydrolysis and absorption of dietary carbohydrates causing diarrhoea, abdominal discomfort and pain. The aim of this study was to investigate the association between the consumption of foods rich in FODMAPs (fermentable oligo-, di- and monosaccharides and polyols) and gastrointestinal symptoms in patients receiving adjuvant therapy for colorectal cancer. The patients (n = 52) filled in a 4-day food diary at baseline and during therapy and kept a symptom diary. The intakes of FODMAP-rich foods were calculated as portions and the intakes were divided into two consumption categories. Patients with high consumption of FODMAP-rich foods had diarrhoea more frequently than those with low consumption (for lactose-rich foods the odds ratio (OR) was 2.63, P = 0.03; and for other FODMAP-rich foods 1.82, P = 0.20). Patients with high consumption of both lactose-rich and other FODMAP-rich foods had an over 4-fold risk of developing diarrhoea as compared to those with low consumption of both (OR, 4.18; P = 0.02). These results were confirmed in multivariate models. Conclusion: Consumption of lactose-rich foods results in an increased risk of diarrhoea during adjuvant therapy for colorectal cancer, especially when the consumption of other FODMAP-rich foods is also high.
Collapse
Affiliation(s)
- Reetta Holma
- Faculty of Medicine, Pharmacology, Medical Nutrition Physiology and Human Microbe Research Program, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland; (R.H.); (R.L.); (R.K.)
| | - Reijo Laatikainen
- Faculty of Medicine, Pharmacology, Medical Nutrition Physiology and Human Microbe Research Program, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland; (R.H.); (R.L.); (R.K.)
- Booston Oy Ltd., Viikinkaari 6, FI-00790 Helsinki, Finland
| | - Helena Orell
- Department of Clinical Nutrition Therapy, Helsinki University Hospital, P.O. Box 100, FI-00029 HUS, Finland;
| | - Heikki Joensuu
- Department of Oncology, Helsinki University Hospital and University of Helsinki, P.O. Box 180, FI-00029 HUS, Finland;
| | - Katri Peuhkuri
- Social Services and Health Care Division, P.O. Box 6230, FI-00099 City of Helsinki, Finland;
| | - Tuija Poussa
- STAT-Consulting, Vahverokatu 6, FI-37130 Nokia, Finland;
| | - Riitta Korpela
- Faculty of Medicine, Pharmacology, Medical Nutrition Physiology and Human Microbe Research Program, University of Helsinki, P.O. Box 63, FI-00014 Helsinki, Finland; (R.H.); (R.L.); (R.K.)
| | - Pia Österlund
- Department of Oncology, Helsinki University Hospital and University of Helsinki, P.O. Box 180, FI-00029 HUS, Finland;
- Department of Oncology, Tampere University Hospital and Tampere university, P.O. Box 2000, FI-33521 Tampere, Finland
- Correspondence: or or ; Tel.: +358-9-4711
| |
Collapse
|
317
|
Kong WJ, Vernieri C, Foiani M, Jiang JD. Berberine in the treatment of metabolism-related chronic diseases: A drug cloud (dCloud) effect to target multifactorial disorders. Pharmacol Ther 2020; 209:107496. [PMID: 32001311 DOI: 10.1016/j.pharmthera.2020.107496] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/17/2020] [Indexed: 12/23/2022]
Abstract
Berberine (BBR) is a multi-target drug (MTD) that has proven effective in the treatment of metabolism-related chronic diseases (CDs). However, the mode of action (MOA) of BBR remains to be clarified. At a cellular level, the inhibitory effect of BBR on mitochondrial enzymes is probably responsible for many of its biological activities, including the activation of low-density lipoprotein receptor (LDLR), AMP-activated protein kinase (AMPK) and insulin receptor (InsR); these biological activities contribute to ameliorate peripheral blood metabolic profiles, e.g. by reducing plasma lipids and glucose levels, thus improving signs and symptoms of metabolic disorders. In this perspective, BBR acts as a targeted therapy. However, it also exerts pleiotropic systemic activities on some root causes of CDs that include antioxidant / anti-inflammatory effects and modifications of gut microbiota composition and metabolism, which may also contribute to its disease-modifying effects. After reviewing the different MOA of BBR, here we propose that BBR acts through a drug-cloud (dCloud) mechanism, as different to a drug-target effect. The dCloud here is defined as a group of terminal molecular events induced by the drug (or/and related metabolites), as well as the network connections among them. In this scenario, the therapeutic efficacy of BBR is the result of its dCloud effect acting on symptoms/signs as well as on root causes of the diseases. The dCloud concept is applicable to other established MTDs, such as aspirin, metformin, statins as well as to nutrient starvation, thus providing a novel instrument for the design of effective therapies against multifactorial metabolism-related CDs.
Collapse
Affiliation(s)
- Wei-Jia Kong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050 Beijing, China
| | - Claudio Vernieri
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Fondazione Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy
| | - Marco Foiani
- Fondazione Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy; University of Milan, Italy.
| | - Jian-Dong Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050 Beijing, China; State Key Laboratory of Bioactive Natural Products and Function, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050 Beijing, China.
| |
Collapse
|
318
|
Short-term starvation reduces IGF-1 levels to sensitize lung tumors to PD-1 immune checkpoint blockade. ACTA ACUST UNITED AC 2020; 1:75-85. [DOI: 10.1038/s43018-019-0007-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/14/2019] [Indexed: 01/31/2023]
|
319
|
Involvement of GABAergic interneuron dysfunction and neuronal network hyperexcitability in Alzheimer's disease: Amelioration by metabolic switching. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:191-205. [DOI: 10.1016/bs.irn.2020.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
320
|
Yan S, Wang C, Zhao H, Pan Y, Wang H, Guo Y, Yao N, Li B, Cui W. Effects of fasting intervention regulating anthropometric and metabolic parameters in subjects with overweight or obesity: a systematic review and meta-analysis. Food Funct 2020; 11:3781-3799. [DOI: 10.1039/d0fo00287a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Previous studies have shown that fasting produces a potential effect in the prevention and treatment of many diseases.
Collapse
Affiliation(s)
- Shoumeng Yan
- Department of Epidemiology and Biostatistics
- School of Public Health
- Jilin University
- Changchun
- P. R. China
| | - Changcong Wang
- Department of Epidemiology and Biostatistics
- School of Public Health
- Jilin University
- Changchun
- P. R. China
| | - Hantong Zhao
- Department of Epidemiology and Biostatistics
- School of Public Health
- Jilin University
- Changchun
- P. R. China
| | - Yingan Pan
- Department of Epidemiology and Biostatistics
- School of Public Health
- Jilin University
- Changchun
- P. R. China
| | - Han Wang
- Department of Epidemiology and Biostatistics
- School of Public Health
- Jilin University
- Changchun
- P. R. China
| | - Yinpei Guo
- Department of Epidemiology and Biostatistics
- School of Public Health
- Jilin University
- Changchun
- P. R. China
| | - Nan Yao
- Department of Epidemiology and Biostatistics
- School of Public Health
- Jilin University
- Changchun
- P. R. China
| | - Bo Li
- Department of Epidemiology and Biostatistics
- School of Public Health
- Jilin University
- Changchun
- P. R. China
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene
- School of Public Health
- Jilin University
- Changchun
- P. R. China
| |
Collapse
|
321
|
Affiliation(s)
- Rafael de Cabo
- From the Translational Gerontology Branch (R.C.) and the Laboratory of Neurosciences (M.P.M.), Intramural Research Program, National Institute on Aging, National Institutes of Health, and the Department of Neuroscience, Johns Hopkins University School of Medicine (M.P.M.) - both in Baltimore
| | - Mark P Mattson
- From the Translational Gerontology Branch (R.C.) and the Laboratory of Neurosciences (M.P.M.), Intramural Research Program, National Institute on Aging, National Institutes of Health, and the Department of Neuroscience, Johns Hopkins University School of Medicine (M.P.M.) - both in Baltimore
| |
Collapse
|
322
|
Francescangeli F, De Angelis ML, Zeuner A. Dietary Factors in the Control of Gut Homeostasis, Intestinal Stem Cells, and Colorectal Cancer. Nutrients 2019; 11:nu11122936. [PMID: 31816977 PMCID: PMC6950549 DOI: 10.3390/nu11122936] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/21/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third commonly diagnosed cancer and the second leading cause of cancer-related deaths worldwide. Global CRC burden is expected to increase by 60% in the next decade, with low-income countries experiencing an escalation of CRC incidence and mortality in parallel to the adoption of western lifestyles. CRC incidence is also sharply increasing in individuals younger than 50 years, often presenting at advanced stages and with aggressive features. Both genetic and environmental factors have been recognized as major contributors for the development of CRC, the latter including diet-related conditions such as chronic inflammation and obesity. In particular, a diet rich in fat and sugars (Western-style diet, WSD) has been shown to induce multiple pathophysiological changes in the intestine linked to an increased risk of CRC. In this scenario, dietary factors have been recently shown to play novel unexpected roles in the regulation of intestinal stem cells (ISCs) and of the gut microbiota, which represent the two main biological systems responsible for intestinal homeostasis. Furthermore, diet is increasingly recognized to play a key role in the neoplastic transformation of ISCs and in the metabolic regulation of colorectal cancer stem cells. This review illustrates novel discoveries on the role of dietary components in regulating intestinal homeostasis and colorectal tumorigenesis. Particular focus is dedicated to new areas of research with potential clinical relevance including the effect of food components on ISCs and cancer stem cells (CSCs), the existence of CRC-specific microbial signatures and the alterations of intestinal homeostasis potentially involved in early-onset CRC. New insights on the role of dietary factors in intestinal regulation will provide new tools not only for the prevention and early diagnosis of CRC but also for improving the effectiveness of current CRC therapies.
Collapse
|
323
|
Chakravorty A, Jetto CT, Manjithaya R. Dysfunctional Mitochondria and Mitophagy as Drivers of Alzheimer's Disease Pathogenesis. Front Aging Neurosci 2019; 11:311. [PMID: 31824296 PMCID: PMC6880761 DOI: 10.3389/fnagi.2019.00311] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022] Open
Abstract
Neurons are highly specialized post-mitotic cells that are inherently dependent on mitochondria owing to their high bioenergetic demand. Mitochondrial dysfunction is therefore associated with various age-related neurodegenerative disorders such as Alzheimer's disease (AD), wherein accumulation of damaged and dysfunctional mitochondria has been reported as an early symptom further contributing to disease progression. In AD, impairment of mitochondrial function causes bioenergetic deficiency, intracellular calcium imbalance and oxidative stress, thereby aggravating the effect of Aβ and tau pathologies, leading to synaptic dysfunction, cognitive impairment and memory loss. Although there are reports suggesting intricate parallelism between mitochondrial dysfunction and AD pathologies such as Aβ aggregation and hyperphosphorylated tau accumulation, the factors that drive the pathogenesis of either are unclear. In addition, emerging evidence suggest that mitochondrial quality control (QC) mechanisms such as mitophagy are impaired in AD. As an important mitochondrial QC mechanism, mitophagy plays a critical role in maintaining neuronal health and function. Studies show that various proteins involved in mitophagy, mitochondrial dynamics, and mitochondrial biogenesis are affected in AD. Compromised mitophagy may also be attributed to impairment in autophagosome-lysosome fusion and defects in lysosomal acidification. Therapeutic interventions aiming to restore mitophagy functions can be used as a strategy for ameliorating AD pathogenesis. Recent evidence implicates the role of microglial activation via mitophagy induction in reducing amyloid plaque load. This review summarizes the current developments in the field of mitophagy and mitochondrial dysfunction in AD.
Collapse
Affiliation(s)
- Anushka Chakravorty
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Cuckoo Teresa Jetto
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| |
Collapse
|
324
|
Sanderson SM, Gao X, Dai Z, Locasale JW. Methionine metabolism in health and cancer: a nexus of diet and precision medicine. Nat Rev Cancer 2019; 19:625-637. [PMID: 31515518 DOI: 10.1038/s41568-019-0187-8] [Citation(s) in RCA: 328] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2019] [Indexed: 01/11/2023]
Abstract
Methionine uptake and metabolism is involved in a host of cellular functions including methylation reactions, redox maintenance, polyamine synthesis and coupling to folate metabolism, thus coordinating nucleotide and redox status. Each of these functions has been shown in many contexts to be relevant for cancer pathogenesis. Intriguingly, the levels of methionine obtained from the diet can have a large effect on cellular methionine metabolism. This establishes a link between nutrition and tumour cell metabolism that may allow for tumour-specific metabolic vulnerabilities that can be influenced by diet. Recently, a number of studies have begun to investigate the molecular and cellular mechanisms that underlie the interaction between nutrition, methionine metabolism and effects on health and cancer.
Collapse
Affiliation(s)
- Sydney M Sanderson
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Xia Gao
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Ziwei Dai
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
325
|
Abstract
The way cancer cells utilize nutrients to support their growth and proliferation is determined by cancer cell-intrinsic and cancer cell-extrinsic factors, including interactions with the environment. These interactions can define therapeutic vulnerabilities and impact the effectiveness of cancer therapy. Diet-mediated changes in whole-body metabolism and systemic nutrient availability can affect the environment that cancer cells are exposed to within tumours, and a better understanding of how diet modulates nutrient availability and utilization by cancer cells is needed. How diet impacts cancer outcomes is also of great interest to patients, yet clear evidence for how diet interacts with therapy and impacts tumour growth is lacking. Here we propose an experimental framework to probe the connections between diet and cancer metabolism. We examine how dietary factors may affect tumour growth by altering the access to and utilization of nutrients by cancer cells. Our growing understanding of how certain cancer types respond to various diets, how diet impacts cancer cell metabolism to mediate these responses and whether dietary interventions may constitute new therapeutic opportunities will begin to provide guidance on how best to use diet and nutrition to manage cancer in patients.
Collapse
Affiliation(s)
- Evan C Lien
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
326
|
Breast Cancer Stem Cells as Drivers of Tumor Chemoresistance, Dormancy and Relapse: New Challenges and Therapeutic Opportunities. Cancers (Basel) 2019; 11:cancers11101569. [PMID: 31619007 PMCID: PMC6826533 DOI: 10.3390/cancers11101569] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most frequent cancer among women worldwide. Therapeutic strategies to prevent or treat metastatic disease are still inadequate although great progress has been made in treating early-stage breast cancer. Cancer stem-like cells (CSCs) that are endowed with high plasticity and self-renewal properties have been shown to play a key role in breast cancer development, progression, and metastasis. A subpopulation of CSCs that combines tumor-initiating capacity and a dormant/quiescent/slow cycling status is present throughout the clinical history of breast cancer patients. Dormant/quiescent/slow cycling CSCs are a key component of tumor heterogeneity and they are responsible for chemoresistance, tumor migration, and metastatic dormancy, defined as the ability of CSCs to survive in target organs and generate metastasis up to two decades after diagnosis. Understanding the strategies that are used by CSCs to resist conventional and targeted therapies, to interact with their niche, to escape immune surveillance, and finally to awaken from dormancy is of key importance to prevent and treat metastatic cancer. This review summarizes the current understanding of mechanisms involved in CSCs chemoresistance, dissemination, and metastasis in breast cancer, with a particular focus on dormant cells. Finally, we discuss how advancements in the detection, molecular understanding, and targeting of dormant CSCs will likely open new therapeutic avenues for breast cancer treatment.
Collapse
|
327
|
Tavakol S, Ashrafizadeh M, Deng S, Azarian M, Abdoli A, Motavaf M, Poormoghadam D, Khanbabaei H, Afshar EG, Mandegary A, Pardakhty A, Yap CT, Mohammadinejad R, Kumar AP. Autophagy Modulators: Mechanistic Aspects and Drug Delivery Systems. Biomolecules 2019; 9:E530. [PMID: 31557936 PMCID: PMC6843293 DOI: 10.3390/biom9100530] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/14/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy modulation is considered to be a promising programmed cell death mechanism to prevent and cure a great number of disorders and diseases. The crucial step in designing an effective therapeutic approach is to understand the correct and accurate causes of diseases and to understand whether autophagy plays a cytoprotective or cytotoxic/cytostatic role in the progression and prevention of disease. This knowledge will help scientists find approaches to manipulate tumor and pathologic cells in order to enhance cellular sensitivity to therapeutics and treat them. Although some conventional therapeutics suffer from poor solubility, bioavailability and controlled release mechanisms, it appears that novel nanoplatforms overcome these obstacles and have led to the design of a theranostic-controlled drug release system with high solubility and active targeting and stimuli-responsive potentials. In this review, we discuss autophagy modulators-related signaling pathways and some of the drug delivery strategies that have been applied to the field of therapeutic application of autophagy modulators. Moreover, we describe how therapeutics will target various steps of the autophagic machinery. Furthermore, nano drug delivery platforms for autophagy targeting and co-delivery of autophagy modulators with chemotherapeutics/siRNA, are also discussed.
Collapse
Affiliation(s)
- Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Milad Ashrafizadeh
- Department of basic science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Maryam Azarian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
- Departament de Bioquímica i Biologia Molecular, Institut de Biotecnologia i Biomedicina (IBB), Universitat Autónoma de Barcelona, Barcelona, Spain.
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | - Mahsa Motavaf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Delaram Poormoghadam
- Department of Medical Nanotechnology, Faculty of Advanced Sciences & Technology, Pharmaceutical Sciences Branch, Islamic Azad University, (IAUPS), Tehran, Iran.
| | - Hashem Khanbabaei
- Medical Physics Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Ali Mandegary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Abbas Pardakhty
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Celestial T Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.
| |
Collapse
|
328
|
De Angelis ML, Francescangeli F, La Torre F, Zeuner A. Stem Cell Plasticity and Dormancy in the Development of Cancer Therapy Resistance. Front Oncol 2019; 9:626. [PMID: 31355143 PMCID: PMC6636659 DOI: 10.3389/fonc.2019.00626] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer treatment with either standard chemotherapy or targeted agents often results in the emergence of drug-refractory cell populations, ultimately leading to therapy failure. The biological features of drug resistant cells are largely overlapping with those of cancer stem cells and include heterogeneity, plasticity, self-renewal ability, and tumor-initiating capacity. Moreover, drug resistance is usually characterized by a suppression of proliferation that can manifest as quiescence, dormancy, senescence, or proliferative slowdown. Alterations in key cellular pathways such as autophagy, unfolded protein response or redox signaling, as well as metabolic adaptations also contribute to the establishment of drug resistance, thus representing attractive therapeutic targets. Moreover, a complex interplay of drug resistant cells with the micro/macroenvironment and with the immune system plays a key role in dictating and maintaining the resistant phenotype. Recent studies have challenged traditional views of cancer drug resistance providing innovative perspectives, establishing new connections between drug resistant cells and their environment and indicating unexpected therapeutic strategies. In this review we discuss recent advancements in understanding the mechanisms underlying drug resistance and we report novel targeting agents able to overcome the drug resistant status, with particular focus on strategies directed against dormant cells. Research on drug resistant cancer cells will take us one step forward toward the development of novel treatment approaches and the improvement of relapse-free survival in solid and hematological cancer patients.
Collapse
Affiliation(s)
- Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Filippo La Torre
- Department of Surgical Sciences Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
329
|
Turbitt WJ, Demark-Wahnefried W, Peterson CM, Norian LA. Targeting Glucose Metabolism to Enhance Immunotherapy: Emerging Evidence on Intermittent Fasting and Calorie Restriction Mimetics. Front Immunol 2019; 10:1402. [PMID: 31293576 PMCID: PMC6603129 DOI: 10.3389/fimmu.2019.01402] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022] Open
Abstract
There is growing interest in harnessing lifestyle and pharmaceutical interventions to boost immune function, reduce tumor growth, and improve cancer treatment efficacy while reducing treatment toxicity. Interventions targeting glucose metabolism are particularly promising, as they have the potential to directly inhibit tumor cell proliferation. However, because anti-tumor immune effector cells also rely on glycolysis to sustain their clonal expansion and function, it remains unclear whether glucose-modulating therapies will support or hinder anti-tumor immunity. In this perspective, we summarize a growing body of literature that evaluates the effects of intermittent fasting, calorie restriction mimetics, and anti-hyperglycemic agents on anti-tumor immunity and immunotherapy outcomes. Based on the limited data currently available, we contend that additional pre-clinical studies and clinical trials are warranted to address the effects of co-administration of anti-hyperglycemic agents or glucose-lowering lifestyle modifications on anti-tumor immunity and cancer treatment outcomes. We stress that there is currently insufficient evidence to provide recommendations regarding these interventions to cancer patients undergoing immunotherapy. However, if found to be safe and effective in clinical trials, interventions targeting glucose metabolism could act as low-cost combinatorial adjuvants for cancer patients receiving immune checkpoint blockade or other immunotherapies.
Collapse
Affiliation(s)
- William J Turbitt
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Wendy Demark-Wahnefried
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, United States.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Courtney M Peterson
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lyse A Norian
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, United States.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
330
|
Catalá-López F, Forés-Martos J, Driver JA, Page MJ, Hutton B, Ridao M, Alonso-Arroyo A, Macías Saint-Gerons D, Gènova-Maleras R, Valderas JM, Vieta E, Valencia A, Tabarés-Seisdedos R. Association of Anorexia Nervosa With Risk of Cancer: A Systematic Review and Meta-analysis. JAMA Netw Open 2019; 2:e195313. [PMID: 31173122 PMCID: PMC6563572 DOI: 10.1001/jamanetworkopen.2019.5313] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPORTANCE Anorexia nervosa is recognized as an important cause of morbidity in young people. However, the risk of cancer in people with anorexia nervosa remains uncertain. OBJECTIVE To evaluate the association of anorexia nervosa with the risk of developing or dying of cancer. DATA SOURCES MEDLINE, Scopus, Embase, and Web of Science from database inception to January 9, 2019. STUDY SELECTION Published observational studies in humans examining the risk of cancer in people with anorexia nervosa compared with the general population or those without anorexia nervosa. Studies needed to report incidence or mortality rate ratios (RRs). DATA EXTRACTION AND SYNTHESIS Screening, data extraction, and methodological quality assessment were performed by at least 2 researchers independently. A random-effects model was used to synthesize individual studies. Heterogeneity (I2) was assessed and 95% prediction intervals (PIs) were calculated. MAIN OUTCOMES AND MEASURES All cancer incidence and cancer mortality associated with anorexia nervosa. Secondary outcomes were site-specific cancer incidence and mortality. RESULTS Seven cohort studies published in 10 articles (42 602 participants with anorexia nervosa) were included. Anorexia nervosa was not associated with risk of developing any cancer (4 studies in women; RR, 0.97; 95% CI, 0.89-1.06; P = .53; I2, 0%; 95% PI, 0.80-1.18; moderate confidence). Anorexia nervosa was associated with decreased breast cancer incidence (5 studies in women; RR, 0.60; 95% CI, 0.50-0.80; P < .001; I2, 0%; 95% PI, 0.44-0.83; high confidence). Conversely, anorexia nervosa was associated with increased risk of developing lung cancer (3 studies in women; RR, 1.50; 95% CI, 1.06-2.12; P = .001; I2, 0%; 95% PI, 0.19-16.46; low confidence) and esophageal cancer (2 studies in women; RR, 6.10; 95% CI, 2.30-16.18; P < .001; I2, 0%; low confidence). CONCLUSIONS AND RELEVANCE Among people with anorexia nervosa, risk of developing cancer did not differ compared with the general population, but a significantly reduced risk of breast cancer was observed. Understanding the mechanisms underlying these associations could have important preventive potential.
Collapse
Affiliation(s)
- Ferrán Catalá-López
- Department of Health Planning and Economics, National School of Public Health, Institute of Health Carlos III, Madrid, Spain
- Department of Medicine, University of Valencia, INCLIVA Health Research Institute, Centro de Investigación en Red de Salud Mental (CIBERSAM), Valencia, Spain
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jaume Forés-Martos
- Department of Medicine, University of Valencia, INCLIVA Health Research Institute, Centro de Investigación en Red de Salud Mental (CIBERSAM), Valencia, Spain
| | - Jane A. Driver
- Geriatric Research Education and Clinical Center, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Division of Aging, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Matthew J. Page
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Brian Hutton
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Manuel Ridao
- Instituto Aragonés de Ciencias de la Salud, Red de Investigación en Servicios de Salud en Enfermedades Crónicas, Zaragoza, Spain
| | - Adolfo Alonso-Arroyo
- Department of History of Science and Documentation, University of Valencia, Valencia, Spain
- Unidad de Información e Investigación Social y Sanitaria, University of Valencia, Spanish National Research Council, Valencia, Spain
| | - Diego Macías Saint-Gerons
- Department of Medicine, University of Valencia, INCLIVA Health Research Institute, Centro de Investigación en Red de Salud Mental (CIBERSAM), Valencia, Spain
| | | | - José M. Valderas
- Health Services and Policy Research Group, Exeter Collaboration for Academic Primary Care, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Eduard Vieta
- Hospital Clínic, Universitat de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Centro de Investigación en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - Alfonso Valencia
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Spain
| | - Rafael Tabarés-Seisdedos
- Department of Medicine, University of Valencia, INCLIVA Health Research Institute, Centro de Investigación en Red de Salud Mental (CIBERSAM), Valencia, Spain
| |
Collapse
|
331
|
CD8 + T cells induce cachexia during chronic viral infection. Nat Immunol 2019; 20:701-710. [PMID: 31110314 PMCID: PMC6531346 DOI: 10.1038/s41590-019-0397-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/08/2019] [Indexed: 12/19/2022]
Abstract
Cachexia represents a leading cause of morbidity and mortality in various cancers, chronic inflammation and infections. Understanding of the mechanisms that drive cachexia has remained limited, especially for infection-associated cachexia (IAC). Here we describe a model of reversible cachexia in mice with chronic viral infection and identify an essential role for CD8+ T cells in IAC. Cytokines linked to cancer-associated cachexia did not contribute to IAC. Instead, virus-specific CD8+ T cells caused morphological and molecular changes in the adipose tissue, which led to depletion of lipid stores. These changes occurred at a time point that preceded the peak of the CD8+ T cell response and required T cell–intrinsic type 1 interferon signaling and antigen-specific priming. Our results link systemic antiviral immune responses to adipose-tissue remodeling and reveal an underappreciated role of CD8+ T cells in IAC.
Collapse
|
332
|
Pérez-Sánchez A, Cuyàs E, Ruiz-Torres V, Agulló-Chazarra L, Verdura S, González-Álvarez I, Bermejo M, Joven J, Micol V, Bosch-Barrera J, Menendez JA. Intestinal Permeability Study of Clinically Relevant Formulations of Silibinin in Caco-2 Cell Monolayers. Int J Mol Sci 2019; 20:E1606. [PMID: 30935093 PMCID: PMC6480586 DOI: 10.3390/ijms20071606] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022] Open
Abstract
An ever-growing number of preclinical studies have investigated the tumoricidal activity of the milk thistle flavonolignan silibinin. The clinical value of silibinin as a bona fide anti-cancer therapy, however, remains uncertain with respect to its bioavailability and blood⁻brain barrier (BBB) permeability. To shed some light on the absorption and bioavailability of silibinin, we utilized the Caco-2 cell monolayer model of human intestinal absorption to evaluate the permeation properties of three different formulations of silibinin: silibinin-meglumine, a water-soluble form of silibinin complexed with the amino-sugar meglumine; silibinin-phosphatidylcholine, the phytolipid delivery system Siliphos; and Eurosil85/Euromed, a milk thistle extract that is the active component of the nutraceutical Legasil with enhanced bioavailability. Our approach predicted differential mechanisms of transport and blood⁻brain barrier permeabilities between the silibinin formulations tested. Our assessment might provide valuable information about an idoneous silibinin formulation capable of reaching target cancer tissues and accounting for the observed clinical effects of silibinin, including a recently reported meaningful central nervous system activity against brain metastases.
Collapse
Affiliation(s)
- Almudena Pérez-Sánchez
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
| | - Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| | - Verónica Ruiz-Torres
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
| | - Luz Agulló-Chazarra
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
| | - Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| | - Isabel González-Álvarez
- Pharmacokinetics and Pharmaceutical Technology Area, Engineering Department, Universidad Miguel Hernández (UMH), San Juan de Alicante, 03202 Alicante, Spain.
| | - Marival Bermejo
- Pharmacokinetics and Pharmaceutical Technology Area, Engineering Department, Universidad Miguel Hernández (UMH), San Juan de Alicante, 03202 Alicante, Spain.
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
| | - Vicente Micol
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), 03202 Elche, Spain.
- CIBER, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III (CB12/03/30038), 07122 Palma de Mallorca, Spain.
| | - Joaquim Bosch-Barrera
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Department of Medical Sciences, Medical School University of Girona, 17003 Girona, Spain.
- Medical Oncology, Catalan Institute of Oncology (ICO), Dr. Josep Trueta University Hospital, 17007 Girona, Spain.
| | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, 17007 Girona, Spain.
- Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain.
| |
Collapse
|
333
|
Shan W, Zheng H, Fu G, Liu C, Li Z, Ye Y, Zhao J, Xu D, Sun L, Wang X, Chen XL, Bi S, Ren L, Fu G. Bioengineered Nanocage from HBc Protein for Combination Cancer Immunotherapy. NANO LETTERS 2019; 19:1719-1727. [PMID: 30724087 DOI: 10.1021/acs.nanolett.8b04722] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Protein nanocages are promising multifunctional platforms for nanomedicine owing to the ability to decorate their surfaces with multiple functionalities through genetic and/or chemical modification to achieve desired properties for therapeutic and diagnostic purposes. Here, we describe a model antigen (OVA peptide) that was conjugated to the surface of a naturally occurring hepatitis B core protein nanocage (HBc NC) by genetic modification. The engineered OVA-HBc nanocages (OVA-HBc NCs), displaying high density repetitive array of epitopes in a limited space by self-assembling into symmetrical structure, not only can induce bone marrow derived dendritic cells (BMDC) maturation effectively but also can be enriched in the draining lymph nodes. Naïve C57BL/6 mice immunized with OVA-HBc NCs are able to generate significant and specific cytotoxic T lymphocyte (CTL) responses. Moreover, OVA-HBc NCs as a robust nanovaccine can trigger preventive antitumor immunity and significantly delay tumor growth. When combined with a low-dose chemotherapy drug (paclitaxel), OVA-HBc NCs could specifically inhibit progression of an established tumor. Our findings support HBc-based nanocages with modularity and scalability as an attractive nanoplatform for combination cancer immunotherapy.
Collapse
Affiliation(s)
- Wenjun Shan
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials , Xiamen University , Xiamen , Fujian 361005 , P. R. China
| | - Haiping Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences , Xiamen University , Xiamen , Fujian 361102 , China
| | - Guofeng Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences , Xiamen University , Xiamen , Fujian 361102 , China
| | - Chenfeng Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences , Xiamen University , Xiamen , Fujian 361102 , China
| | - Zizhen Li
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health , Xiamen University , Xiamen , Fujian 361102 , P. R. China
| | - Yuhan Ye
- Zhongshan Hospital , Xiamen University , Xiamen , Fujian 361005 , P. R. China
| | - Jie Zhao
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials , Xiamen University , Xiamen , Fujian 361005 , P. R. China
| | - Dan Xu
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials , Xiamen University , Xiamen , Fujian 361005 , P. R. China
| | - Liping Sun
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials , Xiamen University , Xiamen , Fujian 361005 , P. R. China
| | - Xin Wang
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health , Xiamen University , Xiamen , Fujian 361102 , P. R. China
| | - Xiao Lei Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences , Xiamen University , Xiamen , Fujian 361102 , China
| | - Shengli Bi
- Chinese Center for Disease Control & Prevention Institute for Viral Disease Control & Prevention , Beijing 102206 , P. R. China
| | - Lei Ren
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials , Xiamen University , Xiamen , Fujian 361005 , P. R. China
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences , Xiamen University , Xiamen , Fujian 361102 , China
- Cancer Research Center of Xiamen University , Xiamen , Fujian 361102 , China
| |
Collapse
|
334
|
Affiliation(s)
- Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
| | | | - Valter D Longo
- IFOM, FIRC Institute of Molecular Oncology, Milano, Italy.
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
335
|
Affiliation(s)
- Riccardo Caccialanza
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| | - Emanuele Cereda
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Paolo Pedrazzoli
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo and Department of Internal Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
336
|
Bland JS. Fasting Physiology and Therapeutic Diets: A Look Back to the Future. Integr Med (Encinitas) 2019; 18:16-21. [PMID: 31341428 PMCID: PMC6601432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The evidence presented at this event demonstrated the multiple clinical benefits of fasting physiology and points toward a future in which the clinical applications of dietary approaches will be well understood and successfully utilized. The conference reflected the scope and breadth of current research efforts in this important clinical area. Clearly, the application of the important new concepts related to fasting physiology that are emerging will require the advocacy and participation of professionals who are well trained in the fields of clinical nutrition and personalized lifestyle medicine.
Collapse
|
337
|
Poljsak B, Kovac V, Dahmane R, Levec T, Starc A. Cancer Etiology: A Metabolic Disease Originating from Life's Major Evolutionary Transition? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7831952. [PMID: 31687086 PMCID: PMC6800902 DOI: 10.1155/2019/7831952] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 07/21/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022]
Abstract
A clear understanding of the origins of cancer is the basis of successful strategies for effective cancer prevention and management. The origin of cancer at the molecular and cellular levels is not well understood. Is the primary cause of the origin of cancer the genomic instability or impaired energy metabolism? An attempt was made to present cancer etiology originating from life's major evolutionary transition. The first evolutionary transition went from simple to complex cells when eukaryotic cells with glycolytic energy production merged with the oxidative mitochondrion (The Endosymbiosis Theory first proposed by Lynn Margulis in the 1960s). The second transition went from single-celled to multicellular organisms once the cells obtained mitochondria, which enabled them to obtain a higher amount of energy. Evidence will be presented that these two transitions, as well as the decline of NAD+ and ATP levels, are the root of cancer diseases. Restoring redox homeostasis and reactivation of mitochondrial oxidative metabolism are important factors in cancer prevention.
Collapse
Affiliation(s)
- B. Poljsak
- 1Faculty of Health Sciences, University of Ljubljana, Laboratory of Oxidative Stress Research, Ljubljana, Slovenia
| | - V. Kovac
- 1Faculty of Health Sciences, University of Ljubljana, Laboratory of Oxidative Stress Research, Ljubljana, Slovenia
| | - R. Dahmane
- 2Faculty of Health Sciences, University of Ljubljana, Chair of Biomedicine in Health Care, Ljubljana, Slovenia
| | - T. Levec
- 3Faculty of Health Sciences, University of Ljubljana, Chair of Public Health, Ljubljana, Slovenia
| | - A. Starc
- 3Faculty of Health Sciences, University of Ljubljana, Chair of Public Health, Ljubljana, Slovenia
| |
Collapse
|