1
|
Wang D, Duan JJ, Guo YF, Chen JJ, Chen TQ, Wang J, Yu SC. Targeting the glutamine-arginine-proline metabolism axis in cancer. J Enzyme Inhib Med Chem 2024; 39:2367129. [PMID: 39051546 PMCID: PMC11275534 DOI: 10.1080/14756366.2024.2367129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 04/27/2024] [Accepted: 06/06/2024] [Indexed: 07/27/2024] Open
Abstract
Metabolic abnormalities are an important feature of tumours. The glutamine-arginine-proline axis is an important node of cancer metabolism and plays a major role in amino acid metabolism. This axis also acts as a scaffold for the synthesis of other nonessential amino acids and essential metabolites. In this paper, we briefly review (1) the glutamine addiction exhibited by tumour cells with accelerated glutamine transport and metabolism; (2) the methods regulating extracellular glutamine entry, intracellular glutamine synthesis and the fate of intracellular glutamine; (3) the glutamine, proline and arginine metabolic pathways and their interaction; and (4) the research progress in tumour therapy targeting the glutamine-arginine-proline metabolic system, with a focus on summarising the therapeutic research progress of strategies targeting of one of the key enzymes of this metabolic system, P5CS (ALDH18A1). This review provides a new basis for treatments targeting the metabolic characteristics of tumours.
Collapse
Affiliation(s)
- Di Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
| | - Jiang-jie Duan
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
- Jin-feng Laboratory, Chongqing, China
| | - Yu-feng Guo
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jun-jie Chen
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
| | - Tian-qing Chen
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
- Jin-feng Laboratory, Chongqing, China
| | - Shi-cang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, China
- Jin-feng Laboratory, Chongqing, China
| |
Collapse
|
2
|
Wang D, Zhu L, Liu H, Feng X, Zhang C, Li T, Liu B, Liu L, Sun J, Chang H, Chen S, Guo S, Yang W. Huangqin tang alleviates colitis-associated colorectal cancer via amino acids homeostasisand PI3K/AKT/mtor pathway modulation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118597. [PMID: 39034016 DOI: 10.1016/j.jep.2024.118597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/29/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqin Tang (HQT), a traditional Chinese medicine formula, is commonly used in clinical practice for the treatment of inflammatory bowel diseases. It has been reported that HQT exerts antitumor effects on colitis-associated colorectal cancer (CAC). However, the mechanism by which HQT interferes with the inflammation-to-cancer transformation remains unclear. AIMS OF THE STUDY The purpose of this study was to dynamically evaluate the efficacy of HQT in alleviating or delaying CAC and to reveal the underlying mechanism. METHODS We established a mouse model of CAC using azoxymethane combined with 1.5% dextran sodium sulphate. The efficacy of HQT was evaluated based on pathological sections and serum biochemical indices. Subsequently, amino acids (AAs) metabolism analyses were performed using ultra-performance liquid chromatography-tandem mass spectrometry, and the phosphatidylinositol 3 kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR) pathway was detected by western blotting. RESULTS The data demonstrated that HQT could alleviate the development of CAC in the animal model. HQT effectively reduced the inflammatory response, particularly interleukin-6 (IL-6), in the inflammation induction stage, as well as in the stages of proliferation initiation and tumorigenesis. During the proliferation initiation and tumorigenesis stages, immunohistochemistry staining showed that the expression of the proliferation marker Ki67 was reduced, while apoptosis was increased in the HQT group. Accordingly, HQT substantially decreased the levels of specific AAs in the colon with CAC, including glutamic acid, glutamine, arginine, and isoleucine. Furthermore, HQT significantly inhibited the activated PI3K/AKT/mTOR pathway, which may contribute to suppression of cell proliferation and enhancement of apoptosis. CONCLUSION HQT is effective in alleviating and delaying the colon "inflammation-to-cancer". The mechanism of action may involve HQT maintained AAs metabolism homeostasis and regulated PI3K/AKT/mTOR pathway, so as to maintain the balance between proliferation and apoptosis, and then interfere in the occurrence and development of CAC.
Collapse
Affiliation(s)
- Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Lin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Haifan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Xue Feng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Caijuan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Tao Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Bin Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Jingwei Sun
- Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Hao Chang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Siyuan Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Shanshan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
3
|
Tebeje BM, Thiex NW, Swanson JA. Growing Macrophages Regulate High Rates of Solute Flux by Pinocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619691. [PMID: 39484410 PMCID: PMC11526976 DOI: 10.1101/2024.10.22.619691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
In metazoan cells, growth factors stimulate solute ingestion by pinocytosis. To examine the role of pinocytosis in cell growth, this study measured cell proliferation and the attendant rates of solute flux by pinocytosis in murine macrophages in response to the growth factor colony-stimulating factor-1 (CSF1). During CSF1-dependent growth in rich medium, macrophages internalized 72 percent of their cell volume in extracellular fluid every hour. Removal of the essential amino acid leucine from growth medium limited rates of protein synthesis and growth, but increased rates of solute accumulation by macropinocytosis. The amount of protein synthesized during leucine-dependent growth exceeded the capacity of pinocytosis to internalize enough soluble leucine to support growth and proliferation. Fluid-phase solute recycling from lysosomes secreted small molecules from the cells at high rates. Inhibitors of pinocytosis and the mechanistic target-of-rapamycin (mTOR) reduced cell growth and solute recycling, indicating roles for pinocytosis in growth and for nutrient sensing in the regulation of solute flux by pinocytosis. Summary Murine macrophages growing in response to colony-stimulating factor-1 (CSF1) require pinocytosis. High rates of solute influx and accumulation by pinocytosis are regulated by CSF1 and leucine. Low molecular weight products of protein hydrolysis in lysosomes recycle efficiently from the cells.
Collapse
|
4
|
Kolaczkowski OM, Goodson BA, Vazquez VM, Jia J, Bhat AQ, Kim TH, Pu J. Synergistic Role of Amino Acids in Enhancing mTOR Activation Through Lysosome Positioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.12.618047. [PMID: 39416115 PMCID: PMC11482915 DOI: 10.1101/2024.10.12.618047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Lysosome positioning, or lysosome cellular distribution, is critical for lysosomal functions in response to both extracellular and intracellular cues. Amino acids, as essential nutrients, have been shown to promote lysosome movement toward the cell periphery. Peripheral lysosomes are involved in processes such as lysosomal exocytosis, cell migration, and metabolic signaling-functions that are particularly important for cancer cell motility and growth. However, the specific types of amino acids that regulate lysosome positioning, their underlying mechanisms, and their connection to amino acid-regulated metabolic signaling remain poorly understood. In this study, we developed a high-content imaging system for unbiased, quantitative analysis of lysosome positioning. We examined the 15 amino acids present in cell culture media and found that 10 promoted lysosome redistribution toward the cell periphery to varying extents, with aromatic amino acids showing the strongest effect. This redistribution was mediated by promoting outward transport through SLC38A9-BORC-kinesin 1/3 axis and simultaneously reducing inward transport via inhibiting the recruitment of Rab7 and JIP4 onto lysosomes. When examining the effects of amino acids on mTOR activation-a central regulator of cell metabolism-we found that the amino acids most strongly promoting lysosome dispersal, such as phenylalanine, did not activate mTOR on their own. However, combining phenylalanine with arginine, which activates mTOR without affecting lysosome positioning, synergistically enhanced mTOR activity. This synergy was lost when lysosomes failed to localize to the cell periphery, as observed in kinesin 1/3 knockout (KO) cells. Furthermore, breast cancer cells exhibited heightened sensitivity to phenylalanine-induced lysosome dispersal compared to noncancerous breast cells. Inhibition of LAT1, the amino acid transporter responsible for phenylalanine uptake, reduced peripheral lysosomes and impaired cancer cell migration and proliferation, highlighting the importance of lysosome positioning in these coordinated cellular activities. In summary, amino acid-regulated lysosome positioning and mTOR signaling depend on distinct sets of amino acids. Combining lysosome-dispersing amino acids with mTOR-activating amino acids synergistically enhances mTOR activation, which may be particularly relevant in cancer cells.
Collapse
Affiliation(s)
- Oralia M. Kolaczkowski
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Baley A. Goodson
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Valeria Montenegro Vazquez
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Jingyue Jia
- Autophagy, Inflammation, and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Aadil Qadir Bhat
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Tae-Hyung Kim
- Autophagy, Inflammation, and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - Jing Pu
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| |
Collapse
|
5
|
Tan J, Wang Z, Huang Z, Huang A, Zhang H, Huang L, Song N, Xin G, Jiang K, Sun X. Glutamine maintains the stability of alveolar structure and function after lung transplantation by inhibiting autophagy. Biochem Biophys Res Commun 2024; 727:150308. [PMID: 38968769 DOI: 10.1016/j.bbrc.2024.150308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/12/2024] [Accepted: 06/21/2024] [Indexed: 07/07/2024]
Abstract
Excessive autophagy may lead to degradation and damage of alveolar epithelial cells after lung transplantation, eventually leading to alveolar epithelial cell loss, affecting the structural integrity and function of alveoli. Glutamine (Gln), a nutritional supplement, regulates autophagy through multiple signaling pathways. In this study, we explored the protective role of Gln on alveolar epithelial cells by inhibiting autophagy. In vivo, a rat orthotopic lung transplant model was carried out to evaluate the therapeutic effect of glutamine. Ischemia/reperfusion (I/R) induced alveolar collapse, edema, epithelial cell apoptosis, and inflammation, which led to a reduction of alveolar physiological function, such as an increase in peak airway pressure, and a decrease in lung compliance and oxygenation index. In comparison, Gln preserved alveolar structure and function by reducing alveolar apoptosis, inflammation, and edema. In vitro, a hypoxia/reoxygenation (H/R) cell model was performed to simulate IR injury on mouse lung epithelial (MLE) cells and human lung bronchus epithelial (Beas-2B) cells. H/R impaired the proliferation of epithelial cells and triggered cell apoptosis. In contrast, Gln normalized cell proliferation and suppressed I/R-induced cell apoptosis. The activation of mTOR and the downregulation of autophagy-related proteins (LC3, Atg5, Beclin1) were observed in Gln-treated lung tissues and alveolar epithelial cells. Both in vivo and in vitro, rapamycin, a classical mTOR inhibitor, reversed the beneficial effects of Gln on alveolar structure and function. Taken together, Glnpreserved alveolar structure and function after lung transplantation by inhibiting autophagy.
Collapse
Affiliation(s)
- Jun Tan
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhaokai Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhihong Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ai Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huan Zhang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lei Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Naicheng Song
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gaojie Xin
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xiangfu Sun
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
6
|
Manning BD, Dibble CC. Growth Signaling Networks Orchestrate Cancer Metabolic Networks. Cold Spring Harb Perspect Med 2024; 14:a041543. [PMID: 38438221 PMCID: PMC11444256 DOI: 10.1101/cshperspect.a041543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Normal cells grow and divide only when instructed to by signaling pathways stimulated by exogenous growth factors. A nearly ubiquitous feature of cancer cells is their capacity to grow independent of such signals, in an uncontrolled, cell-intrinsic manner. This property arises due to the frequent oncogenic activation of core growth factor signaling pathway components, including receptor tyrosine kinases, PI3K-AKT, RAS-RAF, mTORC1, and MYC, leading to the aberrant propagation of pro-growth signals independent of exogenous growth factors. The growth of both normal and cancer cells requires the acquisition of nutrients and their anabolic conversion to the primary macromolecules underlying biomass production (protein, nucleic acids, and lipids). The core growth factor signaling pathways exert tight regulation of these metabolic processes and the oncogenic activation of these pathways drive the key metabolic properties of cancer cells and tumors. Here, we review the molecular mechanisms through which these growth signaling pathways control and coordinate cancer metabolism.
Collapse
Affiliation(s)
- Brendan D Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Christian C Dibble
- Department of Pathology, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
7
|
Pavlova NN, Thompson CB. Oncogenic Control of Metabolism. Cold Spring Harb Perspect Med 2024; 14:a041531. [PMID: 38565265 PMCID: PMC11444253 DOI: 10.1101/cshperspect.a041531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
A cell committed to proliferation must reshape its metabolism to enable robust yet balanced production of building blocks for the assembly of proteins, lipids, nucleic acids, and other macromolecules, from which two functional daughter cells can be produced. The metabolic remodeling associated with proliferation is orchestrated by a number of pro-proliferative signaling nodes, which include phosphatidylinositol-3 kinase (PI3K), the RAS family of small GTPases, and transcription factor c-myc In metazoan cells, these signals are activated in a paracrine manner via growth factor-mediated activation of receptor (or receptor-associated) tyrosine kinases. Such stimuli are limited in duration and therefore allow the metabolism of target cells to return to the resting state once the proliferation demands have been satisfied. Cancer cells acquire activating genetic alterations within common pro-proliferative signaling nodes. These alterations lock cellular nutrient uptake and utilization into a perpetual progrowth state, leading to the aberrant accumulation and spread of cancer cells.
Collapse
Affiliation(s)
- Natalya N Pavlova
- Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
8
|
Tang X, Liu H, Chang L, Wang X, Liu Q, Tang Z, Xia Q, Zhao P. A strategy for improving silk yield and organ size in silk-producing insects. FEBS J 2024; 291:4286-4300. [PMID: 38923388 DOI: 10.1111/febs.17193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/24/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024]
Abstract
Insect silks possess excellent biodegradability, biocompatibility and mechanical properties, and have numerous applications in biomedicine and tissue engineering. However, the application of silk fiber is hindered by its limited supply, especially from non-domesticated insects. In the present study, the silk yield and organ size of Bombyx mori were significantly improved through genetic manipulation of the target of rapamycin complex 1 (TORC1) pathway components. Silk protein synthesis and silk gland size were decreased following rapamycin treatment, inhibiting the TORC1 signaling pathway both in vivo and ex vivo. The overexpression of posterior silk gland-specific Rheb and BmSLC7A5 improved silk protein synthesis and silk gland size by activating the TORC1 signaling pathway. Silk yield in BmSLC7A5-overexpression silkworms was significantly increased by approximately 25%. We have demonstrated that the TORC1 signaling pathway is involved in the transcription and translation of silk genes and transcriptional activators via phosphorylation of p70 S6 kinase 1 and 4E-binding protein 1. Our findings present a strategy for increasing silk yield and organ size in silk-producing insects.
Collapse
Affiliation(s)
- Xin Tang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of the Innovative Chinese Materia Medica & Health Intervention, Chongqing Academy of Chinese Materia Medica, China
| | - Huawei Liu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Li Chang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Institute for Brain Science and Disease, Chongqing Medical University, China
| | - Xin Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Qingsong Liu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Zhangchen Tang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Ping Zhao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| |
Collapse
|
9
|
Soll D, Chu CF, Sun S, Lutz V, Arunkumar M, Gachechiladze M, Schäuble S, Alissa-Alkhalaf M, Nguyen T, Khalil MA, Garcia-Ribelles I, Mueller M, Buder K, Michalke B, Panagiotou G, Ziegler-Martin K, Benz P, Schatzlmaier P, Hiller K, Stockinger H, Luu M, Schober K, Moosmann C, Schamel WW, Huber M, Zielinski CE. Sodium chloride in the tumor microenvironment enhances T cell metabolic fitness and cytotoxicity. Nat Immunol 2024; 25:1830-1844. [PMID: 39198632 PMCID: PMC11436365 DOI: 10.1038/s41590-024-01918-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 07/04/2024] [Indexed: 09/01/2024]
Abstract
The efficacy of antitumor immunity is associated with the metabolic state of cytotoxic T cells, which is sensitive to the tumor microenvironment. Whether ionic signals affect adaptive antitumor immune responses is unclear. In the present study, we show that there is an enrichment of sodium in solid tumors from patients with breast cancer. Sodium chloride (NaCl) enhances the activation state and effector functions of human CD8+ T cells, which is associated with enhanced metabolic fitness. These NaCl-induced effects translate into increased tumor cell killing in vitro and in vivo. Mechanistically, NaCl-induced changes in CD8+ T cells are linked to sodium-induced upregulation of Na+/K+-ATPase activity, followed by membrane hyperpolarization, which magnifies the electromotive force for T cell receptor (TCR)-induced calcium influx and downstream TCR signaling. We therefore propose that NaCl is a positive regulator of acute antitumor immunity that might be modulated for ex vivo conditioning of therapeutic T cells, such as CAR T cells.
Collapse
Affiliation(s)
| | - Chang-Feng Chu
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Shan Sun
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Veronika Lutz
- Institute of Systems Immunology, Philipps-University Marburg, Marburg, Germany
| | - Mahima Arunkumar
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Mariam Gachechiladze
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Sascha Schäuble
- Department of Microbiome Dynamics, Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Maha Alissa-Alkhalaf
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Trang Nguyen
- Institute of Biology III, Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Michelle-Amirah Khalil
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Ignacio Garcia-Ribelles
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Michael Mueller
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | | | - Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich-German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
- Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Kai Ziegler-Martin
- Chair for Cellular Immunotherapy, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Pascal Benz
- Chair for Cellular Immunotherapy, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Philipp Schatzlmaier
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Hannes Stockinger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Maik Luu
- Chair for Cellular Immunotherapy, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Kilian Schober
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Carolin Moosmann
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang W Schamel
- Institute of Biology III, Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Magdalena Huber
- Institute of Systems Immunology, Philipps-University Marburg, Marburg, Germany
| | - Christina E Zielinski
- Technical University of Munich, Munich, Germany.
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany.
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
10
|
Tang X, Liu H, Wang X, Chang L, Liu Q, Xia Q, Zhao P. BmSLC7A5 is essential for silk protein synthesis and larval development in Bombyx mori. INSECT SCIENCE 2024; 31:1425-1439. [PMID: 38284747 DOI: 10.1111/1744-7917.13314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 01/30/2024]
Abstract
Insects produce silk to form cocoons, nests, and webs, which are important for their survival and reproduction. However, little is known about the molecular mechanism of silk protein synthesis at the translation level. The solute carrier family 7 (SLC7) genes are involved in activating the target of rapamycin complex 1 (TORC1) signaling pathway and protein translation process, but the physiological roles of SLC7 genes in silk-producing insects have not been reported. Here, we found that amino acid signaling regulates silk protein synthesis and larval development via the L-type amino acid transporter 1 (LAT1; also known as SLC7A5) in Bombyx mori. A total of 12 SLC7 homologs were identified in the silkworm genome, among which BmSLC7A5 was found to be a silk gland-enriched gene and may be involved in leucine transport. Bioinformatics analysis indicated that SLC7A5 displays high homology and a close phylogenetic relationship in silk-producing insects. Subsequently, we found that leucine treatment significantly increased silk protein synthesis by improving the transcription and protein levels of silk genes. Furthermore, systemic and silk gland-specific knockout of BmSLC7A5 led to decreased silk protein synthesis by inhibiting TORC1 signaling, and somatic mutation also resulted in arrested development from the 5th instar to the early pupal stage. Altogether, our study reveals that BmSLC7A5 is involved in regulating silk protein synthesis and larval development by affecting the TORC1 signaling pathway, which provides a new strategy and target for improving silk yield.
Collapse
Affiliation(s)
- Xin Tang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Chinese Medicine & Health Science, Chongqing Academy of Chinese Materia Medica, Chongqing College of Traditional Chinese Medicine, Chongqing, China
| | - Huawei Liu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Xin Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Li Chang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Qingsong Liu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| | - Ping Zhao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing, China
- Key Laboratory for Germplasm Creation in Upper Reaches of the Yangtze River, Ministry of Agriculture and Rural Affairs, Chongqing, China
| |
Collapse
|
11
|
Huang G, Cai Y, Ren M, Zhang X, Fu Y, Cheng R, Wang Y, Miao M, Zhu L, Yan T. Salidroside sensitizes Triple-negative breast cancer to ferroptosis by SCD1-mediated lipogenesis and NCOA4-mediated ferritinophagy. J Adv Res 2024:S2090-1232(24)00429-6. [PMID: 39353532 DOI: 10.1016/j.jare.2024.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/25/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is the primary cause of breast cancer-induced death in women. Literature has confirmed the benefits of Salidroside (Sal) in treating TNBC. However, the study about potential therapeutic targets and mechanisms of Sal-anchored TNBC remains limited. OBJECTIVE This study was designed to explore the main targets and potential mechanisms of Sal against TNBC. METHODS Network pharmacology, bioinformatics, and machine learning algorithm strategies were integrated to examine the role, potential targets, and mechanisms of the Sal act in TNBC. MDA-MB-231 cells and tumor-bearing nude mice were chosen for in vitro and in vivo experimentation. Cell viability and cytotoxicity were determined using CCK-8, LDH test, and Calcein-AM/PI staining. Antioxidant defense, lipid peroxidation, and iron metabolism were explored using glutathione, glutathione peroxidase, malondialdehyde (MDA), C11-BODIPY 581/591 probe, and FerroOrange dye. Glutathione peroxidase 4 (GPX4) or stearoyl-CoA desaturase 1 (SCD1) overexpression or nuclear receptor co-activator 4 (NCOA4) deficiency was performed to demonstrate the mechanism of Sal on TNBC. RESULTS The prediction results confirmed that 22 ferroptosis-related genes were identified in Sal and TNBC, revealing that the potential mechanism of the Sal act on TNBC was linked with ferroptosis. Besides, these genes were mainly involved in the mTOR, PI3K/AKT, and autophagy signaling pathway by functional enrichment analysis. The in vitro validation results confirmed that Sal inhibited TNBC cell proliferation by modulating ferroptosis via elevation of intracellular Fe2+ and lipid peroxidation. Mechanistically, Sal sensitized TNBC cells to ferroptosis by inhibiting the PI3K/AKT/mTOR axis, thereby suppressing SCD1-mediated lipogenesis of monounsaturated fatty acids to induce lipid peroxidation, additionally facilitating NCOA4-mediated ferritinophagy to increase intracellular Fe2+ content. The GPX4 or SCD1 overexpression or NCOA4 deficiency results further supported our mechanistic studies. In vivo experimentation confirmed that Sal is vital for slowing down tumor growth by inducing ferroptosis. CONCLUSIONS Overall, this study elucidates TNBC pathogenesis closely linked to ferroptosis and identifies potential biomarkers in TNBC. Meanwhile, the study elucidates that Sal sensitizes TNBC to ferroptosis by SCD1-mediated lipogenesis and NCOA4-mediated ferritinophagy, regulated by PI3K/AKT/mTOR signaling pathways. Our findings provide a theoretical basis for applying Sal to treat TNBC.
Collapse
Affiliation(s)
- Guiqin Huang
- School of Basic Medical Science and Clinical Pharmacy, China Pharmaceutic University, Nanjing, Jiangsu 210009, China
| | - Yawen Cai
- School of Basic Medical Science and Clinical Pharmacy, China Pharmaceutic University, Nanjing, Jiangsu 210009, China
| | - Menghui Ren
- School of Basic Medical Science and Clinical Pharmacy, China Pharmaceutic University, Nanjing, Jiangsu 210009, China
| | - Xiaoyu Zhang
- School of Basic Medical Science and Clinical Pharmacy, China Pharmaceutic University, Nanjing, Jiangsu 210009, China
| | - Yu Fu
- School of Basic Medical Science and Clinical Pharmacy, China Pharmaceutic University, Nanjing, Jiangsu 210009, China
| | - Run Cheng
- School of Basic Medical Science and Clinical Pharmacy, China Pharmaceutic University, Nanjing, Jiangsu 210009, China
| | - Yingdi Wang
- School of Basic Medical Science and Clinical Pharmacy, China Pharmaceutic University, Nanjing, Jiangsu 210009, China
| | - Mingxing Miao
- National Experimental Teaching Demonstration Center of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| | - Lingpeng Zhu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China.
| | - Tianhua Yan
- School of Basic Medical Science and Clinical Pharmacy, China Pharmaceutic University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
12
|
Lobel GP, Han N, Arocho WAM, Silber M, Shoush J, Noji MC, Jerrick To TK, Zhai L, Lesner NP, Simon MC, Haldar M. Glutamine availability regulates cDC subsets in tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613574. [PMID: 39345449 PMCID: PMC11429688 DOI: 10.1101/2024.09.17.613574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Proliferating tumor cells take up glutamine for anabolic processes engendering glutamine deficiency in the tumor microenvironment. How this might impact immune cells is not well understood. Using multiple mouse models of soft tissue sarcomas, glutamine antagonists, as well as genetic and pharmacological inhibition of glutamine utilization, we found that the number and frequency of conventional dendritic cells (cDC) is dependent on microenvironmental glutamine levels. cDCs comprise two distinct subsets - cDC1 and cDC2, with the former subset playing a critical role in antigen cross-presentation and tumor immunity. While both subsets show dependence on Glutamine, cDC1s are particularly sensitive. Notably, glutamine antagonism did not reduce the frequency of DC precursors but decreased proliferation and survival of cDC1s. Further studies suggest a role of the nutrient sensing mTOR signaling pathway in this process. Taken together, these findings uncover glutamine dependence of cDC1s that is coopted by tumors to escape immune responses. One Sentence Summary Type 1 conventional dendritic cells require glutamine to maintain their number in non-lymphoid tissue. Significance Immune evasion is a key hallmark of cancer; however, the underlying pathways are diverse, tumor-specific and not fully elucidated. Many tumor cells avidly import glutamine to support their anabolic needs, creating a glutamine-deficient tumor microenvironment (TME). Herein, using mouse models of soft tissue sarcomas, we show that glutamine depletion in TME leads to reduced type 1 conventional dendritic cells - a cell type that is critical for adaptive immune responses. This work is a paradigm for how tumor cell metabolism can regulate anti-tumor immune responses and will be foundational to future efforts targeting glutamine metabolism for cancer immunotherapy.
Collapse
|
13
|
Carrà G, Petiti J, Tolino F, Vacca R, Orso F. MicroRNAs in metabolism for precision treatment of lung cancer. Cell Mol Biol Lett 2024; 29:121. [PMID: 39256662 PMCID: PMC11384722 DOI: 10.1186/s11658-024-00632-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
The dysregulation of miRNAs in lung cancer has been extensively documented, with specific miRNAs acting as both tumor suppressors and oncogenes, depending on their target genes. Recent research has unveiled the regulatory roles of miRNAs in key metabolic pathways, such as glycolysis, the tricarboxylic acid cycle, fatty acid metabolism, and autophagy, which collectively contribute to the aberrant energy metabolism characteristic of cancer cells. Furthermore, miRNAs are increasingly recognized as critical modulators of the tumor microenvironment, impacting immune response and angiogenesis. This review embarks on a comprehensive journey into the world of miRNAs, unraveling their multifaceted roles, and more notably, their emerging significance in the context of cancer, with a particular focus on lung cancer. As we navigate this extensive terrain, we will explore the fascinating realm of miRNA-mediated metabolic rewiring, a phenomenon that plays a pivotal role in the progression of lung cancer and holds promise in the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy.
- San Luigi Gonzaga Hospital, Orbassano, Italy.
| | - Jessica Petiti
- Division of Advanced Materials Metrology and Life Sciences, Istituto Nazionale di Ricerca Metrologica (INRiM), 10135, Turin, Italy
| | - Federico Tolino
- Department of Translational Medicine (DIMET), University of Eastern Piedmont, Novara, Italy
| | - Rita Vacca
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Turin, Italy
| | - Francesca Orso
- Department of Translational Medicine (DIMET), University of Eastern Piedmont, Novara, Italy.
| |
Collapse
|
14
|
Wang Z, Xiong S, Wu Z, Wang X, Gong Y, Zhu WG, Xu X. VCP/p97 UFMylation stabilizes BECN1 and facilitates the initiation of autophagy. Autophagy 2024; 20:2041-2054. [PMID: 38762759 PMCID: PMC11346537 DOI: 10.1080/15548627.2024.2356488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 05/05/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024] Open
Abstract
Macroautophagy/autophagy is essential for the degradation and recycling of cytoplasmic materials. The initiation of this process is determined by phosphatidylinositol-3-kinase (PtdIns3K) complex, which is regulated by factor BECN1 (beclin 1). UFMylation is a novel ubiquitin-like modification that has been demonstrated to modulate several cellular activities. However, the role of UFMylation in regulating autophagy has not been fully elucidated. Here, we found that VCP/p97 is UFMylated on K109 by the E3 UFL1 (UFM1 specific ligase 1) and this modification promotes BECN1 stabilization and assembly of the PtdIns3K complex, suggesting a role for VCP/p97 UFMylation in autophagy initiation. Mechanistically, VCP/p97 UFMylation stabilizes BECN1 through ATXN3 (ataxin 3)-mediated deubiquitination. As a key component of the PtdIns3K complex, stabilized BECN1 facilitates assembly of this complex. Re-expression of VCP/p97, but not the UFMylation-defective mutant, rescued the VCP/p97 depletion-induced increase in MAP1LC3B/LC3B protein expression. We also showed that several pathogenic VCP/p97 mutations identified in a variety of neurological disorders and cancers were associated with reduced UFMylation, thus implicating VCP/p97 UFMylation as a potential therapeutic target for these diseases. Abbreviation: ATG14:autophagy related 14; Baf A1:bafilomycin A1;CMT2Y: Charcot-Marie-Toothdisease, axonal, 2Y; CYB5R3: cytochromeb5 reductase 3; DDRGK1: DDRGK domain containing 1; DMEM:Dulbecco'smodified Eagle's medium;ER:endoplasmic reticulum; FBS:fetalbovine serum;FTDALS6:frontotemporaldementia and/or amyotrophic lateral sclerosis 6; IBMPFD1:inclusion bodymyopathy with early-onset Paget disease with or withoutfrontotemporal dementia 1; LC-MS/MS:liquid chromatography tandem mass spectrometry; MAP1LC3B/LC3B:microtubule associated protein 1 light chain 3 beta; MS: massspectrometry; NPLOC4: NPL4 homolog, ubiquitin recognition factor;PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3;PIK3R4: phosphoinositide-3-kinase regulatory subunit 4; PtdIns3K:phosphatidylinositol 3-kinase; RPL26: ribosomal protein L26; RPN1:ribophorin I; SQSTM1/p62: sequestosome 1; UBA5: ubiquitin likemodifier activating enzyme 5; UFC1: ubiquitin-fold modifierconjugating enzyme 1; UFD1: ubiquitin recognition factor in ERassociated degradation 1; UFL1: UFM1 specific ligase 1; UFM1:ubiquitin fold modifier 1; UFSP2: UFM1 specific peptidase 2; UVRAG:UV radiation resistance associated; VCP/p97: valosin containingprotein; WT: wild-type.
Collapse
Affiliation(s)
- Zhifeng Wang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Shuhui Xiong
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Zhaoyi Wu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Xingde Wang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Yamin Gong
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Wei-Guo Zhu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Xingzhi Xu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
15
|
Wang Z, Ma L, Meng Y, Fang J, Xu D, Lu Z. The interplay of the circadian clock and metabolic tumorigenesis. Trends Cell Biol 2024; 34:742-755. [PMID: 38061936 DOI: 10.1016/j.tcb.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 09/08/2024]
Abstract
The circadian clock and cell metabolism are both dysregulated in cancer cells through intrinsic cell-autonomous mechanisms and external influences from the tumor microenvironment. The intricate interplay between the circadian clock and cancer cell metabolism exerts control over various metabolic processes, including aerobic glycolysis, de novo nucleotide synthesis, glutamine and protein metabolism, lipid metabolism, mitochondrial metabolism, and redox homeostasis in cancer cells. Importantly, oncogenic signaling can confer a moonlighting function on core clock genes, effectively reshaping cellular metabolism to fuel cancer cell proliferation and drive tumor growth. These interwoven regulatory mechanisms constitute a distinctive feature of cancer cell metabolism.
Collapse
Affiliation(s)
- Zheng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Leina Ma
- Department of Oncology, The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, Shandong 266003, China
| | - Ying Meng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Jing Fang
- Department of Oncology, The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, Shandong 266003, China.
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China.
| |
Collapse
|
16
|
Ma Q, Li H, Song Z, Deng Z, Huang W, Liu Q. Fueling the fight against cancer: Exploring the impact of branched-chain amino acid catalyzation on cancer and cancer immune microenvironment. Metabolism 2024; 161:156016. [PMID: 39222743 DOI: 10.1016/j.metabol.2024.156016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Metabolism of Branched-chain amino acids (BCAAs) is essential for the nutrient necessities in mammals. Catalytic enzymes serve to direct the whole-body BCAAs oxidation which involve in the development of various metabolic disorders. The reprogrammed metabolic elements are also responsible for malignant oncogenic processes, and favor the formation of distinctive immunosuppressive microenvironment surrounding different cancers. The impotent immune surveillance related to BCAAs dysfunction is a novel topic to investigate. Here we focus on the BCAA catalysts that contribute to metabolic changes and dysregulated immune reactions in cancer progression. We summarize the current knowledge of BCAA catalyzation, highlighting the interesting roles of BCAA metabolism in the treatment of cancers.
Collapse
Affiliation(s)
- Qianquan Ma
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, China
| | - Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province
| | - Zhihao Song
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province.
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center For Skull Base Surgery and Neurooncology In Hunan Province.
| |
Collapse
|
17
|
Gubser PM, Wijesinghe S, Heyden L, Gabriel SS, de Souza DP, Hess C, McConville MM, Utzschneider DT, Kallies A. Aerobic glycolysis but not GLS1-dependent glutamine metabolism is critical for anti-tumor immunity and response to checkpoint inhibition. Cell Rep 2024; 43:114632. [PMID: 39159042 DOI: 10.1016/j.celrep.2024.114632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/04/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024] Open
Abstract
Tumor cells undergo uncontrolled proliferation driven by enhanced anabolic metabolism including glycolysis and glutaminolysis. Targeting these pathways to inhibit cancer growth is a strategy for cancer treatment. Critically, however, tumor-responsive T cells share metabolic features with cancer cells, making them susceptible to these treatments as well. Here, we assess the impact on anti-tumor T cell immunity and T cell exhaustion by genetic ablation of lactate dehydrogenase A (LDHA) and glutaminase1 (GLS1), key enzymes in aerobic glycolysis and glutaminolysis. Loss of LDHA severely impairs expansion of T cells in response to tumors and chronic infection. In contrast, T cells lacking GLS1 can compensate for impaired glutaminolysis by engaging alternative pathways, including upregulation of asparagine synthetase, and thus efficiently respond to tumor challenge and chronic infection as well as immune checkpoint blockade. Targeting GLS1-dependent glutaminolysis, but not aerobic glycolysis, may therefore be a successful strategy in cancer treatment, particularly in combination with immunotherapy.
Collapse
Affiliation(s)
- Patrick M Gubser
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia
| | - Sharanya Wijesinghe
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia
| | - Leonie Heyden
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia
| | - Sarah S Gabriel
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia
| | - David P de Souza
- Metabolomics Australia, Bio21 Molecular Science & Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Christoph Hess
- Department of Biomedicine, Immunobiology, University of Basel and University Hospital of Basel, 4031 Basel, Switzerland; Department of Medicine, CITIID, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Malcolm M McConville
- Metabolomics Australia, Bio21 Molecular Science & Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia; Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, Australia
| | - Daniel T Utzschneider
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
18
|
Grobben Y. Targeting amino acid-metabolizing enzymes for cancer immunotherapy. Front Immunol 2024; 15:1440269. [PMID: 39211039 PMCID: PMC11359565 DOI: 10.3389/fimmu.2024.1440269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Despite the immune system's role in the detection and eradication of abnormal cells, cancer cells often evade elimination by exploitation of various immune escape mechanisms. Among these mechanisms is the ability of cancer cells to upregulate amino acid-metabolizing enzymes, or to induce these enzymes in tumor-infiltrating immunosuppressive cells. Amino acids are fundamental cellular nutrients required for a variety of physiological processes, and their inadequacy can severely impact immune cell function. Amino acid-derived metabolites can additionally dampen the anti-tumor immune response by means of their immunosuppressive activities, whilst some can also promote tumor growth directly. Based on their evident role in tumor immune escape, the amino acid-metabolizing enzymes glutaminase 1 (GLS1), arginase 1 (ARG1), inducible nitric oxide synthase (iNOS), indoleamine 2,3-dioxygenase 1 (IDO1), tryptophan 2,3-dioxygenase (TDO) and interleukin 4 induced 1 (IL4I1) each serve as a promising target for immunotherapeutic intervention. This review summarizes and discusses the involvement of these enzymes in cancer, their effect on the anti-tumor immune response and the recent progress made in the preclinical and clinical evaluation of inhibitors targeting these enzymes.
Collapse
|
19
|
Strobl EV, Gamazon E. Discovering Root Causal Genes with High Throughput Perturbations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.13.574491. [PMID: 38260506 PMCID: PMC10802597 DOI: 10.1101/2024.01.13.574491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Root causal gene expression levels - or root causal genes for short - correspond to the initial changes to gene expression that generate patient symptoms as a downstream effect. Identifying root causal genes is critical towards developing treatments that modify disease near its onset, but no existing algorithms attempt to identify root causal genes from data. RNA-sequencing (RNA-seq) data introduces challenges such as measurement error, high dimensionality and non-linearity that compromise accurate estimation of root causal effects even with state-of-the-art approaches. We therefore instead leverage Perturb-seq, or high throughput perturbations with single cell RNA-seq readout, to learn the causal order between the genes. We then transfer the causal order to bulk RNA-seq and identify root causal genes specific to a given patient for the first time using a novel statistic. Experiments demonstrate large improvements in performance. Applications to macular degeneration and multiple sclerosis also reveal root causal genes that lie on known pathogenic pathways, delineate patient subgroups and implicate a newly defined omnigenic root causal model.
Collapse
|
20
|
Xu Z, Amakye WK, Ren Z, Xu Y, Liu W, Gong C, Wong C, Gao L, Zhao Z, Wang M, Yan T, Ye Z, Zhong J, Hou C, Zhao M, Qiu C, Tan J, Xu X, Liu G, Yao M, Ren J. Soy Peptide Supplementation Mitigates Undernutrition through Reprogramming Hepatic Metabolism in a Novel Undernourished Non-Human Primate Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306890. [PMID: 38816931 PMCID: PMC11304262 DOI: 10.1002/advs.202306890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/23/2024] [Indexed: 06/01/2024]
Abstract
In spite of recent advances in the field of undernutrition, current dietary therapy relying on the supply of high protein high calorie formulas is still plagued with transient recovery of impaired organs resulting in significant relapse of cases. This is partly attributed to the inadequacy of current research models in recapitulating clinical undernutrition for mechanistic exploration. Using 1636 Macaca fascicularis monkeys, a human-relevant criterion for determining undernutrition weight-for-age z-score (WAZ), with a cutoff point of ≤ -1.83 is established as the benchmark for identifying undernourished nonhuman primates (U-NHPs). In U-NHPs, pathological anomalies in multi-organs are revealed. In particular, severe dysregulation of hepatic lipid metabolism characterized by impaired fatty acid oxidation due to mitochondria dysfunction, but unlikely peroxisome disorder, is identified as the anchor metabolic aberration in U-NHPs. Mitochondria dysfunction is typified by reduced mito-number, accumulated long-chain fatty acids, and disruption of OXPHOS complexes. Soy peptide-treated U-NHPs increase in WAZ scores, in addition to attenuated mitochondria dysfunction and restored OXPHOS complex levels. Herein, innovative criteria for identifying U-NHPs are developed, and unknown molecular mechanisms of undernutrition are revealed hitherto, and it is further proved that soypeptide supplementation reprogramed mitochondrial function to re-establish lipid metabolism balance and mitigated undernutrition.
Collapse
Affiliation(s)
- Zhenzhen Xu
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - William Kwame Amakye
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Zhengyu Ren
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory DiseaseGuangzhou510182China
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
| | - Yongzhao Xu
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Wei Liu
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
- Huazhen Laboratory Animal Breeding CenterGuangzhou510900China
| | - Congcong Gong
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Chiwai Wong
- Huazhen Laboratory Animal Breeding CenterGuangzhou510900China
| | - Li Gao
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Zikuan Zhao
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Min Wang
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Tao Yan
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Zhiming Ye
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory DiseaseGuangzhou510182China
| | - Jun Zhong
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Chuanli Hou
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Miao Zhao
- Center for Medical Genetics and Hunan Key Laboratory of Medical GeneticsSchool of Life ScienceCentral South UniversityChangsha410013P. R. China
| | - Can Qiu
- Center for Medical Genetics and Hunan Key Laboratory of Medical GeneticsSchool of Life ScienceCentral South UniversityChangsha410013P. R. China
| | - Jieqiong Tan
- Center for Medical Genetics and Hunan Key Laboratory of Medical GeneticsSchool of Life ScienceCentral South UniversityChangsha410013P. R. China
| | - Xin Xu
- College of Food Science and EngineeringYangzhou UniversityYangzhou225127China
| | - Guoyan Liu
- College of Food Science and EngineeringYangzhou UniversityYangzhou225127China
| | - Maojin Yao
- The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory DiseaseGuangzhou510182China
| | - Jiaoyan Ren
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhou510640China
| |
Collapse
|
21
|
Zhao X, Ma D, Yang B, Wang Y, Zhang L. Research progress of T cell autophagy in autoimmune diseases. Front Immunol 2024; 15:1425443. [PMID: 39104538 PMCID: PMC11298352 DOI: 10.3389/fimmu.2024.1425443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024] Open
Abstract
T cells, as a major lymphocyte population involved in the adaptive immune response, play an important immunomodulatory role in the early stages of autoimmune diseases. Autophagy is a cellular catabolism mediated by lysosomes. Autophagy maintains cell homeostasis by recycling degraded cytoplasmic components and damaged organelles. Autophagy has a protective effect on cells and plays an important role in regulating T cell development, activation, proliferation and differentiation. Autophagy mediates the participation of T cells in the acquired immune response and plays a key role in antigen processing as well as in the maintenance of T cell homeostasis. In autoimmune diseases, dysregulated autophagy of T cells largely influences the pathological changes. Therefore, it is of great significance to study how T cells play a role in the immune mechanism of autoimmune diseases through autophagy pathway to guide the clinical treatment of diseases.
Collapse
Affiliation(s)
| | | | | | | | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
22
|
Elias MG, Fatima S, Mann TJ, Karan S, Mikhael M, de Souza P, Gordon CP, Scott KF, Aldrich-Wright JR. Anticancer Effect of Pt IIPHEN SS, Pt II5ME SS, Pt II56ME SS and Their Platinum(IV)-Dihydroxy Derivatives against Triple-Negative Breast Cancer and Cisplatin-Resistant Colorectal Cancer. Cancers (Basel) 2024; 16:2544. [PMID: 39061185 PMCID: PMC11274883 DOI: 10.3390/cancers16142544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Development of resistance to cisplatin, oxaliplatin and carboplatin remains a challenge for their use as chemotherapies, particularly in breast and colorectal cancer. Here, we compare the anticancer effect of novel complexes [Pt(1,10-phenanthroline)(1S,2S-diaminocyclohexane)](NO3)2 (PtIIPHENSS), [Pt(5-methyl-1,10-phenanthroline)(1S,2S-diaminocyclohexane)](NO3)2 (PtII5MESS) and [Pt(5,6-dimethyl-1,10-phenanthroline)(1S,2S-diaminocyclohexane)](NO3)2 (PtII56MESS) and their platinum(IV)-dihydroxy derivatives with cisplatin. Complexes are greater than 11-fold more potent than cisplatin in both 2D and 3D cell line cultures with increased selectivity for cancer cells over genetically stable cells. ICP-MS studies showed cellular uptake occurred through an active transport mechanism with considerably altered platinum concentrations found in the cytoskeleton across all complexes after 24 h. Significant reactive oxygen species generation was observed, with reduced mitochondrial membrane potential at 72 h of treatment. Late apoptosis/necrosis was shown by Annexin V-FITC/PI flow cytometry assay, accompanied by increased sub-G0/G1 cells compared with untreated cells. An increase in S and G2+M cells was seen with all complexes. Treatment resulted in significant changes in actin and tubulin staining. Intrinsic and extrinsic apoptosis markers, MAPK/ERK and PI3K/AKT activation markers, together with autophagy markers showed significant activation of these pathways by Western blot. The proteomic profile investigated post-72 h of treatment identified 1597 MDA-MB-231 and 1859 HT29 proteins quantified by mass spectroscopy, with several differentially expressed proteins relative to no treatment. GO enrichment analysis revealed a statistically significant enrichment of RNA/DNA-associated proteins in both the cell lines and specific additional processes for individual drugs. This study shows that these novel agents function as multi-mechanistic chemotherapeutics, offering promising anticancer potential, and thereby supporting further research into their application as cancer therapeutics.
Collapse
Affiliation(s)
- Maria George Elias
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
| | - Shadma Fatima
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Timothy J. Mann
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Shawan Karan
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
| | - Meena Mikhael
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
| | - Paul de Souza
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Kingswood, NSW 2747, Australia;
| | - Christopher P. Gordon
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
| | - Kieran F. Scott
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Janice R. Aldrich-Wright
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| |
Collapse
|
23
|
Dou X, Chen K, Brown MR, Strand MR. Reciprocal interactions between neuropeptide F and RYamide regulate host attraction in the mosquito Aedes aegypti. Proc Natl Acad Sci U S A 2024; 121:e2408072121. [PMID: 38950363 PMCID: PMC11252962 DOI: 10.1073/pnas.2408072121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 07/03/2024] Open
Abstract
Female mosquitoes produce eggs in gonadotrophic cycles that are divided between a previtellogenic and vitellogenic phase. Previtellogenic females consume water and sugar sources like nectar while also being attracted to hosts for blood feeding. Consumption of a blood meal activates the vitellogenic phase, which produces mature eggs and suppresses host attraction. In this study, we tested the hypothesis that neuropeptide Y-like hormones differentially modulate host attraction behavior in the mosquito Aedes aegypti. A series of experiments collectively indicated that enteroendocrine cells (EECs) in the posterior midgut produce and release neuropeptide F (NPF) into the hemolymph during the previtellogenic phase which stimulates attraction to humans and biting behavior. Consumption of a blood meal, which primarily consists of protein by dry weight, down-regulated NPF in EECs until mature eggs developed, which was associated with a decline in hemolymph titer. NPF depletion depended on protein digestion but was not associated with EEC loss. Other experiments showed that neurons in the terminal ganglion extend axons to the posterior midgut and produce RYamide, which showed evidence of increased secretion into circulation after a blood meal. Injection of RYamide-1 and -2 into previtellogenic females suppressed host attraction, while coinjection of RYamides with or without short NPF-2 also inhibited the host attraction activity of NPF. Overall, our results identify NPF and RYamide as gut-associated hormones in A. aegypti that link host attraction behavior to shifts in diet during sequential gonadotrophic cycles.
Collapse
Affiliation(s)
- Xiaoyi Dou
- Department of Entomology, University of Georgia, Athens, GA30602
| | - Kangkang Chen
- Department of Entomology, University of Georgia, Athens, GA30602
| | - Mark R. Brown
- Department of Entomology, University of Georgia, Athens, GA30602
| | | |
Collapse
|
24
|
Zhao J, Duan L, Li J, Yao C, Wang G, Mi J, Yu Y, Ding L, Zhao Y, Yan G, Li J, Zhao Z, Wang X, Li M. New insights into the interplay between autophagy, gut microbiota and insulin resistance in metabolic syndrome. Biomed Pharmacother 2024; 176:116807. [PMID: 38795644 DOI: 10.1016/j.biopha.2024.116807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Metabolic syndrome (MetS) is a widespread and multifactorial disorder, and the study of its pathogenesis and treatment remains challenging. Autophagy, an intracellular degradation system that maintains cellular renewal and homeostasis, is essential for maintaining antimicrobial defense, preserving epithelial barrier integrity, promoting mucosal immune response, maintaining intestinal homeostasis, and regulating gut microbiota and microbial metabolites. Dysfunctional autophagy is implicated in the pathological mechanisms of MetS, involving insulin resistance (IR), chronic inflammation, oxidative stress, and endoplasmic reticulum (ER) stress, with IR being a predominant feature. The study of autophagy represents a valuable field of research with significant clinical implications for identifying autophagy-related signals, pathways, mechanisms, and treatment options for MetS. Given the multifactorial etiology and various potential risk factors, it is imperative to explore the interplay between autophagy and gut microbiota in MetS more thoroughly. This will facilitate the elucidation of new mechanisms underlying the crosstalk among autophagy, gut microbiota, and MetS, thereby providing new insights into the diagnosis and treatment of MetS.
Collapse
Affiliation(s)
- Jinyue Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Liyun Duan
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jiarui Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Chensi Yao
- Molecular Biology Laboratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guoqiang Wang
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jia Mi
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yongjiang Yu
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Lu Ding
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yunyun Zhao
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Guanchi Yan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jing Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Zhixuan Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Xiuge Wang
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China.
| | - Min Li
- Molecular Biology Laboratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
25
|
Villagomez FR, Lang J, Rosario FJ, Nunez-Avellaneda D, Webb P, Neville M, Woodruff ER, Bitler BG. Claudin-4 Modulates Autophagy via SLC1A5/LAT1 as a Mechanism to Regulate Micronuclei. CANCER RESEARCH COMMUNICATIONS 2024; 4:1625-1642. [PMID: 38867360 PMCID: PMC11218812 DOI: 10.1158/2767-9764.crc-24-0240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/21/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
Genome instability is a hallmark of cancer crucial for tumor heterogeneity and is often a result of defects in cell division and DNA damage repair. Tumors tolerate genomic instability, but the accumulation of genetic aberrations is regulated to avoid catastrophic chromosomal alterations and cell death. In ovarian cancer tumors, claudin-4 is frequently upregulated and closely associated with genome instability and worse patient outcomes. However, its biological association with regulating genomic instability is poorly understood. Here, we used CRISPR interference and a claudin mimic peptide to modulate the claudin-4 expression and its function in vitro and in vivo. We found that claudin-4 promotes a tolerance mechanism for genomic instability through micronuclei generation in tumor cells. Disruption of claudin-4 increased autophagy and was associated with the engulfment of cytoplasm-localized DNA. Mechanistically, we observed that claudin-4 establishes a biological axis with the amino acid transporters SLC1A5 and LAT1, which regulate autophagy upstream of mTOR. Furthermore, the claudin-4/SLC1A5/LAT1 axis was linked to the transport of amino acids across the plasma membrane as one of the potential cellular processes that significantly decreased survival in ovarian cancer patients. Together, our results show that the upregulation of claudin-4 contributes to increasing the threshold of tolerance for genomic instability in ovarian tumor cells by limiting its accumulation through autophagy. SIGNIFICANCE Autophagy regulation via claudin-4/SLC1A5/LAT1 has the potential to be a targetable mechanism to interfere with genomic instability in ovarian tumor cells.
Collapse
Affiliation(s)
- Fabian R. Villagomez
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Julie Lang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Fredrick J. Rosario
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Daniel Nunez-Avellaneda
- Deputy Directorate of Technological Development, Linkage, and Innovation, National Council of Humanities, Sciences, and Technologies, Mexico City, Mexico.
| | - Patricia Webb
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Margaret Neville
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Elizabeth R. Woodruff
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Benjamin G. Bitler
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
26
|
Guo S, Wang X, Wang Y, Bai J, Liu Y, Shao Z. The potential therapeutic targets of glutamine metabolism in head and neck squamous cell carcinoma. Biomed Pharmacother 2024; 176:116906. [PMID: 38876051 DOI: 10.1016/j.biopha.2024.116906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024] Open
Abstract
Targeting metabolic reprogramming may be an effective strategy to enhance cancer treatment efficacy. Glutamine serves as a vital nutrient for cancer cells. Inhibiting glutamine metabolism has shown promise in preventing tumor growth both in vivo and in vitro through various mechanisms. Therefore, this review collates recent scientific literature concerning the correlation between glutamine metabolism and cancer treatment. Novel treatment modalities based on amino acid transporters, metabolites, and glutaminase are discussed. Moreover, we demonstrate the relationship between glutamine metabolism and tumor proliferation, drug resistance, and the tumor immune microenvironment, offering new perspectives for the clinical treatment of head and neck squamous cell carcinoma, particularly for combined therapies. Identifying innovative approaches for enhancing the efficacy of glutamine-based metabolic therapy is crucial to improving HNSCC treatment.
Collapse
Affiliation(s)
- Shutian Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China; Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xinmiao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China; Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yifan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China; Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junqiang Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China; Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi Liu
- Department of stomatology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Huangshi 435000, China.
| | - Zhe Shao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China; Day Surgery Center, School and Hospital of Stomatology, Wuhan University, China.
| |
Collapse
|
27
|
Zheng M, Wang Z, Li M, Yang N, Lu H, Zhang Z, Dong Y, Chen Y, Zhu Z, Tong A, Yang H. A novel SLC3A2-targeting antibody-drug conjugate exerts potent antitumor efficacy in head and neck squamous cell cancer. Transl Oncol 2024; 45:101981. [PMID: 38703658 PMCID: PMC11088350 DOI: 10.1016/j.tranon.2024.101981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/03/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024] Open
Abstract
The development of innovative therapeutic strategies for head and neck squamous cell carcinoma (HNSCC) is a critical medical requirement. Antibody-drug conjugates (ADC) targeting tumor-specific surface antigens have demonstrated clinical effectiveness in treating hematologic and solid malignancies. Our investigation revealed high expression levels of SLC3A2 in HNSCC tissue and cell lines. This study aimed to develop a novel anti-SLC3A2 ADC and assess its antitumor effects on HNSCC both in vitro and in vivo. This study developed a potent anti-SLC3A2 ADC (19G4-MMAE) and systematically investigated its drug delivery potential and antitumor efficacy in preclinical models. This study revealed that 19G4-MMAE exhibited specific binding to SLC3A2 and effectively targeted lysosomes. Moreover, 19G4-MMAE induced a significant accumulation of reactive oxygen species (ROS) and apoptosis in SLC3A2-positive HNSCC cells. The compound demonstrated potent antitumor effects derived from MMAE against SLC3A2-expressing HNSCC in preclinical models, displaying a favorable safety profile. These findings suggest that targeting SLC3A2 with an anti-SLC3A2 ADC could be a promising therapeutic approach for treating HNSCC patients.
Collapse
Affiliation(s)
- Meijun Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Zeng Wang
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Mengyao Li
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Nian Yang
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Huaqing Lu
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Yijun Dong
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Yongdong Chen
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Zhixiong Zhu
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Aiping Tong
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China.
| | - Hui Yang
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, PR China.
| |
Collapse
|
28
|
Jiang J, Tang Y, Cao Z, Zhou C, Yu Z. Effects of hypo-osmotic stress on osmoregulation, antioxidant response, and energy metabolism in sea cucumber Holothuria moebii under desalination environment. ENVIRONMENTAL RESEARCH 2024; 252:118800. [PMID: 38555088 DOI: 10.1016/j.envres.2024.118800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
With global climate changing, hypo-salinity events are increasing in frequency and duration because of continuous rainfall and freshwater inflow, which causes reduced cytosolic osmolarity and cellular stress responses in aquatic animals. Sea cucumbers are considered stenohaline because they lack osmoregulatory organs and are vulnerable to salinity fluctuations. In this study, we performed multiple biochemical assays, de novo transcriptomics, and widely targeted metabolomics to comprehensively explore the osmoregulatory mechanisms and physiological responses of sea cucumber Holothuria moebii to hypo-osmotic stress, which is a representative specie that is frequently exposed to hypo-saline intertidal zones. Our results found that H. moebii contracted their ambulacral feet and oral tentacles, and the coelomic fluid ion concentrations were reduced to be consistent with the environment. The microvilli of intestines and respiratory trees underwent degeneration, and the cytoplasm exhibited swelling and vacuolation. Moreover, the Na+, K+, and Cl- concentrations and Na+/K+-ATPase activity were significantly reduced under hypo-osmotic stress. The decrease in protein kinase A activity and increase in 5'-AMP level indicated a significant inhibition of the cAMP signaling pathway to regulate ion concentrations. And small intracellular organic molecules (amino acids, nucleotides and their derivatives) also play crucial roles in osmoregulation through oxidative deamination of glutamate, nucleotide catabolism, and nucleic acid synthesis. Moreover, lysosomes and peroxisomes removed oxidative damage, whereas antioxidant metabolites, such as N-acetyl amino acids and glutathione, were increased to resist oxidative stress. With prolonged hypo-osmotic stress, glycerophospholipid metabolism was enhanced to maintain membrane stability. Furthermore, acyl-CoA-binding protein activity was significantly inhibited, and only a small amount of acylcarnitine was significantly accumulated, which indicated a disruption in energy metabolism. PPAR signaling pathway and choline content were up-regulated to promote fatty acid metabolism under hypo-osmotic stress. Overall, our results provide new insights into the osmoregulatory mechanisms and physiological responses of sea cucumbers to hypo-osmotic stress.
Collapse
Affiliation(s)
- Junyang Jiang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yanna Tang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Zhaozhao Cao
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Cong Zhou
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China.
| | - Zonghe Yu
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
29
|
Gröner B, Hoffmann C, Endepols H, Urusova EA, Brugger M, Neumaier F, Timmer M, Neumaier B, Zlatopolskiy BD. Radiosynthesis and Preclinical Evaluation of m-[ 18F]FET and [ 18F]FET-OMe as Novel [ 18F]FET Analogs for Brain Tumor Imaging. Mol Pharm 2024; 21:2795-2812. [PMID: 38747353 DOI: 10.1021/acs.molpharmaceut.3c01215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
O-([18F]Fluoroethyl)-l-tyrosine ([18F]FET) is actively transported into the brain and cancer cells by LAT1 and possibly other amino acid transporters, which enables brain tumor imaging by positron emission tomography (PET). However, tumor delivery of this probe in the presence of competing amino acids may be limited by a relatively low affinity for LAT1. The aim of the present work was to evaluate the meta-substituted [18F]FET analog m-[18F]FET and the methyl ester [18F]FET-OMe, which were designed to improve tumor delivery by altering the physicochemical, pharmacokinetic, and/or transport properties. Both tracers could be prepared with good radiochemical yields of 41-56% within 66-90 min. Preclinical evaluation with [18F]FET as a reference tracer demonstrated reduced in vitro uptake of [18F]FET-OMe by U87 glioblastoma cells and no advantage for in vivo tumor imaging. In contrast, m-[18F]FET showed significantly improved in vitro uptake and accelerated in vivo tumor accumulation in an orthotopic glioblastoma model. As such, our work identifies m-[18F]FET as a promising alternative to [18F]FET for brain tumor imaging that deserves further evaluation with regard to its transport properties and in vivo biodistribution.
Collapse
Affiliation(s)
- Benedikt Gröner
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Chris Hoffmann
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Heike Endepols
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
- Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Elizaveta A Urusova
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Melanie Brugger
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
| | - Felix Neumaier
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Marco Timmer
- Faculty of Medicine and University Hospital Cologne, Center for Neurosurgery, Department of General Neurosurgery, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Bernd Neumaier
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| | - Boris D Zlatopolskiy
- Forschungszentrum Jülich GmbH, Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Wilhelm-Johnen-Straße, Jülich 52428, Germany
- Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Kerpener Straße 62, Cologne 50937, Germany
| |
Collapse
|
30
|
Das C, Bhattacharya A, Adhikari S, Mondal A, Mondal P, Adhikary S, Roy S, Ramos K, Yadav KK, Tainer JA, Pandita TK. A prismatic view of the epigenetic-metabolic regulatory axis in breast cancer therapy resistance. Oncogene 2024; 43:1727-1741. [PMID: 38719949 PMCID: PMC11161412 DOI: 10.1038/s41388-024-03054-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
Epigenetic regulation established during development to maintain patterns of transcriptional expression and silencing for metabolism and other fundamental cell processes can be reprogrammed in cancer, providing a molecular mechanism for persistent alterations in phenotype. Metabolic deregulation and reprogramming are thus an emerging hallmark of cancer with opportunities for molecular classification as a critical preliminary step for precision therapeutic intervention. Yet, acquisition of therapy resistance against most conventional treatment regimens coupled with tumor relapse, continue to pose unsolved problems for precision healthcare, as exemplified in breast cancer where existing data informs both cancer genotype and phenotype. Furthermore, epigenetic reprograming of the metabolic milieu of cancer cells is among the most crucial determinants of therapeutic resistance and cancer relapse. Importantly, subtype-specific epigenetic-metabolic interplay profoundly affects malignant transformation, resistance to chemotherapy, and response to targeted therapies. In this review, we therefore prismatically dissect interconnected epigenetic and metabolic regulatory pathways and then integrate them into an observable cancer metabolism-therapy-resistance axis that may inform clinical intervention. Optimally coupling genome-wide analysis with an understanding of metabolic elements, epigenetic reprogramming, and their integration by metabolic profiling may decode missing molecular mechanisms at the level of individual tumors. The proposed approach of linking metabolic biochemistry back to genotype, epigenetics, and phenotype for specific tumors and their microenvironment may thus enable successful mechanistic targeting of epigenetic modifiers and oncometabolites despite tumor metabolic heterogeneity.
Collapse
Affiliation(s)
- Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India.
- Homi Bhabha National Institute, Mumbai, 400094, India.
| | - Apoorva Bhattacharya
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
| | - Swagata Adhikari
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Payel Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Siddhartha Roy
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Kenneth Ramos
- Center for Genomics and Precision Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA
| | - Kamlesh K Yadav
- Center for Genomics and Precision Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA
- School of Engineering Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA
| | - John A Tainer
- The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Tej K Pandita
- Center for Genomics and Precision Medicine, Texas A&M University, School of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
31
|
Simon-Molas H, Montironi C, Kabanova A, Eldering E. Metabolic reprogramming in the CLL TME; potential for new therapeutic targets. Semin Hematol 2024; 61:155-162. [PMID: 38493076 DOI: 10.1053/j.seminhematol.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/28/2024] [Accepted: 02/12/2024] [Indexed: 03/18/2024]
Abstract
Chronic lymphocytic leukemia (CLL) cells circulate between peripheral (PB) blood and lymph node (LN) compartments, and strictly depend on microenvironmental factors for proliferation, survival and drug resistance. All cancer cells display metabolic reprogramming and CLL is no exception - though the inert status of the PB CLL cells has hampered detailed insight into these processes. We summarize previous work on reactive oxygen species (ROS), oxidative stress, and hypoxia, as well as the important roles of Myc, and PI3K/Akt/mTor pathways. In vitro co-culture systems and gene expression analyses have provided a partial picture of CLL LN metabolism. New broad omics techniques allow to obtain molecular and also single-cell level understanding of CLL plasticity and metabolic reprogramming. We summarize recent developments and describe the new concept of glutamine addiction for CLL, which may hold therapeutic promise.
Collapse
Affiliation(s)
- Helga Simon-Molas
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands; Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Cancer Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Chiara Montironi
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands; Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Cancer Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Anna Kabanova
- Tumour Immunology Unit, Toscana Life Sciences Foundation, Siena, Italy
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands; Cancer Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands; Cancer Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
32
|
Chen Y, Liu T, Hu D, Hu T, Ye C, Mu W. Histology, fatty acid composition, antioxidant and glycolipid metabolism, and transcriptome analyses of the acute cold stress response in Phoxinus lagowskii. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 50:101242. [PMID: 38729031 DOI: 10.1016/j.cbd.2024.101242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
Water temperature is a crucial environmental factor that significantly affects the physiological and biochemical processes of fish. Due to the occurrence of cold events in aquaculture, it is imperative to investigate how fish respond to cold stress. This study aims to uncover the mechanisms responds to acute cold stress by conducting a comprehensive analysis of the histomorphology, glycolipid metabolic and antioxidant enzymes, fatty acid composition and transcriptome at three temperatures (16 °C, 10 °C and 4 °C) in Phoxinus lagowskii. Our results showed that cold stress not damaged muscle microstructure but caused autophagy (at 10 °C). In addition, serum glucose (Glu) and triglycerides (TG) increased during cold stress. The activities of reactive oxygen species (ROS), superoxide dismutase (SOD), catalase (CAT), fructose phosphokinase (PFK), hexokinase (HK), pyruvate kinase (PK), and malondialdehyde (MDA) content in muscle were measured and analyzed. During cold stress, superoxide dismutase and catalase activities increased, reactive oxygen species content decreased. No significant difference in Glutathione peroxidase (GPx) activity, malondialdehyde and total cholesterol (T-CHO) contents among groups. Phosphokinase and pyruvate kinase activities decreased, and HK activity increased during cold stress. Our study resulted in the identification of a total of 25,400 genes, with 2524 genes showing differential expression across different temperature treatments. Furthermore, KEGG pathway indicated that some pathways upregulated during light cold stress (at 10 °C, including autophagy, and AMP-activated protein kinase (AMPK) signaling pathway. Additionally, circadian rhythm is among the most enriched pathways in genes up-regulated during severe cold stress (at 4 °C). Our findings offer valuable insights into how cold-water fish respond to cold stress.
Collapse
Affiliation(s)
- Yingqiao Chen
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Tianmei Liu
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Deer Hu
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Tingting Hu
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Cunrun Ye
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China
| | - Weijie Mu
- Key Laboratory of Biodiversity of Aquatic Organisms, College of Life Science and Technology, Harbin Normal University, Harbin 150025, China.
| |
Collapse
|
33
|
Göttig L, Schreiner S. E4orf1: The triple agent of adenovirus - Unraveling its roles in oncogenesis, infectious obesity and immune responses in virus replication and vector therapy. Tumour Virus Res 2024; 17:200277. [PMID: 38428735 PMCID: PMC10937242 DOI: 10.1016/j.tvr.2024.200277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
Human Adenoviruses (HAdV) are nearly ubiquitous pathogens comprising numerous sub-types that infect various tissues and organs. Among many encoded proteins that facilitate viral replication and subversion of host cellular processes, the viral E4orf1 protein has emerged as an intriguing yet under-investigated player in the complex interplay between the virus and its host. E4orf1 has gained attention as a metabolism activator and oncogenic agent, while recent research is showing that E4orf1 may play a more important role in modulating cellular pathways such as PI3K-Akt-mTOR, Ras, the immune response and further HAdV replication stages than previously anticipated. In this review, we aim to explore the structure, molecular mechanisms, and biological functions of E4orf1, shedding light on its potentially multifaceted roles during HAdV infection, including metabolic diseases and oncogenesis. Furthermore, we discuss the role of functional E4orf1 in biotechnological applications such as Adenovirus (AdV) vaccine vectors and oncolytic AdV. By dissecting the intricate relationships between HAdV types and E4orf1 proteins, this review provides valuable insights into viral pathogenesis and points to promising areas of future research.
Collapse
Affiliation(s)
- Lilian Göttig
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
| | - Sabrina Schreiner
- Institute of Virology, School of Medicine, Technical University of Munich, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (Resolving Infection Susceptibility; EXC 2155), Hannover, Germany; Institute of Virology, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
34
|
Shi J, Pabon K, Ding R, Scotto KW. ABCG2 and SLC1A5 functionally interact to rewire metabolism and confer a survival advantage to cancer cells under oxidative stress. J Biol Chem 2024; 300:107299. [PMID: 38641063 PMCID: PMC11131071 DOI: 10.1016/j.jbc.2024.107299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/21/2024] Open
Abstract
ABCG2, a member of the ABC transporter superfamily, is overexpressed in many human tumors and has long been studied for its ability to export a variety of chemotherapeutic agents, thereby conferring a multidrug resistance (MDR) phenotype. However, several studies have shown that ABCG2 can also confer an MDR-independent survival advantage to tumor cells exposed to stress. While investigating the mechanism by which ABCG2 enhances survival in stressful milieus, we have identified a physical and functional interaction between ABCG2 and SLC1A5, a member of the solute transporter superfamily and the primary transporter of glutamine in cancer cells. This interaction was accompanied by increased glutamine uptake, increased glutaminolysis, and rewired cellular metabolism, as evidenced by an increase in key metabolic enzymes and alteration of glutamine-dependent metabolic pathways. Specifically, we observed an increase in glutamine metabolites shuttled to the TCA cycle, and an increase in the synthesis of glutathione, accompanied by a decrease in basal levels of reactive oxygen species and a marked increase in cell survival in the face of oxidative stress. Notably, the knockdown of SLC1A5 or depletion of exogenous glutamine diminished ABCG2-enhanced autophagy flux, further implicating this solute transporter in ABCG2-mediated cell survival. This is, to our knowledge, the first report of a functionally significant physical interaction between members of the two major transporter superfamilies. Moreover, these observations may underlie the protective role of ABCG2 in cancer cells under duress and suggest a novel role for ABCG2 in the regulation of metabolism in normal and diseased states.
Collapse
Affiliation(s)
- Jia Shi
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Kirk Pabon
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Rui Ding
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA; Clinical Pharmacology, Translational Medicine, Servier Pharmaceuticals LLC, Boston, Massachusetts, USA
| | - Kathleen W Scotto
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA.
| |
Collapse
|
35
|
Ni M, Yue Z, Tian M, Luo X, Wang W, Shi H, Luo J, Deng L, Li C. Leucine-Mediated SLC7A5 Promotes Milk Protein and Milk Fat Synthesis through mTOR Signaling Pathway in Goat Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:13728-13739. [PMID: 38807030 PMCID: PMC11192034 DOI: 10.1021/acs.jafc.4c02087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
The SLC7A5 gene encodes a Na+ and pH-independent transporter protein that regulates cell growth by regulating the uptake of AA. This study, utilizing RNA-seq, aimed to explore the effect of SLC7A5 on the synthesis of milk proteins and fats in goat mammary epithelial cells (GMECs) through gene interference and overexpression techniques. The results demonstrated that the overexpression of SLC7A5 resulted in a significant increase in the expression of CSN1S1, SCD, CEBPB, ACACA, αS1-casein, p-S6K, and p-S6. The levels of p-S6K and p-S6 gradually increased as the AA/Leu stimulation time lengthened. The overexpression of SLC7A5 rescued the role of Torin1 in GMECs. In conclusion, SLC7A5 plays a crucial role in promoting the synthesis of milk proteins and milk fats through the mTOR signaling pathway in GMECs, providing a theoretical foundation for improving the quality of goat milk.
Collapse
Affiliation(s)
- Mengke Ni
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Ziting Yue
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Min Tian
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Xinran Luo
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Wanting Wang
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Huaiping Shi
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Jun Luo
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Lu Deng
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Cong Li
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
36
|
Lyda BR, Leary GP, Farnsworth J, Seaver B, Silvius D, Kavanaugh MP, Esslinger CS, Natale NR. Discovery and Synthesis of Hydroxy-l-Proline Blockers of the Neutral Amino Acid Transporters SLC1A4 (ASCT1) and SLC1A5 (ASCT2). Molecules 2024; 29:2330. [PMID: 38792190 PMCID: PMC11123841 DOI: 10.3390/molecules29102330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
As a conformationally restricted amino acid, hydroxy-l-proline is a versatile scaffold for the synthesis of diverse multi-functionalized pyrrolidines for probing the ligand binding sites of biological targets. With the goal to develop new inhibitors of the widely expressed amino acid transporters SLC1A4 and SLC1A5 (also known as ASCT1 and ASCT2), we synthesized and functionally screened synthetic hydroxy-l-proline derivatives using electrophysiological and radiolabeled uptake methods against amino acid transporters from the SLC1, SLC7, and SLC38 solute carrier families. We have discovered a novel class of alkoxy hydroxy-pyrrolidine carboxylic acids (AHPCs) that act as selective high-affinity inhibitors of the SLC1 family neutral amino acid transporters SLC1A4 and SLC1A5. AHPCs were computationally docked into a homology model and assessed with respect to predicted molecular orientation and functional activity. The series of hydroxyproline analogs identified here represent promising new agents to pharmacologically modulate SLC1A4 and SLC1A5 amino acid exchangers which are implicated in numerous pathophysiological processes such as cancer and neurological diseases.
Collapse
Affiliation(s)
- Brent R. Lyda
- Division of Biological Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Gregory P. Leary
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Jill Farnsworth
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Benjamin Seaver
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Derek Silvius
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Michael P. Kavanaugh
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - C. Sean Esslinger
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
| | - Nicholas R. Natale
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA (B.S.); (D.S.)
- Medicinal Chemistry Graduate Program, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA
| |
Collapse
|
37
|
Ogawa M, Tanaka A, Maekawa M, Namba K, Otani Y, Shia J, Wang JY, Roehrl MH. Protein expression of the amino acid transporter SLC7A5 in tumor tissue is prognostic in early-stage colorectal cancer. PLoS One 2024; 19:e0298362. [PMID: 38722983 PMCID: PMC11081336 DOI: 10.1371/journal.pone.0298362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 01/24/2024] [Indexed: 05/13/2024] Open
Abstract
Proteins overexpressed in early-stage cancers may serve as early diagnosis and prognosis markers as well as targets for cancer therapies. In this study, we examined the expression of an essential amino acid carrier SLC7A5 (LAT1, CD98, or 4F2 light chain) in cancer tissue from two well-annotated cohorts of 575 cases of early-stage and 106 cases of late-stage colorectal cancer patients. Immunohistochemistry showed SLC7A5 overexpression in 72.0% of early-stage and 56.6% of late-stage cases. SLC7A5 expression was not influenced by patient gender, age, location, or mismatch repair status, although it appeared to be slightly less prevalent in tumors of mucinous differentiation or with lymphovascular invasion. Statistical analyses revealed a positive correlation between SLC7A5 overexpression and both overall survival and disease-free survival in early-stage but not late-stage cancers. Co-expression analyses of the TCGA and CPTAC colorectal cancer cohorts identified a network of gene transcripts positively related to SLC7A5, with its heterodimer partner SLC3A2 having the highest co-expression score. Network analysis uncovered the SLC7A network to be significantly associated with ncRNA such as tRNA processing and the mitotic cell cycle. Since SLC7A5 is also a marker of activated lymphocytes such as NK, T, and B lymphocytes, SLC7A5 overexpression in early colorectal cancers might trigger a strong anti-tumor immune response which could results in better clinical outcome. Overall, our study provides clear evidence of differential SLC7A5 expression and its prognostic value for early-stage colorectal cancer, although the understanding of its functions in colorectal tumorigenesis and cancer immunity is currently rather limited and awaits further characterization.
Collapse
Affiliation(s)
- Makiko Ogawa
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Atsushi Tanaka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Masaki Maekawa
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Kei Namba
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Yusuke Otani
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Jinru Shia
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | | | - Michael H. Roehrl
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
38
|
Profir M, Roşu OA, Creţoiu SM, Gaspar BS. Friend or Foe: Exploring the Relationship between the Gut Microbiota and the Pathogenesis and Treatment of Digestive Cancers. Microorganisms 2024; 12:955. [PMID: 38792785 PMCID: PMC11124004 DOI: 10.3390/microorganisms12050955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Digestive cancers are among the leading causes of cancer death in the world. However, the mechanisms of cancer development and progression are not fully understood. Accumulating evidence in recent years pointing to the bidirectional interactions between gut dysbiosis and the development of a specific type of gastrointestinal cancer is shedding light on the importance of this "unseen organ"-the microbiota. This review focuses on the local role of the gut microbiota imbalance in different digestive tract organs and annexes related to the carcinogenic mechanisms. Microbiota modulation, either by probiotic administration or by dietary changes, plays an important role in the future therapies of various digestive cancers.
Collapse
Affiliation(s)
- Monica Profir
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Severus Gaspar
- Surgery Clinic, Emergency Clinical Hospital of Bucharest, 014461 Bucharest, Romania;
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
39
|
Ziki RA, Colnot S. Glutamine metabolism, a double agent combating or fuelling hepatocellular carcinoma. JHEP Rep 2024; 6:101077. [PMID: 38699532 PMCID: PMC11063524 DOI: 10.1016/j.jhepr.2024.101077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/16/2024] [Accepted: 02/28/2024] [Indexed: 05/05/2024] Open
Abstract
The reprogramming of glutamine metabolism is a key event in cancer more generally and in hepatocellular carcinoma (HCC) in particular. Glutamine consumption supplies tumours with ATP and metabolites through anaplerosis of the tricarboxylic acid cycle, while glutamine production can be enhanced by the overexpression of glutamine synthetase. In HCC, increased glutamine production is driven by activating mutations in the CTNNB1 gene encoding β-catenin. Increased glutamine synthesis or utilisation impacts tumour epigenetics, oxidative stress, autophagy, immunity and associated pathways, such as the mTOR (mammalian target of rapamycin) pathway. In this review, we will discuss studies which emphasise the pro-tumoral or tumour-suppressive effect of glutamine overproduction. It is clear that more comprehensive studies are needed as a foundation from which to develop suitable therapies targeting glutamine metabolic pathways, depending on the predicted pro- or anti-tumour role of dysregulated glutamine metabolism in distinct genetic contexts.
Collapse
Affiliation(s)
- Razan Abou Ziki
- INSERM, Sorbonne Université, Centre de Recherche des Cordeliers (CRC), Paris, F-75006, France
- Équipe labellisée Ligue Nationale Contre le Cancer, France
| | - Sabine Colnot
- INSERM, Sorbonne Université, Centre de Recherche des Cordeliers (CRC), Paris, F-75006, France
- Équipe labellisée Ligue Nationale Contre le Cancer, France
| |
Collapse
|
40
|
Ma Y, Okuda S, Okanishi H, Xu M, Jin C, Endou H, Ohgaki R, Kanai Y. Upregulation of ATF4 mediates the cellular adaptation to pharmacologic inhibition of amino acid transporter LAT1 in pancreatic ductal adenocarcinoma cells. J Pharmacol Sci 2024; 155:14-20. [PMID: 38553134 DOI: 10.1016/j.jphs.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/05/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
L-type amino acid transporter 1 (LAT1) is recognized as a promising target for cancer therapy; however, the cellular adaptive response to its pharmacological inhibition remains largely unexplored. This study examined the adaptive response to LAT1 inhibition using nanvuranlat, a high-affinity LAT1 inhibitor. Proteomic analysis revealed the activation of a stress-induced transcription factor ATF4 following LAT1 inhibition, aligning with the known cellular responses to amino acid deprivation. This activation was linked to the GCN2-eIF2α pathway which regulates translation initiation. Our results show that ATF4 upregulation counteracts the suppressive effect of nanvuranlat on cell proliferation in pancreatic ductal adenocarcinoma cell lines, suggesting a role for ATF4 in cellular adaptation to LAT1 inhibition. Importantly, dual targeting of LAT1 and ATF4 exhibited more substantial anti-proliferative effects in vitro than individual treatments. This study underscores the potential of combining LAT1 and ATF4 inhibition as a therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Yu Ma
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Suguru Okuda
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroki Okanishi
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Minhui Xu
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Chunhuan Jin
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hitoshi Endou
- J-Pharma Co., Ltd., Yokohama, Kanagawa, 230-0046, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
41
|
Chang Y, Chen J, Zhu H, Huang R, Wu J, Lin Y, Li Q, Shen G, Feng J. Metabolic Characteristics and Discriminative Diagnosis of Growth Hormone Deficiency and Idiopathic Short Stature in Preadolescents and Adolescents. Molecules 2024; 29:1661. [PMID: 38611940 PMCID: PMC11013616 DOI: 10.3390/molecules29071661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
Growth hormone deficiency (GHD) and idiopathic short stature (ISS) are the most common types of short stature (SS), but little is known about their pathogenesis, and even less is known about the study of adolescent SS. In this study, nuclear magnetic resonance (NMR)-based metabolomic analysis combined with least absolute shrinkage and selection operator (LASSO) were performed to identify the biomarkers of different types of SS (including 94 preadolescent GHD (PAG), 61 preadolescent ISS (PAI), 43 adolescent GHD (ADG), and 19 adolescent ISS (ADI)), and the receiver operating characteristic curve (ROC) was further used to evaluate the predictive power of potential biomarkers. The results showed that fourteen, eleven, nine, and fifteen metabolites were identified as the potential biomarkers of PAG, PAI, ADG, and ADI compared with their corresponding controls, respectively. The disturbed metabolic pathways in preadolescent SS were mainly carbohydrate metabolism and lipid metabolism, while disorders of amino acid metabolism played an important role in adolescent SS. The combination of aspartate, ethanolamine, phosphocholine, and trimethylamine was screened out to identify PAI from PAG, and alanine, histidine, isobutyrate, methanol, and phosphocholine gave a high classification accuracy for ADI and ADC. The differences in metabolic characteristics between GHD and ISS in preadolescents and adolescents will contribute to the development of individualized clinical treatments in short stature.
Collapse
Affiliation(s)
- Yajie Chang
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China; (Y.C.); (J.W.); (Q.L.)
| | - Jing Chen
- Department of Child Health, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361003, China; (J.C.); (R.H.); (Y.L.)
| | - Hongwei Zhu
- Education Section and Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China;
| | - Rong Huang
- Department of Child Health, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361003, China; (J.C.); (R.H.); (Y.L.)
| | - Jinxia Wu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China; (Y.C.); (J.W.); (Q.L.)
| | - Yanyan Lin
- Department of Child Health, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361003, China; (J.C.); (R.H.); (Y.L.)
| | - Quanquan Li
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China; (Y.C.); (J.W.); (Q.L.)
| | - Guiping Shen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China; (Y.C.); (J.W.); (Q.L.)
| | - Jianghua Feng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen 361005, China; (Y.C.); (J.W.); (Q.L.)
| |
Collapse
|
42
|
Wang Y, Lu Y, Fang Z, Yan H, Li J, Ye Z, Yu Y, Shan W. Brisk walking improves motor function and lower limb muscle strength in Chinese women aged 80 years and older. Sci Rep 2024; 14:7933. [PMID: 38575643 PMCID: PMC10995214 DOI: 10.1038/s41598-024-55925-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/29/2024] [Indexed: 04/06/2024] Open
Abstract
This study investigates the effects of a 12-week brisk walking exercise regimen on motor function improvements in elderly women. Twenty-six elderly women, aged 84.2 ± 3.2 years, participated in a 12-week brisk walking exercise program. Fitness assessments and blood biomarker analyses (including CHO, HDLC, LDLC, TC) were conducted pre- and post-intervention. Additionally, targeted metabolomics was employed to measure short-chain fatty acids, amino acids, and vitamin metabolites. The intervention led to significant enhancements in participants' flexibility (p < 0.05), lower limb muscle strength (p < 0.01), and cardiorespiratory endurance (p < 0.01), while muscle mass showed no significant changes. Fifteen significant differential metabolites were identified (VIP > 1.0, FC > 1.2 or < 0.8, and p < 0.05), with arginine, ornithine, aspartic acid, glutamine, phenylalanine, tyrosine, and pantothenic acid playing key roles across seven metabolic pathways. A 12-week brisk walking exercise program significantly enhanced flexibility, lower limb muscle strength, and cardiorespiratory endurance among elderly women. These improvements did not extend to muscle mass or upper limb muscle strength. The observed enhancement in exercise capacity may be attributed to improved regulation of neurotransmitters.
Collapse
Affiliation(s)
- Yang Wang
- The School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University, Beijing, 100084, China
| | - Yifan Lu
- The School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University, Beijing, 100084, China.
| | - Zilong Fang
- The School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University, Beijing, 100084, China
| | - Huiping Yan
- The School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University, Beijing, 100084, China
| | - Jiahao Li
- The School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University, Beijing, 100084, China
| | - Zhifan Ye
- The School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University, Beijing, 100084, China
| | - Yichao Yu
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University, Beijing, 100084, China
- The School of Sports Coaching, Beijing Sport University, Beijing, 100084, China
| | - Wei Shan
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University, Beijing, 100084, China.
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
43
|
Wang X, Cornish AE, Do MH, Brunner JS, Hsu TW, Xu Z, Malik I, Edwards C, Capistrano KJ, Zhang X, Ginsberg MH, Finley LWS, Lim MS, Horwitz SM, Li MO. Onco-Circuit Addiction and Onco-Nutrient mTORC1 Signaling Vulnerability in a Model of Aggressive T Cell Malignancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587917. [PMID: 38617314 PMCID: PMC11014592 DOI: 10.1101/2024.04.03.587917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
How genetic lesions drive cell transformation and whether they can be circumvented without compromising function of non-transformed cells are enduring questions in oncology. Here we show that in mature T cells-in which physiologic clonal proliferation is a cardinal feature- constitutive MYC transcription and Tsc1 loss in mice modeled aggressive human malignancy by reinforcing each other's oncogenic programs. This cooperation was supported by MYC-induced large neutral amino acid transporter chaperone SLC3A2 and dietary leucine, which in synergy with Tsc1 deletion overstimulated mTORC1 to promote mitochondrial fitness and MYC protein overexpression in a positive feedback circuit. A low leucine diet was therapeutic even in late-stage disease but did not hinder T cell immunity to infectious challenge, nor impede T cell transformation driven by constitutive nutrient mTORC1 signaling via Depdc5 loss. Thus, mTORC1 signaling hypersensitivity to leucine as an onco-nutrient enables an onco-circuit, decoupling pathologic from physiologic utilization of nutrient acquisition pathways.
Collapse
|
44
|
Liu X, Nishikubo K, Ohgaki R, Okanishi H, Okuda S, Xu M, Kanai Y. Identification of tumor-suppressive miRNAs that target amino acid transporter LAT1 and exhibit anti-proliferative effects on cholangiocarcinoma cells. J Pharmacol Sci 2024; 154:301-311. [PMID: 38485348 DOI: 10.1016/j.jphs.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/24/2024] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
Amino acid transporter LAT1 is highly upregulated in various cancer types, including cholangiocarcinoma (CHOL), and contributes to the rapid proliferation of cancer cells and disease progression. However, the molecular mechanisms underlying the pathological upregulation of LAT1 remain largely unknown. This study pursued the possibility of miRNA-mediated regulation of the LAT1 expression in CHOL cells. Using online target prediction methods, we extracted five candidate miRNAs commonly predicted to regulate the LAT1 expression. Three of them, miR-194-5p, miR-122-5p, and miR-126-3p, were significantly downregulated in CHOL cancer compared to normal tissues. Correlation analysis revealed weak-to-moderate negative correlations between the expression of these miRNAs and LAT1 mRNA in CHOL cancer tissues. We selected miR-194-5p and miR-122-5p for further analyses and found that both miRNAs functionally target 3'UTR of LAT1 mRNA by a luciferase-based reporter assay. Transfection of the miRNA mimics significantly suppressed the LAT1 expression at mRNA and protein levels and inhibited the proliferation of CHOL cells, with a trend of affecting intracellular amino acids and amino acid-related signaling pathways. This study indicates that the decreased expression of these LAT1-targeting tumor-suppressive miRNAs contributes to the upregulation of LAT1 and the proliferation of CHOL cells, highlighting their potential for developing novel cancer therapeutics and diagnostics.
Collapse
Affiliation(s)
- Xingming Liu
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kou Nishikubo
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hiroki Okanishi
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Suguru Okuda
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Minhui Xu
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
45
|
Wu X, Fang Y, Gu Y, Shen H, Xu Y, Xu T, Shi R, Xu D, Zhang J, Leng K, Shu Y, Ma P. Fat mass and obesity-associated protein (FTO) mediated m 6A modification of circFAM192A promoted gastric cancer proliferation by suppressing SLC7A5 decay. MOLECULAR BIOMEDICINE 2024; 5:11. [PMID: 38556586 PMCID: PMC10982225 DOI: 10.1186/s43556-024-00172-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/21/2024] [Indexed: 04/02/2024] Open
Abstract
Gastric cancer (GC) is a common malignant tumor worldwide, especially in East Asia, with high incidence and mortality rate. Epigenetic modifications have been reported to participate in the progression of gastric cancer, among which m6A is the most abundant and important chemical modification in RNAs. Fat mass and obesity-associated protein (FTO) is the first identified RNA demethylase but little is known about its role in gastric cancer. In our study, data from TCGA and clinical samples showed that FTO was highly expressed in gastric cancer tissues. Kaplan-Meier plotter suggested that patients with the high level of FTO had a poor prognosis. In vitro and in vivo experiments confirmed the role of FTO in promoting gastric cancer cell proliferation. Mechanistically, we found that FTO bound to circFAM192A at the specific site and removed the m6A modification in circFAM192A, protecting it from degradation. CircFAM192A subsequently interacted with the leucine transporter solute carrier family 7 member 5 (SLC7A5) and enhancing its stability. As a result, an increased amount of SLC7A5 was on the membrane, which facilitated leucine uptake and activated the mTOR signaling pathway. Therefore, our study demonstrated that FTO promoted gastric cancer proliferation through the circFAM192A/SLC7A5 axis in the m6A-dependent manner. Our study shed new light on the role of FTO in gastric cancer progression.
Collapse
Affiliation(s)
- Xi Wu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Yuan Fang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Yunru Gu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Haoyang Shen
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Yangyue Xu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Tingting Xu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Run Shi
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Duo Xu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Jingxin Zhang
- Department of General Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang Clinic School of Nanjing Medical University, Zhenjiang, People's Republic of China
| | - Kai Leng
- Department of Medical Informatics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Yongqian Shu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China.
| | - Pei Ma
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
46
|
Zhu K, Liu C, Guo X, Zhang X, Xie J, Xie S, Qi Q, Yang B. Exosomal miR-126-3p: Potential protection against vascular damage by regulating the SLC7A5/mTOR Signalling pathway in human umbilical vein endothelial cells. Scand J Immunol 2024; 99:e13354. [PMID: 39008522 DOI: 10.1111/sji.13354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/19/2023] [Accepted: 12/27/2023] [Indexed: 07/17/2024]
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune connective tissue disease. Vascular damage is one of the important features of SSc, which affects the progression and prognosis of the disease. MiR-126-3p is an important microRNA (miRNA) that regulates vascular structure and function, which can be transported through exosomes. However, the role of miR-126-3p in vascular damage in SSc is still unclear. Therefore, we focused on the connection between miR-126-3p and vascular damage in SSc, as well as investigated the potential role of miR-126-3p in vascular damage in SSc. First, this study successfully extracted extracellular vesicles from clinical plasma samples and characterized the exosomes within them. Then, we predicted and screened the target pathway mammalian/mechanistic target of rapamycin (mTOR) and the target gene SLC7A5 of miR-126-3p through online databases. Next, we constructed SSc mice for in vivo studies. The results showed that the expression of miR-126-3p was decreased in the plasma exosomes, while the SLC7A5 expression, autophagy, and lipid peroxidation were increased in the aorta. Luciferase reporter gene assays demonstrated that miR-126-3p can bind to SLC7A5, resulting in a decrease in its expression. In vitro experiments have shown that exosomal miR-126-3p can be internalized by human umbilical vein endothelial cells (HUVECs). The miR-126-3p group exhibited enhanced cell viability and tube formation ability, along with increased expression of the vascular formation marker CD31. Additionally, miR-126-3p downregulated the protein expression of SLC7A5 and LC3 in HUVECs, while upregulating the protein expression of mTOR, P62, PPARγ, and CPT-1. However, the effects of miR-126-3p on HUVECs were counteracted by mTOR inhibitors and enhanced by mTOR activators. The results indicated that exosomal miR-126-3p has the potential to protect against vascular injury in SSc by regulating the SLC7A5/mTOR signalling pathway in HUVECs.
Collapse
Affiliation(s)
- Ke Zhu
- Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Dermatology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chen Liu
- Department of Dermatology, Shenzhen People's Hospital, Shenzhen, China
| | - Xiaofang Guo
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuting Zhang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaxin Xie
- Department of Dermatology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Songmiao Xie
- Department of Dermatology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qing Qi
- Department of Dermatology, The Second Hospital Affiliated to Guangzhou Medical University, Guangzhou, China
| | - Bin Yang
- Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
47
|
Bao L, Fu L, Su Y, Chen Z, Peng Z, Sun L, Gonzalez FJ, Wu C, Zhang H, Shi B, Shi YB. Amino acid transporter SLC7A5 regulates cell proliferation and secretary cell differentiation and distribution in the mouse intestine. Int J Biol Sci 2024; 20:2187-2201. [PMID: 38617535 PMCID: PMC11008275 DOI: 10.7150/ijbs.94297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/16/2024] [Indexed: 04/16/2024] Open
Abstract
The intestine is critical for not only processing nutrients but also protecting the organism from the environment. These functions are mainly carried out by the epithelium, which is constantly being self-renewed. Many genes and pathways can influence intestinal epithelial cell proliferation. Among them is mTORC1, whose activation increases cell proliferation. Here, we report the first intestinal epithelial cell (IEC)-specific knockout (ΔIEC) of an amino acid transporter capable of activating mTORC1. We show that the transporter, SLC7A5, is highly expressed in mouse intestinal crypt and Slc7a5ΔIEC reduces mTORC1 signaling. Surprisingly, adult Slc7a5ΔIEC intestinal crypts have increased cell proliferation but reduced mature Paneth cells. Goblet cells, the other major secretory cell type in the small intestine, are increased in the crypts but reduced in the villi. Analyses with scRNA-seq and electron microscopy have revealed dedifferentiation of Paneth cells in Slc7a5ΔIEC mice, leading to markedly reduced secretory granules with little effect on Paneth cell number. Thus, SLC7A5 likely regulates secretory cell differentiation to affect stem cell niche and indirectly regulate cell proliferation.
Collapse
Affiliation(s)
- Lingyu Bao
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine. No.277, Yanta West Road, Xi'an, Shaanxi, 710061, P.R. China
| | - Liezhen Fu
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
| | - Yijun Su
- Laboratory of High Resolution Optical Imaging and Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhaoyi Peng
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine. No.277, Yanta West Road, Xi'an, Shaanxi, 710061, P.R. China
| | - Lulu Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hongen Zhang
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine. No.277, Yanta West Road, Xi'an, Shaanxi, 710061, P.R. China
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
| |
Collapse
|
48
|
Wang B, Pei J, Xu S, Liu J, Yu J. A glutamine tug-of-war between cancer and immune cells: recent advances in unraveling the ongoing battle. J Exp Clin Cancer Res 2024; 43:74. [PMID: 38459595 PMCID: PMC10921613 DOI: 10.1186/s13046-024-02994-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/22/2024] [Indexed: 03/10/2024] Open
Abstract
Glutamine metabolism plays a pivotal role in cancer progression, immune cell function, and the modulation of the tumor microenvironment. Dysregulated glutamine metabolism has been implicated in cancer development and immune responses, supported by mounting evidence. Cancer cells heavily rely on glutamine as a critical nutrient for survival and proliferation, while immune cells require glutamine for activation and proliferation during immune reactions. This metabolic competition creates a dynamic tug-of-war between cancer and immune cells. Targeting glutamine transporters and downstream enzymes involved in glutamine metabolism holds significant promise in enhancing anti-tumor immunity. A comprehensive understanding of the intricate molecular mechanisms underlying this interplay is crucial for developing innovative therapeutic approaches that improve anti-tumor immunity and patient outcomes. In this review, we provide a comprehensive overview of recent advances in unraveling the tug-of-war of glutamine metabolism between cancer and immune cells and explore potential applications of basic science discoveries in the clinical setting. Further investigations into the regulation of glutamine metabolism in cancer and immune cells are expected to yield valuable insights, paving the way for future therapeutic interventions.
Collapse
Affiliation(s)
- Bolin Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinli Pei
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Shengnan Xu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China
| | - Jie Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
49
|
Yuan Q, Yin L, He J, Zeng Q, Liang Y, Shen Y, Zu X. Metabolism of asparagine in the physiological state and cancer. Cell Commun Signal 2024; 22:163. [PMID: 38448969 PMCID: PMC10916255 DOI: 10.1186/s12964-024-01540-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/22/2024] [Indexed: 03/08/2024] Open
Abstract
Asparagine, an important amino acid in mammals, is produced in several organs and is widely used for the production of other nutrients such as glucose, proteins, lipids, and nucleotides. Asparagine has also been reported to play a vital role in the development of cancer cells. Although several types of cancer cells can synthesise asparagine alone, their synthesis levels are insufficient to meet their requirements. These cells must rely on the supply of exogenous asparagine, which is why asparagine is considered a semi-essential amino acid. Therefore, nutritional inhibition by targeting asparagine is often considered as an anti-cancer strategy and has shown success in the treatment of leukaemia. However, asparagine limitation alone does not achieve an ideal therapeutic effect because of stress responses that upregulate asparagine synthase (ASNS) to meet the requirements for asparagine in cancer cells. Various cancer cells initiate different reprogramming processes in response to the deficiency of asparagine. Therefore, it is necessary to comprehensively understand the asparagine metabolism in cancers. This review primarily discusses the physiological role of asparagine and the current progress in the field of cancer research.
Collapse
Affiliation(s)
- Qiong Yuan
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Liyang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Qiting Zeng
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yuxin Liang
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
50
|
Huang TQ, Chen YX, Zeng SL, Lin Y, Li F, Jiang ZM, Liu EH. Bergenin Alleviates Ulcerative Colitis By Decreasing Gut Commensal Bacteroides vulgatus-Mediated Elevated Branched-Chain Amino Acids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3606-3621. [PMID: 38324392 DOI: 10.1021/acs.jafc.3c09448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Ulcerative colitis is closely associated with the dysregulation of gut microbiota. There is growing evidence that natural products may improve ulcerative colitis by regulating the gut microbiota. In this research, we demonstrated that bergenin, a naturally occurring isocoumarin, significantly ameliorates colitis symptoms in dextran sulfate sodium (DSS)-induced mice. Transcriptomic analysis and Caco-2 cell assays revealed that bergenin could ameliorate ulcerative colitis by inhibiting TLR4 and regulating NF-κB and mTOR phosphorylation. 16S rRNA sequencing and metabolomics analyses revealed that bergenin could improve gut microbiota dysbiosis by decreasing branched-chain amino acid (BCAA) levels. BCAA intervention mediated the mTOR/p70S6K signaling pathway to exacerbate the symptoms of ulcerative colitis in mice. Notably, bergenin greatly decreased the symbiotic bacteria Bacteroides vulgatus (B. vulgatus), and the gavage of B. vulgatus increased BCAA concentrations and aggravated the symptoms of ulcerative colitis in mice. Our findings suggest that gut microbiota-mediated BCAA metabolism plays a vital role in the protective effect of bergenin on ulcerative colitis, providing novel insights for ulcerative colitis prevention through manipulation of the gut microbiota.
Collapse
Affiliation(s)
- Tian-Qing Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Yu-Xin Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Su-Ling Zeng
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| | - Yang Lin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Fei Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zheng-Meng Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
- College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Road, Nanjing 210023, China
| | - E-Hu Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
- College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Road, Nanjing 210023, China
| |
Collapse
|