1
|
Li J, Peng Y, Zhen D, Guo C, Peng W. NEDD4 enhances bone‑tendon healing in rotator cuff tears by reducing fatty infiltration. Mol Med Rep 2025; 31:55. [PMID: 39704218 DOI: 10.3892/mmr.2024.13420] [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: 07/18/2024] [Accepted: 11/14/2024] [Indexed: 12/21/2024] Open
Abstract
Rotator cuff tears (RCT) can cause shoulder pain, weakness and stiffness, significantly affecting daily life. Analysis of the GSE103266 dataset revealed significant changes in the mTOR/PI3K/Akt signaling pathway and lipid metabolism‑related pathways, suggesting that fatty infiltration may affect RCT. The analysis indicated that the ubiquitin ligase NEDD4 plays a critical role in RCT. NEDD4 was found to be highly associated with the mTOR/PI3K/Akt signaling pathway. An RCT model in Sprague‑Dawley (SD) rats was established to study the role of NEDD4 in regulating the mTOR pathway and investigate its effects on fatty infiltration. SD rats were divided into NEDD4 overexpression and knockout groups. Tissue recovery, apoptosis and fat deposition were measured through histological staining, reverse transcription‑quantitative PCR and western blotting. Additionally, cell culture of fibro‑adipogenic progenitors and lentiviral transfection were conducted to investigate the effect of NEDD4 on adipocyte differentiation. NEDD4 overexpression significantly reduced lipid accumulation, whereas NEDD4 knockdown enhanced lipid accumulation. NEDD4 was found to regulate the mTOR pathway and the expression of adipogenesis‑related genes, promoting fat metabolism and inhibiting adipocyte differentiation. Histological analysis indicated that NEDD4 overexpression improved tissue recovery and reduced apoptosis. Targeting NEDD4 offers a potential therapeutic strategy to improve the clinical outcomes of patients with RCT by modulating the mTOR pathway and fat metabolism.
Collapse
Affiliation(s)
- Jian Li
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Ying Peng
- Thyroid Disease Diagnosis and Treatment Center, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Dong Zhen
- Department of Sports Medicine, The Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, Guizhou 550014, P.R. China
| | - Caifen Guo
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Wuxun Peng
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
2
|
Lim YN, Ryu IS, Jung YJ, Helmlinger G, Kim I, Park HW, Kang H, Lee J, Lee HJ, Lee KS, Jang HN, Ha DI, Park J, Won J, Lim KS, Jeon CY, Cho HJ, Min HS, Ryu JH. l-Type amino acid transporter 1-targeting nanoparticles for antisense oligonucleotide delivery to the CNS. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102340. [PMID: 39411247 PMCID: PMC11474373 DOI: 10.1016/j.omtn.2024.102340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024]
Abstract
l-Type amino acid transporter 1 (LAT1)-specific ligands and polyion complexes are used as brain-specific targets to deliver RNA-based drugs across the blood-brain barrier. We characterized an LAT1-targeting antisense oligonucleotide (ASO)-encapsulated nanoparticle, Phe-NPs/ASO. A 25% density of phenylalanine effectively binds to the surface of LAT1-targeting NPs in the GL261-Luc cells, and Phe-NPs/ASO shows higher binding affinity compared to that without phenylalanine by cellular binding assay. To further characterize the blood-brain barrier-targeting effect and tissue distribution following a single-dose intravenous injection in mice, we performed in vivo biodistribution studies using fluorescence imaging. The Phe-NPs/ASOs were detected in the brain tissue 1 h post-intravenous injection at an approximately 64-fold higher ratio than that of the same ASOs administered in the absence of any NP carrier. The brain tissue delivery of ASO-loaded Phe-NPs was also confirmed in a fluorescence imaging study performed in non-human primates. These results demonstrate that Phe-NPs may successfully deliver an ASO to the brain tissue across brain regions. Phe-NPs loaded with RNA-based drugs have the potential to treat diseases of the CNS, including all forms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu Na Lim
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - In Soo Ryu
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Yeon-Joo Jung
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Gabriel Helmlinger
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Insun Kim
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Hye Won Park
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Hansol Kang
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Jina Lee
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Hyo Jin Lee
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Kang Seon Lee
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Ha-Na Jang
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Dae-In Ha
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Junghyung Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, South Korea
| | - Jinyoung Won
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, South Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, South Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, South Korea
| | - Hyun-Jeong Cho
- Department of Biomedical Laboratory Science, College of Medical Science, Konyang University, Daejeon 35365, South Korea
| | - Hyun Su Min
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| | - Jin-Hyeob Ryu
- BIORCHESTRA Co., Ltd, 1, Gukjegwahak 2-ro, Yuseong-gu, Daejeon 34000, South Korea
| |
Collapse
|
3
|
Zhang X, Teng J, Chen Z, Zhao C, Jiang L, Zhang Q. S100A9 Affects Milk Protein Content by Regulating Amino Acid Transporters and the PI3K-Akt, WNT, and mTOR Signaling Pathways. Genes (Basel) 2024; 15:1486. [PMID: 39596686 PMCID: PMC11594203 DOI: 10.3390/genes15111486] [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: 09/23/2024] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Calgranulin B (S100A9) was found to be strongly associated with milk protein percentage in dairy cattle in our previous genome-wide association study. METHODS SNPs in S100A9 were identified via pooled sequencing, and genotyping of 1054 cows was performed individually using MassArray with MALDI-TOFMS technology. Association analyses between the S100A9 SNPs and five milk production traits were conducted using SAS 9.2 software. Functional studies of S100A9 were conducted using quantitative PCR, Western blot, CCK-8, and immunofluorescence assays. RESULTS In the present study, we further verified that two SNPs in S100A9, g.17115387 C>A and g.17115176 C>A, were significantly associated with milk protein percentage. We found that S100A9 could affect the expressions of caseins CSN1S1, CSN2, and CSN3 in MAC-T cells by regulating the expressions of amino acid transporter genes. We investigated the effects of S100A9 on the PI3K-Akt, WNT, and mTOR pathways, which are well known to play important roles in mammary gland development and milk protein synthesis. Our results suggest that S100A9 regulates the expressions of the relevant genes in these pathways, and thus potentially influences the protein synthesis in the mammary gland. CONCLUSIONS This study demonstrates the important role of the S100A9 gene in the milk protein trait of dairy cattle and provides new insights into the molecular mechanism of milk protein content.
Collapse
Affiliation(s)
- Xinyi Zhang
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science, Shandong Agricultural University, Tai’an 271018, China; (X.Z.); (J.T.); (Z.C.); (C.Z.)
| | - Jun Teng
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science, Shandong Agricultural University, Tai’an 271018, China; (X.Z.); (J.T.); (Z.C.); (C.Z.)
| | - Zhujun Chen
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science, Shandong Agricultural University, Tai’an 271018, China; (X.Z.); (J.T.); (Z.C.); (C.Z.)
| | - Changheng Zhao
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science, Shandong Agricultural University, Tai’an 271018, China; (X.Z.); (J.T.); (Z.C.); (C.Z.)
| | - Li Jiang
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China
| | - Qin Zhang
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science, Shandong Agricultural University, Tai’an 271018, China; (X.Z.); (J.T.); (Z.C.); (C.Z.)
| |
Collapse
|
4
|
Liu Y, Li J, Ding C, Tong H, Yan Y, Li S, Li S, Cao Y. Leu promotes C2C12 cell differentiation by regulating the GSK3β/β-catenin signaling pathway through facilitating the interaction between SESN2 and RPN2. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:6696-6705. [PMID: 38551359 DOI: 10.1002/jsfa.13496] [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: 04/06/2023] [Revised: 03/02/2024] [Accepted: 03/29/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Leucine (Leu) is an essential amino acid that facilitates skeletal muscle satellite cell differentiation, yet its mechanism remains underexplored. Sestrin2 (SESN2) serves as a Leu sensor, binding directly to Leu, while ribophorin II (RPN2) acts as a signaling factor in multiple pathways. This study aimed to elucidate Leu's impact on mouse C2C12 cell differentiation and skeletal muscle injury repair by modulating RPN2 expression through SESN2, offering a theoretical foundation for clinical skeletal muscle injury prevention and treatment. RESULTS Leu addition promoted C2C12 cell differentiation compared to the control, enhancing early differentiation via myogenic determinant (MYOD) up-regulation. Sequencing revealed SESN2 binding to and interacting with RPN2. RPN2 overexpression up-regulated MYOD, myogenin and myosin heavy chain 2, concurrently decreased p-GSK3β and increased nuclear β-catenin. Conversely, RPN2 knockdown yielded opposite results. Combining RPN2 knockdown with Leu rescued increased p-GSK3β and decreased nuclear β-catenin compared to Leu absence. Hematoxylin and eosin staining results showed that Leu addition accelerated mouse muscle damage repair, up-regulating Pax7, MYOD and RPN2 in the cytoplasm, and nuclear β-catenin, confirming that the role of Leu in muscle injury repair was consistent with the results for C2C12 cells. CONCLUSION Leu, bound with SESN2, up-regulated RPN2 expression, activated the GSK3β/β-catenin pathway, enhanced C2C12 differentiation and expedited skeletal muscle damage repair. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yifan Liu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Jinping Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Cong Ding
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Huili Tong
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Yunqin Yan
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Shuang Li
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Shufeng Li
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Yunkao Cao
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
5
|
Mora S, Adegoke OAJ. Maintenance of the branched-chain amino acid transporter LAT1 counteracts myotube atrophy following chemotherapy. Am J Physiol Cell Physiol 2024; 326:C866-C879. [PMID: 38284122 DOI: 10.1152/ajpcell.00537.2023] [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: 10/13/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/30/2024]
Abstract
Prevention/management of cachexia remains a critical issue in muscle wasting conditions. The branched-chain amino acids (BCAA) have anabolic properties in skeletal muscle, but their use in treating cachexia has minimal benefits. This may be related to altered BCAA metabolism consequent to the use of chemotherapy, a main cancer treatment. Since this topic is minimally studied, we investigated the effect of chemotherapy on BCAA concentrations, transporter expression, and their metabolism. L6 myotubes were treated with vehicle (1.4 μL/mL DMSO) or a chemotherapy drug cocktail, FOLFIRI [CPT-11 (20 μg/mL), leucovorin (10 μg/mL), and 5-fluorouracil (50 μg/mL)] for 24-48 h. Chemotherapy reduced myotube diameter (-43%), myofibrillar protein content (-50%), and phosphorylation of the mechanistic target of rapamycin complex 1 (mTORC1) substrate S6K1thr389 (-80%). Drug-treated myotubes exhibited decreased BCAA concentrations (-52%) and expression of their transporter, L-type amino acid transporter 1 (LAT1; -67%). BCAA transaminase BCAT2 level was increased, but there was a reduction in PP2CM (-54%), along with increased inhibitory phosphorylation of BCKD-E1αser293 (+98%), corresponding with decreased BCKD enzyme activity (-23%) in chemotherapy-treated myotubes. Decreases in BCAA concentrations were a later response, preceded by decreases in LAT1 and BCKD activity. Although supplementation with the BCAA restored myotube BCAA levels, it had minimal effects on preventing the loss of myofibrillar proteins. However, RNAi-mediated depletion of neural precursor cell-expressed developmentally downregulated gene 4 (NEdd4), the protein ligase responsible for ubiquitin-dependent degradation of LAT1, attenuated the effects of chemotherapy on BCAA concentrations, anabolic signaling, protein synthesis, and myofibrillar protein abundance. Thus, if our findings are validated in preclinical models, interventions regulating muscle amino acid transporters might represent a promising strategy to treat cachexia.NEW & NOTEWORTHY This is the first study to attenuate chemotherapy-induced myotube atrophy by manipulating a BCAA transporter. Our findings suggest that positive regulation of amino acid transporters may be a promising strategy to treat cachexia.
Collapse
Affiliation(s)
- Stephen Mora
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Olasunkanmi A J Adegoke
- School of Kinesiology and Health Science and Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Koe JC, Parker SJ. The posttranslational regulation of amino acid transporters is critical for their function in the tumor microenvironment. Curr Opin Biotechnol 2024; 85:103022. [PMID: 38056204 DOI: 10.1016/j.copbio.2023.103022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
Amino acid transporters (AATs) facilitate nutrient uptake and nutrient exchange between cancer and stromal cells. The posttranslational modification (PTM) of transporters is an important mechanism that tumor-associated cells use to dynamically regulate their function and stability in response to microenvironmental cues. In this review, we summarize recent findings that demonstrate the significance of N-glycosylation, phosphorylation, and ubiquitylation for the function of AATs. We also highlight powerful approaches that hijack the PTM machinery that could be used as therapeutics or tools to modulate transporter activity.
Collapse
Affiliation(s)
- Jessica C Koe
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada
| | - Seth J Parker
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada; British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
7
|
Tae K, Kim SJ, Cho SW, Lee H, Cha HS, Choi CY. L-Type Amino Acid Transporter 1 (LAT1) Promotes PMA-Induced Cell Migration through mTORC2 Activation at the Lysosome. Cells 2023; 12:2504. [PMID: 37887348 PMCID: PMC10605051 DOI: 10.3390/cells12202504] [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: 08/28/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 10/28/2023] Open
Abstract
The mTOR signaling pathway integrates signaling inputs from nutrients, including glucose and amino acids, which are precisely regulated by transporters depending on nutrient levels. The L-type amino acid transporter 1 (LAT1) affects the activity of mTORC1 through upstream regulators that sense intracellular amino acid levels. While mTORC1 activation by LAT1 has been thoroughly investigated in cultured cells, the effects of LAT1 expression on the activity of mTORC2 has scarcely been studied. Here, we provide evidence that LAT1 recruits and activates mTORC2 on the lysosome for PMA-induced cell migration. LAT1 is translocated to the lysosomes in cells treated with PMA in a dose- and time-dependent manner. Lysosomal LAT1 interacted with mTORC2 through a direct interaction with Rictor, leading to the lysosomal localization of mTORC2. Furthermore, the depletion of LAT1 reduced PMA-induced cell migration in a wound-healing assay. Consistent with these results, the LAT1 N3KR mutant, which is defective in PMA-induced endocytosis and lysosomal localization, did not induce mTORC2 recruitment to the lysosome, with the activation of mTORC2 determined via Akt phosphorylation or the LAT1-mediated promotion of cell migration. Taken together, lysosomal LAT1 recruits and activates the mTORC2 complex and downstream Akt for PMA-mediated cell migration. These results provide insights into the development of therapeutic drugs targeting the LAT1 amino acid transporter to block metastasis, as well as disease progression in various types of cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Cheol-Yong Choi
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; (K.T.); (S.-J.K.); (S.-W.C.); (H.L.); (H.-S.C.)
| |
Collapse
|
8
|
Wei S, Wang W, Liu S, Sun B, Zeng Q, Wang G, Luo P, Zhang A. Genome-wide DNA methylation pattern in whole blood of patients with coal-burning arsenic poisoning. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 248:114323. [PMID: 36436256 DOI: 10.1016/j.ecoenv.2022.114323] [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: 05/26/2022] [Revised: 08/31/2022] [Accepted: 11/20/2022] [Indexed: 06/16/2023]
Abstract
Exposure to coal-burning arsenic leads to an increased risk of cancer, multi-systems damage and chronic diseases, with DNA methylation one potential mechanism of arsenic toxicity. There are few studies on genome-wide methylation in the coal-burning arsenic poisoning population. Illumina 850 K methylation beadchip is the most suitable technology for DNA methylation of epigenome-wide association analysis. This study used 850 K to detect changes in Genome-wide DNA methylation in whole blood samples of 12 patients with coal-burning arsenic poisoning ( Arsenic poisoning group) and four healthy control participants (Healthy control group). There is clearly abnormal genome-wide DNA methylation in coal-burning arsenic poisoning, with 647 significantly different methylation positions, 524 different methylation regions and 335 significantly different methylation genes in arsenic poisoning patients compared with healthy controls. Further functional analysis of Gene ontology (GO) and Kyoto encyclopedia of genes (KEGG) found 592 GO items and 131 KEGG pathways between patients of coal-burning arsenic poisoning and healthy control. Then, analysis of gene degree and combined-score identified NAPRT1, NT5C3B, NEDD4L, SLC22A3 and RAB11B as target genes. Further validation by qRT-PCR indicates that mRNA expression of five genes changes significantly in the arsenic poisoning group (n = 72) compared to the healthy control group (n = 72). These results showed the genome-wide methylation pattern and highlighted five critical genes within the coal-burning arsenic poisoning population that involve Nicotinate and nicotinamide metabolism, Choline metabolism in cancer, and Ubiquitin mediated proteolysis. Next, the methylation profile of coal burning arsenic poisoning will be further excavation and the mechanism of coal burning arsenic poisoning will be further explored from five genes related pathways and functions.
Collapse
Affiliation(s)
- Shaofeng Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China; The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China.
| | - Wenjing Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China; The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Shiwen Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China; The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Baofei Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China; The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Qibing Zeng
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China; The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Guoze Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China; The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Peng Luo
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China; The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Aihua Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China; The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China.
| |
Collapse
|
9
|
Targeting Transporters for Drug Delivery to the Brain: Can We Do Better? Pharm Res 2022; 39:1415-1455. [PMID: 35359241 PMCID: PMC9246765 DOI: 10.1007/s11095-022-03241-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/21/2022] [Indexed: 12/11/2022]
Abstract
Limited drug delivery to the brain is one of the major reasons for high failure rates of central nervous system (CNS) drug candidates. The blood–brain barrier (BBB) with its tight junctions, membrane transporters, receptors and metabolizing enzymes is a main player in drug delivery to the brain, restricting the entrance of the drugs and other xenobiotics. Current knowledge about the uptake transporters expressed at the BBB and brain parenchymal cells has been used for delivery of CNS drugs to the brain via targeting transporters. Although many transporter-utilizing (pro)drugs and nanocarriers have been developed to improve the uptake of drugs to the brain, their success rate of translation from preclinical development to humans is negligible. In the present review, we provide a systematic summary of the current progress in development of transporter-utilizing (pro)drugs and nanocarriers for delivery of drugs to the brain. In addition, we applied CNS pharmacokinetic concepts for evaluation of the limitations and gaps in investigation of the developed transporter-utilizing (pro)drugs and nanocarriers. Finally, we give recommendations for a rational development of transporter-utilizing drug delivery systems targeting the brain based on CNS pharmacokinetic principles.
Collapse
|
10
|
Johnson M, Nowlan S, Sahin G, Barnett DA, Joy AP, Touaibia M, Cuperlovic-Culf M, Zofija Avizonis D, Turcotte S. Decrease of Intracellular Glutamine by STF-62247 Results in the Accumulation of Lipid Droplets in von Hippel-Lindau Deficient Cells. Front Oncol 2022; 12:841054. [PMID: 35223522 PMCID: PMC8865074 DOI: 10.3389/fonc.2022.841054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/13/2022] [Indexed: 01/01/2023] Open
Abstract
Kidney cancer is one of the top ten cancer diagnosed worldwide and its incidence has increased the last 20 years. Clear Cell Renal Cell Carcinoma (ccRCC) are characterized by mutations that inactivate the von Hippel-Lindau (VHL) tumor suppressor gene and evidence indicated alterations in metabolic pathways, particularly in glutamine metabolism. We previously identified a small molecule, STF-62247, which target VHL-deficient renal tumors by affecting late-stages of autophagy and lysosomal signaling. In this study, we investigated ccRCC metabolism in VHL-deficient and proficient cells exposed to the small molecule. Metabolomics profiling using 1H NMR demonstrated that STF-62247 increases levels of glucose, pyruvate, glycerol 3-phosphate while glutamate, asparagine, and glutathione significantly decreased. Diminution of glutamate and glutamine was further investigated using mass spectrometry, western blot analyses, enzymatic activities, and viability assays. We found that expression of SLC1A5 increases in VHL-deficient cells treated with STF-62247, possibly to stimulate glutamine uptake intracellularly to counteract the diminution of this amino acid. However, exogenous addition of glutamine was not able to rescue cell viability induced by the small molecule. Instead, our results showed that VHL-deficient cells utilize glutamine to produce fatty acid in response to STF-62247. Surprisingly, this occurs through oxidative phosphorylation in STF-treated cells while control cells use reductive carboxylation to sustain lipogenesis. We also demonstrated that STF-62247 stimulated expression of stearoyl-CoA desaturase (SCD1) and peripilin2 (PLIN2) to generate accumulation of lipid droplets in VHL-deficient cells. Moreover, the carnitine palmitoyltransferase 1A (CPT1A), which control the entry of fatty acid into mitochondria for β-oxidation, also increased in response to STF-62247. CPT1A overexpression in ccRCC is known to limit tumor growth. Together, our results demonstrated that STF-62247 modulates cellular metabolism of glutamine, an amino acid involved in the autophagy-lysosome process, to support lipogenesis, which could be implicated in the signaling driving to cell death.
Collapse
Affiliation(s)
- Mathieu Johnson
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada.,Atlantic Cancer Research Institute, Moncton, NB, Canada
| | - Sarah Nowlan
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada.,Atlantic Cancer Research Institute, Moncton, NB, Canada
| | - Gülsüm Sahin
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada.,Atlantic Cancer Research Institute, Moncton, NB, Canada
| | | | - Andrew P Joy
- Atlantic Cancer Research Institute, Moncton, NB, Canada
| | - Mohamed Touaibia
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
| | | | | | - Sandra Turcotte
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada.,Atlantic Cancer Research Institute, Moncton, NB, Canada
| |
Collapse
|
11
|
Huttunen J, Agami M, Tampio J, Montaser AB, Huttunen KM. Comparison of Experimental Strategies to Study l-Type Amino Acid Transporter 1 (LAT1) Utilization by Ligands. Molecules 2021; 27:molecules27010037. [PMID: 35011270 PMCID: PMC8746705 DOI: 10.3390/molecules27010037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 01/08/2023] Open
Abstract
l-Type amino acid transporter 1 (LAT1), expressed abundantly in the brain and placenta and overexpressed in several cancer cell types, has gained a lot of interest in drug research and development, as it can be utilized for brain-targeted drug delivery, as well as inhibiting the essential amino acid supply to cancer cells. The structure of LAT1 is today very well-known and the interactions of ligands at the binding site of LAT1 can be modeled and explained. However, less is known of LAT1′s life cycle within the cells. Moreover, the functionality of LAT1 can be measured by several different methods, which may vary between the laboratories and make the comparison of the results challenging. In the present study, the usefulness of indirect cis-inhibition methods and direct cellular uptake methods and their variations to interpret the interactions of LAT1-ligands were evaluated. Moreover, this study also highlights the importance of understanding the intracellular kinetics of LAT1-ligands, and how they can affect the regular function of LAT1 in critical tissues, such as the brain. Hence, it is discussed herein how the selected methodology influences the outcome and created knowledge of LAT1-utilizing compounds.
Collapse
|
12
|
Lee DY, Lin HY, Ramasamy M, Kuo SC, Lee PC, Hsieh MT. Synthesis and Characterization of the Ethylene-Carbonate-Linked L-Valine Derivatives of 4,4-Dimethylcurcumin with Potential Anticancer Activities. Molecules 2021; 26:molecules26227050. [PMID: 34834146 PMCID: PMC8624457 DOI: 10.3390/molecules26227050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/19/2022] Open
Abstract
Natural phenolic products from herbal medicines and dietary plants constitute the main source of lead compounds for the development of the new drug. 4,4-Dimethylcurcumin (DMCU) is a synthetic curcumin derivative and exhibits anticancer activities against breast, colon, lung, and liver cancers. However, further development of DMCU is limited by unfavorable compound properties such as very low aqueous solubility and moderate stability. To increase its solubility, we installed either or both of the ethylene-carbonate-linked L-valine side chains to DMCU phenolic groups and produced targeted 1-trifluoroacetic acid (1-TFA) and 2-trifluoroacetic acid (2-TFA) derivatives. The terminus L-valine of ethylene-carbonate-linked side chain is known to be a L-type amino acid transporter 1 (LAT1) recognition element and therefore, these two derivatives were expected to readily enter into LAT1-expressing cancer cells. In practice, 1-TFA or 2-TFA were synthesized from DMCU in four steps with 34–48% overall yield. Based on the corresponding LC-MS analysis, water solubility of DMCU, 1-TFA, and 2-TFA at room temperature (25 ± 1 °C) were 0.018, 249.7, and 375.8 mg/mL, respectively, indicating >10,000-fold higher solubility of 1-TFA and 2-TFA than DMCU. Importantly, anti-proliferative assay demonstrated that 2-TFA is a potent anti-cancer agent against LAT1-expressing lung cancer cells NCI-H460, NCI-H358, and A549 cells due to its high intracellular uptake compared to DMCU and 1-TFA. In this study, we logically designed and synthesized the targeted compounds, established the LC-MS analytical methods for evaluations of drug solubility and intracellular uptake levels, and showed improved solubility and anti-cancer activities of 2-TFA. Our results provide a strategical direction for the future development of curcuminoid-like phenolic compounds.
Collapse
Affiliation(s)
- Der-Yen Lee
- Graduate Institute of Integrated Medicine, China Medical University, No. 91, Hsueh-Shih Road, Taichung 40402, Taiwan;
| | - Hui-Yi Lin
- Research Center for Chinese Herbal Medicine, China Medical University, Taichung 40402, Taiwan; (H.-Y.L.); (S.-C.K.)
| | | | - Sheng-Chu Kuo
- Research Center for Chinese Herbal Medicine, China Medical University, Taichung 40402, Taiwan; (H.-Y.L.); (S.-C.K.)
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan;
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung 40447, Taiwan
| | - Pei-Chih Lee
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
- Correspondence: (P.-C.L.); (M.-T.H.)
| | - Min-Tsang Hsieh
- Research Center for Chinese Herbal Medicine, China Medical University, Taichung 40402, Taiwan; (H.-Y.L.); (S.-C.K.)
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan;
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung 40447, Taiwan
- Correspondence: (P.-C.L.); (M.-T.H.)
| |
Collapse
|
13
|
Sanada Y, Takata T, Tanaka H, Sakurai Y, Watanabe T, Suzuki M, Masunaga SI. HIF-1α affects sensitivity of murine squamous cell carcinoma to boron neutron capture therapy with BPA. Int J Radiat Biol 2021; 97:1441-1449. [PMID: 34264166 DOI: 10.1080/09553002.2021.1956004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Purpose To examine whether hypoxia and Hif-1α affect sensitivity of murine squamous cell carcinoma cells to boron neutron capture therapy (BNCT).Materials and methods SCC VII and SCC VII Hif-1α-deficient mouse tumor cells were incubated under normoxic or hypoxic conditions, and cell survival after BNCT was assessed. The intracellular concentration of the 10B-carrier, boronophenylalanine-10B (BPA), was estimated using an autoradiography technique. The expression profile of SLC7A5, which is involved in the uptake of BPA, and the amount of DNA damage caused by BNCT with BPA were examined. A cell survival assay was performed on cell suspensions prepared from tumor-bearing mice.Results Hypoxia ameliorated SCC VII cell survival after neutron irradiation with BPA, but not BSH. Hypoxia-treated SCC VII cells showed decreased intracellular concentrations of BPA and the down-regulated expression of the SLC7A5 protein. BPA uptake and the SLC7A5 protein were not decreased in hypoxia-treated Hif-1α-deficient cells, the survival of which was lower than that of SCC VII cells. More DNA damage was induced in SCC VII Hif-1α-deficient cells than in SCC VII cells. In experiments using tumor-bearing mice, the survival of SCC VII Hif-1α-deficient cells was lower than that of SCC VII cells.Conclusion. Hypoxia may decrease the effects of BNCT with BPA, whereas the disruption of Hif-1α enhanced sensitivity to BNCT with BPA.
Collapse
Affiliation(s)
- Yu Sanada
- Particle Radiation Biology, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Kumatori, Japan
| | - Takushi Takata
- Particle Radiation Medical Physics, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Kumatori, Japan
| | - Hiroki Tanaka
- Particle Radiation Medical Physics, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Kumatori, Japan
| | - Yoshinori Sakurai
- Particle Radiation Medical Physics, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Kumatori, Japan
| | - Tsubasa Watanabe
- Particle Radiation Biology, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Kumatori, Japan
| | - Minoru Suzuki
- Particle Radiation Oncology, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Kumatori, Japan
| | - Shin-Ichiro Masunaga
- Particle Radiation Biology, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Kumatori, Japan
| |
Collapse
|
14
|
Nachef M, Ali AK, Almutairi SM, Lee SH. Targeting SLC1A5 and SLC3A2/SLC7A5 as a Potential Strategy to Strengthen Anti-Tumor Immunity in the Tumor Microenvironment. Front Immunol 2021; 12:624324. [PMID: 33953707 PMCID: PMC8089370 DOI: 10.3389/fimmu.2021.624324] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 03/31/2021] [Indexed: 12/18/2022] Open
Abstract
Cancer cells are metabolically vigorous and are superior in the uptake of nutrients and in the release of the tumor microenvironment (TME)-specific metabolites. They create an acidic, hypoxic, and nutrient-depleted TME that makes it difficult for the cytotoxic immune cells to adapt to the metabolically hostile environment. Since a robust metabolism in immune cells is required for optimal anti-tumor effector functions, the challenges caused by the TME result in severe defects in the invasion and destruction of the established tumors. There have been many recent developments in NK and T cell-mediated immunotherapy, such as engineering them to express chimeric antigen receptors (CARs) to enhance tumor-recognition and infiltration. However, to defeat the tumor and overcome the limitations of the TME, it is essential to fortify these novel therapies by improving the metabolism of the immune cells. One potential strategy to enhance the metabolic fitness of immune cells is to upregulate the expression of nutrient transporters, specifically glucose and amino acid transporters. In particular, the amino acid transporters SLC1A5 and SLC7A5 as well as the ancillary subunit SLC3A2, which are required for efficient uptake of glutamine and leucine respectively, could strengthen the metabolic capabilities and effector functions of tumor-directed CAR-NK and T cells. In addition to enabling the influx and efflux of essential amino acids through the plasma membrane and within subcellular compartments such as the lysosome and the mitochondria, accumulating evidence has demonstrated that the amino acid transporters participate in sensing amino acid levels and thereby activate mTORC1, a master metabolic regulator that promotes cell metabolism, and induce the expression of c-Myc, a transcription factor essential for cell growth and proliferation. In this review, we discuss the regulatory pathways of these amino acid transporters and how we can take advantage of these processes to strengthen immunotherapy against cancer.
Collapse
Affiliation(s)
- Marianna Nachef
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Alaa Kassim Ali
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Saeedah Musaed Almutairi
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Botany and Microbiology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,The University of Ottawa Centre for Infection, Immunity, and Inflammation, Ottawa, ON, Canada
| |
Collapse
|
15
|
Morio H, Reien Y, Hirayama Y, Hashimoto H, Anzai N. Protein kinase C activation upregulates human L-type amino acid transporter 2 function. J Physiol Sci 2021; 71:11. [PMID: 33789576 PMCID: PMC10716992 DOI: 10.1186/s12576-021-00795-0] [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: 10/13/2020] [Accepted: 03/05/2021] [Indexed: 11/10/2022]
Abstract
L-type amino acid transporter 2 (LAT2) is a Na+-independent neutral amino acid transporter, whose function regulation system remains unclarified. Since protein kinase C (PKC) is known to regulate the functions of various transporters, we investigated whether human LAT2 (hLAT2) function is regulated by PKC. In mouse proximal tubule S2 cells, hLAT2 transport activity was upregulated by PKC activation. However, we found that the mRNA and protein expression of hLAT2 was not affected by PKC activation and that the upregulation was independent of the three potential PKC consensus sites in the hLAT2 amino acid sequence. Moreover, we found that PKC activation upregulated the Vmax value for hLAT2-mediated alanine transport, which was not accompanied by the induction of hLAT2 membrane insertion. In conclusion, we showed that hLAT2 function is upregulated by PKC activation, which is not related to either the de novo synthesis, the phosphorylation or the membrane insertion of hLAT2.
Collapse
Affiliation(s)
- Hanae Morio
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan
| | - Yoshie Reien
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan
| | - Yuri Hirayama
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan
| | - Hirofumi Hashimoto
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan.
- Department of Pharmacology and Toxicology, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu-cho, Shimotsuga-gun, Tochigi, 321-0293, Japan.
| |
Collapse
|