1
|
Poudel SB, Chlebek C, Ruff RR, He Z, Xu F, Yildirim G, Hu B, De Jesus CL, Shinde AR, Nayak VV, Witek L, Bromage T, Neubert TA, Rosen CJ, Yakar S. Canagliflozin-Induced Adaptive Metabolism in Bone. Diabetes 2025; 74:812-826. [PMID: 39932694 PMCID: PMC12015145 DOI: 10.2337/db24-0955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/06/2025] [Indexed: 04/23/2025]
Abstract
Sodium-glucose transporter-2 inhibitor (SGLT2i) drugs are widely used for lowering blood glucose levels independent of insulin. Beyond this, these drugs induce various metabolic changes, including weight loss and impaired bone integrity. A significant gap exists in understanding SGLT2i-induced skeletal changes, as SGLT2 is not expressed in osteoblasts or osteocytes, which use glucose to remodel the bone matrix. We studied the impact of 1, 3, or 6 months of canagliflozin (CANA), an SGLT2i treatment, on the skeleton of 6-month-old genetically heterogeneous UM-HET3 mice. Significant metabolic adaptations to CANA were evident as early as 1.5 months after treatment, specifically in male mice. CANA-treated male mice exhibited notable reductions in body weight and decreased proinflammatory and bone remodeling markers associated with reduced cortical bone remodeling indices. Bone tissue metabolome indicated enrichment in metabolites related to amino acid transport and tryptophan catabolism in CANA-treated male mice. In contrast, CANA-treated female mice showed increases in nucleic acid metabolism. An integrOmics approach of source-matched bone tissue metabolome and bone marrow RNA sequencing indicated a positive correlation between the two omics data sets in male mice. Three clusters of transcripts and metabolites involved in energy metabolism, oxidative stress response, and cellular proliferation and differentiation were reduced in CANA-treated male mice. In conclusion, CANA affects bone metabolism mainly via the "glucose restriction state" it induces and impacts bone cell proliferation and differentiation. These findings underline the effects of SGLT2i on bone health and highlight the need to consider sex-specific responses when developing clinical treatments that alter substrate availability. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| | | | - Ryan R. Ruff
- David B. Kriser Dental Center, Department of Epidemiology and Health Promotion, New York University College of Dentistry, New York, NY
| | - Zhiming He
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| | - Fangxi Xu
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| | - Gozde Yildirim
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| | - Bin Hu
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| | - Christopher Lawrence De Jesus
- Department of Neuroscience and Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY
| | - Ankita Raja Shinde
- Department of Mechanical Engineering, New York University Tandon School of Engineering, Brooklyn, NY
| | | | - Lukasz Witek
- Biomaterials Division, New York University College of Dentistry, New York, NY
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, NY
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY
| | - Timothy Bromage
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| | - Thomas A. Neubert
- Department of Neuroscience and Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY
| | | | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| |
Collapse
|
2
|
Jiang C, Qian Y, Bai X, Li S, Zhang L, Xie Y, Lu Y, Lu Z, Liu B, Jiang BH. SLC7A5/E2F1/PTBP1/PKM2 axis mediates progression and therapy effect of triple-negative breast cancer through the crosstalk of amino acid metabolism and glycolysis pathway. Cancer Lett 2025; 617:217612. [PMID: 40054655 DOI: 10.1016/j.canlet.2025.217612] [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: 01/20/2025] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Triple-negative breast cancer (TNBC) is one of the most challenging malignancies with the highest mortality rates among women. TNBC relies on both amino acid metabolism and glycolysis to fuel its bioenergetic and biosynthetic demands. However, the potential crosstalk between these two metabolic pathways and its impact on TNBC progression remain largely unexplored. In this study, we observed that SLC7A5, a key amino acid transporter, was upregulated in TNBC and strongly associated with poor patient prognosis. We demonstrated that the elevated SLC7A5 expression activated the amino acid pathway and promoted cell proliferation, tumor growth, and therapeutic resistance by inducing the switch from PKM1 to PKM2 expression, thereby mediating the crosstalk between amino acid metabolism and glycolysis. We further identified that the upregulation of SLC7A5 resulted from miR-152 suppression, which regulates TNBC cellular function and tumor growth. In addition, the miR-152/SLC7A5 axis mediated the expression of PTBP1, which maintains the balance between PKM1 and PKM2, linking amino acid signaling with the glycolysis pathway. To further understand the mechanism of PTBP1 upregulation, we identified that E2F1 transcriptionally activated PTBP1 expression through direct binding at the seed site, while E2F1 expression was also induced by SLC7A5 in TNBC. This novel SLC7A5/E2F1/PTBP1 axis plays a crucial role in regulating the crosstalk between amino acid signaling and glycolysis in TNBC and is essential for TNBC progression and therapeutic effectiveness. Our findings offer valuable insights into the molecular mechanisms underlying TNBC metabolic reprogramming and highlight potential targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Chengfei Jiang
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China; Department of Pathology, Nanjing Medical University, Nanjing, 210029, China
| | - Yingchen Qian
- Department of Pathology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, China
| | - Xiaoming Bai
- Department of Pathology, Nanjing Medical University, Nanjing, 210029, China
| | - Shuangya Li
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Liyuan Zhang
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Yunxia Xie
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Yifan Lu
- Department of Pathology, Nanjing Medical University, Nanjing, 210029, China
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310029, China
| | - Bingjie Liu
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China.
| | - Bing-Hua Jiang
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
3
|
Zhao Y, Gao J, Feng H, Jiang L. GRAMMAR-Lambda Delivers Efficient Understanding of the Genetic Basis for Head Size in Catfish. BIOLOGY 2025; 14:63. [PMID: 39857294 PMCID: PMC11760490 DOI: 10.3390/biology14010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025]
Abstract
The shape of the skull plays a crucial role in the evolution and adaptation of species to their environments. In the case of aquaculture fish, the size of the head is also an important economic trait, as it is linked to fillet yield and ornamental value. This study applies our GRAMMAR-Lambda method to perform a genome-wide association study analysis on loci related to head size in catfish. Compared with traditional GWAS methods, the GRAMMAR-Lambda method offers higher computational efficiency, statistical power, and stability, especially in complex population structures. This research identifies many candidate genes closely related to cranial morphology in terms of head length, width, and depth in catfish, including bmpr1bb, fgfrl1b, nipbl, foxp2, and pax5, etc. Based on the results of gene-gene interaction analysis, we speculate that there may be frequent genetic interactions between chromosome 19 and chromosome 29 in bone development. Additionally, many candidate genes, gene families, and mechanisms (such as SOCE mechanisms) affecting skeletal development and morphology have been identified. These findings contribute to our understanding of the genetic architecture of head size and will support marker-assisted breeding in aquaculture, also reflecting the potential application of the GRAMMAR-Lambda method in genetic studies of complex traits.
Collapse
Affiliation(s)
- Yunfeng Zhao
- Hainan Fisheries Innovation Research Institute, Chinese Academy of Fishery Sciences, Sanya 572024, China
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Jin Gao
- Hainan Academy of Ocean and Fisheries Sciences, Haikou 571126, China;
| | - Hong Feng
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong SAR, China;
| | - Li Jiang
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| |
Collapse
|
4
|
Su S, Tian L. Association Between Dietary Tryptophan Intake and Bone Health: A Cross-Sectional Study. Calcif Tissue Int 2024; 116:6. [PMID: 39673557 DOI: 10.1007/s00223-024-01329-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
The relationship between dietary tryptophan intake and the risk of low bone mineral density (LBMD) has not been thoroughly evaluated. This study aimed to examine the relationship between dietary tryptophan intake and LBMD. A total of 12,003 participants aged 50 years and older with complete data on bone mineral density (BMD) and tryptophan intake from the National Health and Nutrition Examination Survey (NHANES) 2005 to 2020 were included in this cross-sectional study. The median dietary tryptophan intake among the 12,003 participants was 1822.14 mg/day, with significantly lower levels observed in individuals with LBMD compared to those with normal bone mass (1740.45 mg/day vs. 2041.39 mg/day, p < 0.001). For every 2.7-fold increase in dietary tryptophan intake, the risk of low BMD decreases by 22%. When dietary tryptophan intake was categorized into quartiles, significantly lower risks of LBMD were observed in the third [Odds Ratio (OR) = 0.68, 95% confidence interval (CI): 0.51-0.91] and fourth (OR = 0.65, 95% CI: 0.49-0.87) quartiles compared to the reference group after multivariable adjustment. Moreover, the restricted cubic spline (RCS) results revealed a negative nonlinear relationship between dietary tryptophan intake and LBMD (p for overall < 0.001, p for nonlinear < 0.05), with this correlation remaining consistent across various population subgroups and exhibiting no significant interaction according to stratification variables. Sensitivity analyses further substantiated these findings. Overall, we found that increased dietary tryptophan intake may be associated with a lower risk of LBMD among individuals aged ≥ 50 years, highlighting the importance of optimizing tryptophan nutrition for reducing osteoporosis risk.
Collapse
Affiliation(s)
- Shan Su
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Limin Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China.
- Department of Endocrinology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| |
Collapse
|
5
|
Hendi NN, Bengoechea-Alonso MT, Ericsson J, Nemer G. Functional characterization of the SDR42E1 reveals its role in vitamin D biosynthesis. Heliyon 2024; 10:e36466. [PMID: 39263177 PMCID: PMC11387231 DOI: 10.1016/j.heliyon.2024.e36466] [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: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
Vitamin D deficiency poses a widespread health challenge, shaped by environmental and genetic determinants. A recent discovery identified a genetic regulator, rs11542462, in the SDR42E1 gene, though its biological implications remain largely unexplored. Our bioinformatic assessments revealed pronounced SDR42E1 expression in skin keratinocytes and the analogous HaCaT human keratinocyte cell lines, prompting us to select the latter as an experimental model. Employing CRISPR/Cas9 gene-editing technology and multi-omics approach, we discovered that depleting SDR42E1 showed a 1.6-fold disruption in steroid biosynthesis pathway (P-value = 0.03), considerably affecting crucial vitamin D biosynthesis regulators. Notably, SERPINB2 (P-value = 2.17 × 10-103), EBP (P-value = 2.46 × 10-13), and DHCR7 (P-value = 8.03 × 10-09) elevated by ∼2-3 fold, while ALPP (P-value <2.2 × 10-308), SLC7A5 (P-value = 1.96 × 10-215), and CYP26A1 (P-value = 1.06 × 10-08) downregulated by ∼1.5-3 fold. These alterations resulted in accumulation of 7-dehydrocholesterol precursor and reduction of vitamin D3 production, as evidenced by the drug enrichment (P-value = 4.39 × 10-06) and total vitamin D quantification (R2 = 0.935, P-value = 0.0016) analyses. Our investigation unveils SDR42E1's significance in vitamin D homeostasis, emphasizing the potential of precision medicine in addressing vitamin D deficiency through understanding its genetic basis.
Collapse
Affiliation(s)
- Nagham Nafiz Hendi
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, P.O. Box 34110, Qatar
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, P.O. Box 34110, Qatar
- Systems Biology and Immunology Department, Sidra Medicine, Doha, P.O. Box 26999, Qatar
| | - Maria Teresa Bengoechea-Alonso
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, P.O. Box 34110, Qatar
| | - Johan Ericsson
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, P.O. Box 34110, Qatar
| | - Georges Nemer
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, P.O. Box 34110, Qatar
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, P.O. Box 110236, Lebanon
| |
Collapse
|
6
|
Stegen S, Carmeliet G. Metabolic regulation of skeletal cell fate and function. Nat Rev Endocrinol 2024; 20:399-413. [PMID: 38499689 DOI: 10.1038/s41574-024-00969-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/23/2024] [Indexed: 03/20/2024]
Abstract
Bone development and bone remodelling during adult life are highly anabolic processes requiring an adequate supply of oxygen and nutrients. Bone-forming osteoblasts and bone-resorbing osteoclasts interact closely to preserve bone mass and architecture and are often located close to blood vessels. Chondrocytes within the developing growth plate ensure that bone lengthening occurs before puberty, but these cells function in an avascular environment. With ageing, numerous bone marrow adipocytes appear, often with negative effects on bone properties. Many studies have now indicated that skeletal cells have specific metabolic profiles that correspond to the nutritional microenvironment and their stage-specific functions. These metabolic networks provide not only skeletal cells with sufficient energy, but also biosynthetic intermediates that are necessary for proliferation and extracellular matrix synthesis. Moreover, these metabolic pathways control redox homeostasis to avoid oxidative stress and safeguard cell survival. Finally, several intracellular metabolites regulate the activity of epigenetic enzymes and thus control the fate and function of skeletal cells. The metabolic profile of skeletal cells therefore not only reflects their cellular state, but can also drive cellular activity. Insight into skeletal cell metabolism will thus not only advance our understanding of skeletal development and homeostasis, but also of skeletal disorders, such as osteoarthritis, diabetic bone disease and bone malignancies.
Collapse
Affiliation(s)
- Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Rosario FJ, Barentsen K, Powell TL, Urschitz J, Brown TL, Kanai Y, Jansson T. Trophoblast-specific overexpression of the LAT1 increases transplacental transport of essential amino acids and fetal growth in mice. PNAS NEXUS 2024; 3:pgae207. [PMID: 38894879 PMCID: PMC11184900 DOI: 10.1093/pnasnexus/pgae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024]
Abstract
Placental System L amino acid transporter activity is decreased in pregnancies complicated by intrauterine growth restriction (IUGR) and increased in fetal overgrowth. However, it is unknown if changes in the expression/activity of placental Large Neutral Amino Acid Transporter Small Subunit 1 (Slc7a5/LAT1) are mechanistically linked to placental function and fetal growth. We hypothesized that trophoblast-specific Slc7a5 overexpression increases placental transport of essential amino acids, activates the placental mechanistic target of rapamycin (mTOR) signaling, and promotes fetal growth in mice. Using lentiviral transduction of blastocysts with a Slc7a5 transgene, we achieved trophoblast-specific overexpression of Slc7a5 (Slc7a5 OX) with increased fetal (+27%) and placental weights (+10%). Trophoblast-specific Slc7a5 overexpression increased trophoblast plasma membrane (TPM) LAT1 protein abundance and TPM System L transporter (+53%) and System A transporter activity (+ 21%). Slc7a5 overexpression also increased transplacental transport of leucine (+ 85%) but not of the System A tracer, 14C-methylamino isobutyric acid, in vivo. Trophoblast-specific overexpression of Slc7a5 activated placental mTORC1, as assessed by increased (+44%) phosphorylation of S6 ribosomal protein (Ser 235/236), and mTORC2 as indicated by phosphorylation of PKCα-Tyr-657 (+47%) and Akt-Ser 473 (+96%). This is the first demonstration that placental transport of essential amino acids is mechanistically linked to fetal growth. The decreased placental System L activity in human IUGR and the increased placental activity of this transporter in some cases of fetal overgrowth may directly contribute to the development of these pregnancy complications.
Collapse
Affiliation(s)
- Fredrick J Rosario
- Department of Obstetrics and Gynecology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth Barentsen
- Department of Obstetrics and Gynecology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Theresa L Powell
- Department of Obstetrics and Gynecology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Johann Urschitz
- Institue of Biogenesis, University of Hawaii, Honolulu, HI 96822, USA
| | - Thomas L Brown
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University Boonshoft School of Medicine, Dayton, OH 45435, USA
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
8
|
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
|
9
|
Takegahara N, Kim H, Choi Y. Unraveling the intricacies of osteoclast differentiation and maturation: insight into novel therapeutic strategies for bone-destructive diseases. Exp Mol Med 2024; 56:264-272. [PMID: 38297158 PMCID: PMC10907717 DOI: 10.1038/s12276-024-01157-7] [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: 08/18/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 02/02/2024] Open
Abstract
Osteoclasts are the principal cells that efficiently resorb bone. Numerous studies have attempted to reveal the molecular pathways leading to the differentiation and activation of osteoclasts to improve the treatment and prevention of osteoporosis and other bone-destructive diseases. While the cumulative knowledge of osteoclast regulatory molecules, such as receptor activator of nuclear factor-kB ligand (RANKL) and nuclear factor of activated T cells 1 (NFATc1), contributes to the understanding of the developmental progression of osteoclasts, little is known about how the discrete steps of osteoclastogenesis modify osteoclast status but not the absolute number of osteoclasts. The regulatory mechanisms involved in osteoclast maturation but not those involved in differentiation deserve special attention due to their potential use in establishing a more effective treatment strategy: targeting late-phase differentiation while preserving coupled bone formation. Recent studies have shed light on the molecules that govern late-phase osteoclast differentiation and maturation, as well as the metabolic changes needed to adapt to shifting metabolic demands. This review outlines the current understanding of the regulation of osteoclast differentiation, as well as osteoclast metabolic adaptation as a differentiation control mechanism. Additionally, this review introduces molecules that regulate the late-phase osteoclast differentiation and thus minimally impact coupled bone formation.
Collapse
Affiliation(s)
- Noriko Takegahara
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Hyunsoo Kim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
10
|
Zheng Y, Gao N, Li Y, Fan M, Tian W, Jiang Y, Wang Y, Cui M, Suo C, Zhang T, Jin L, Xu K, Chen X. Unraveling the role of serum metabolites in the relationship between plant-based diets and bone health in community-dwelling older adults. Curr Res Food Sci 2024; 8:100687. [PMID: 38318314 PMCID: PMC10839558 DOI: 10.1016/j.crfs.2024.100687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
The potential adverse effects of the plant-based dietary pattern on bone health have received widespread attention. However, the biological mechanisms underlying the adverse effects of plant-based diets on bone health remain incompletely understood. The objective of this study was to identify potential biomarkers between plant-based diets and bone loss utilizing metabolomic techniques in the Taizhou Imaging Study (TIS) (N = 788). Plant-based diet indexes (overall plant-based diet index (PDI), healthy plant-based diet index (hPDI), and unhealthy plant-based diet index (uPDI)) were calculated using the food frequency questionnaire, and bone mineral density (BMD) was measured using dual-energy X-ray absorptiometry. A multinomial logistic regression was used to explore the associations of plant-based diet indexes with bone loss. Furthermore, mediation analysis and exploratory factor analysis (EFA) were performed to explore the mediated effects of metabolites on the association of plant-based diets with BMD T-score. Our results showed that higher hPDI and uPDI were positively associated with bone loss. Moreover, nineteen metabolites were significantly associated with BMD T-score, among them, seven metabolites were associated with uPDI. Except for cholesterol esters in VLDL-1, the remaining six metabolites significantly mediated the negative association between uPDI and BMD T-score. Interestingly, we observed that the same six metabolites mediated the positive association between fresh fruit and BMD T-score. Collectively, our results support the deleterious effects of plant-based diets on bone health and discover the potential mediation effect of metabolites on the association of plant-based diets with bone loss. The findings offer valuable insights that could optimize dietary recommendations and interventions, contributing to alleviate the potential adverse effects associated with plant-based diets.
Collapse
Affiliation(s)
- Yi Zheng
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Ningxin Gao
- Department of Biostatistics, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Yucan Li
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Min Fan
- Taixing Disease Control and Prevention Center, Taizhou, Jiangsu, China
| | - Weizhong Tian
- Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Yingzhe Wang
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Mei Cui
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Suo
- Department of Biostatistics, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Tiejun Zhang
- Department of Biostatistics, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Kelin Xu
- Department of Biostatistics, School of Public Health, The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, China
| |
Collapse
|
11
|
Ledesma-Colunga MG, Passin V, Lademann F, Hofbauer LC, Rauner M. Novel Insights into Osteoclast Energy Metabolism. Curr Osteoporos Rep 2023; 21:660-669. [PMID: 37816910 PMCID: PMC10724336 DOI: 10.1007/s11914-023-00825-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2023] [Indexed: 10/12/2023]
Abstract
PURPOSE OF REVIEW Osteoclasts are crucial for the dynamic remodeling of bone as they resorb old and damaged bone, making space for new bone. Metabolic reprogramming in these cells not only supports phenotypic changes, but also provides the necessary energy for their highly energy-consuming activity, bone resorption. In this review, we highlight recent developments in our understanding of the metabolic adaptations that influence osteoclast behavior and the overall remodeling of bone tissue. RECENT FINDINGS Osteoclasts undergo metabolic reprogramming to meet the energy demands during their transition from precursor cells to fully mature bone-resorbing osteoclasts. Recent research has made considerable progress in pinpointing crucial metabolic adaptations and checkpoint proteins in this process. Notably, glucose metabolism, mitochondrial biogenesis, and oxidative respiration were identified as essential pathways involved in osteoclast differentiation, cytoskeletal organization, and resorptive activity. Furthermore, the interaction between these pathways and amino acid and lipid metabolism adds to the complexity of the process. These interconnected processes can function as diverse fuel sources or have independent regulatory effects, significantly influencing osteoclast function. Energy metabolism in osteoclasts involves various substrates and pathways to meet the energetic requirements of osteoclasts throughout their maturation stages. This understanding of osteoclast biology may provide valuable insights for modulating osteoclast activity during the pathogenesis of bone-related disorders and may pave the way for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Maria G Ledesma-Colunga
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany
| | - Vanessa Passin
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany
| | - Franziska Lademann
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany.
| |
Collapse
|
12
|
Grahnemo L, Eriksson AL, Nethander M, Johansson R, Lorentzon M, Mellström D, Pettersson-Kymmer U, Ohlsson C. Low Circulating Valine Associate With High Risk of Hip Fractures. J Clin Endocrinol Metab 2023; 108:e1384-e1393. [PMID: 37178220 PMCID: PMC10583993 DOI: 10.1210/clinem/dgad268] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
CONTEXT Hip fractures constitute a major health concern. An adequate supply of amino acids is crucial to ensure optimal acquisition and remodeling of bone. Circulating amino acid levels have been proposed as markers of bone mineral density, but data on their ability to predict incident fractures are scarce. OBJECTIVES To investigate the associations between circulating amino acids and incident fractures. METHODS We used UK Biobank (n = 111 257; 901 hip fracture cases) as a discovery cohort and the Umeå Fracture and Osteoporosis (UFO) hip fracture study (hip fracture cases n = 2225; controls n = 2225) for replication. Associations with bone microstructure parameters were tested in a subsample of Osteoporotic Fractures in Men Sweden (n = 449). RESULTS Circulating valine was robustly associated with hip fractures in the UK Biobank (HR per SD increase 0.79, 95% CI 0.73-0.84), and this finding was replicated in the UFO study (combined meta-analysis including 3126 incident hip fracture cases, odds ratio per SD increase 0.84, 95% CI 0.80-0.88). Detailed bone microstructure analyses showed that high circulating valine was associated with high cortical bone area and trabecular thickness. CONCLUSION Low circulating valine is a robust predictor of incident hip fractures. We propose that circulating valine may add information for hip fracture prediction. Future studies are warranted to determine whether low valine is causally associated with hip fractures.
Collapse
Affiliation(s)
- Louise Grahnemo
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Anna L Eriksson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden
- Region Västra Götaland, Department of Drug Treatment, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
| | - Maria Nethander
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden
- Sahlgrenska Academy, Bioinformatics and Data Centre, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Robert Johansson
- The Biobank Research Unit, Umeå University, SE-90187 Umeå, Sweden
| | - Mattias Lorentzon
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden
- Geriatric Medicine, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg and Geriatric Medicine, Sahlgrenska University Hospital, 43180 Mölndal, Sweden
- Mary McKillop Institute for Health Research, Australian Catholic University, 3000 VIC, Melbourne, Australia
| | - Dan Mellström
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden
- Geriatric Medicine, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg and Geriatric Medicine, Sahlgrenska University Hospital, 43180 Mölndal, Sweden
| | - Ulrika Pettersson-Kymmer
- Clinical Pharmacology, Department of Integrative Medical Biology, Umeå University, SE-90197 Umeå, Sweden
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, University of Gothenburg, SE-413 45 Gothenburg, Sweden
- Region Västra Götaland, Department of Drug Treatment, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
| |
Collapse
|
13
|
Iwasaki R, Yoshikawa R, Umeno R, Seki A, Matsukawa T, Takeno S, Yokoyama K, Mori T, Suzuki M, Ono K. The effects of BPA-BNCT on normal bone: determination of the CBE value in mice‡. JOURNAL OF RADIATION RESEARCH 2023; 64:795-803. [PMID: 37517393 PMCID: PMC10516729 DOI: 10.1093/jrr/rrad054] [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: 06/05/2023] [Revised: 07/03/2023] [Indexed: 08/01/2023]
Abstract
Boron neutron capture therapy (BNCT) with p-boronophenylalanine (BPA) is expected to have less effect on the decrease in normal bone strength than X-ray therapy. However, the compound biological effectiveness (CBE) value necessary to convert the boron neutron capture reaction (BNCR) dose into a bioequivalent X-ray dose has not been determined yet. The purpose of this study was to evaluate the influence of BNCT on normal bone in mice and to elucidate the CBE factor. We first searched the distribution of BPA in the normal bone of C3H/He mice and then measured the changes in bone strength after irradiation. The CBE value was determined when the decrease in bone strength was set as an index of the BNCT effect. The 10B concentrations in the tibia after subcutaneous injection of 125, 250 and 500 mg/kg BPA were measured by prompt gamma-ray spectroscopy and inductively coupled plasma (ICP)-atomic emission spectrometry. The 10B mapping in the tibia was examined by alpha-track autoradiography and laser ablation-ICP-mass spectrometry. The 10B concentration increased dose-dependently; moreover, the concentrations were maintained until 120 min after BPA administration. The administered 10B in the tibia was abundantly accumulated in the growth cartilage, trabecular bone and bone marrow. The bone strength was analyzed by a three-point bending test 12 weeks after irradiation. The bending strength of the tibia decreased dose-dependently after the irradiation of X-ray, neutron and BNCR. The CBE factor was obtained as 2.27 by comparing these dose-effect curves; the value determined in this study will enable an accurate dosimetry of normal bone.
Collapse
Affiliation(s)
- Ryota Iwasaki
- Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Ryutaro Yoshikawa
- Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Ryo Umeno
- Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Azusa Seki
- HAMRI Co. Ltd., 2638-2 Ozaki, Koga-shi, Ibaragi 306-0101, Japan
| | - Takehisa Matsukawa
- Department of Epidemiology and Environmental Health, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Forensic Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Satoshi Takeno
- Department of Radiation Oncology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi Takatsuki-shi, Osaka 569-8686, Japan
- Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi Takatsuki-shi, Osaka 569-8686, Japan
| | - Kazuhito Yokoyama
- Department of Epidemiology and Environmental Health, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Epidemiology and Social Medicine, International University of Health and Welfare, 4-1-26 Akasaka, Minato-ku, Tokyo 107-8402, Japan
| | - Takashi Mori
- Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Minoru Suzuki
- Particle Radiation Oncology Research Center, Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Koji Ono
- Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi Takatsuki-shi, Osaka 569-8686, Japan
| |
Collapse
|
14
|
Wu G, Liu Z, Mu C, Song D, Wang J, Meng X, Li Z, Qing H, Dong Y, Xie HY, Pang DW. Enhanced Proliferation of Visualizable Mesenchymal Stem Cell-Platelet Hybrid Cell for Versatile Intracerebral Hemorrhage Treatment. ACS NANO 2023; 17:7352-7365. [PMID: 37037487 DOI: 10.1021/acsnano.2c11329] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The intrinsic features and functions of platelets and mesenchymal stem cells (MSCs) indicate their great potential in the treatment of intracerebral hemorrhage (ICH). However, neither of them can completely overcome ICH because of the stealth process and the complex pathology of ICH. Here, we fabricate hybrid cells for versatile and highly efficient ICH therapy by fusing MSCs with platelets and loading with lysophosphatidic acid-modified PbS quantum dots (LPA-QDs). The obtained LPA-QDs@FCs (FCs = fusion cells) not only inherit the capabilities of both platelets and MSCs but also exhibit clearly enhanced proliferation activated by LPA. After systemic administration, many proliferating LPA-QDs@FCs rapidly accumulate in ICH areas for responding to the vascular damage and inflammation and then efficiently prevent both the primary and secondary injuries of ICH but with no obvious side effects. Moreover, the treatment process can be tracked by near-infrared II fluorescence imaging with highly spatiotemporal resolution, providing a promising solution for ICH therapy.
Collapse
Affiliation(s)
- Guanghao Wu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Zhenya Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
| | - Changwen Mu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Da Song
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaxin Wang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, P. R. China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, P. R. China
| | - Ziyuan Li
- Department of Biomedical Engineering, Peking University, Beijing 100871, P. R. China
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yuping Dong
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Hai-Yan Xie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
15
|
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 Paneth cell function to affect the intestinal inflammatory response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.524966. [PMID: 36789439 PMCID: PMC9928054 DOI: 10.1101/2023.01.24.524966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The intestine is critical for not only processing and resorbing 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-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 Slc7a5 ΔIEC reduces mTORC1 signaling. Surprisingly, Slc7a5 ΔIEC mice have increased cell proliferation but reduced secretory cells, particularly mature Paneth cells. scRNA-seq and electron microscopic analyses revealed dedifferentiation of Paneth cells in Slc7a5 ΔIEC mice, leading to markedly reduced secretory granules with little effect on Paneth cell number. We further show that Slc7a5 ΔIEC mice are prone to experimental colitis. Thus, SLC7A5 regulates secretory cell differentiation to affect stem cell niche and/or inflammatory response to regulate cell proliferation.
Collapse
|
16
|
Blinova AA, Karamirzoev AA, Guseynova AR, Maglakelidze DG, Ilyaeva TA, Gusov BA, Meliksetyants AP, Pirumian MM, Taravanov MA, Pirogov MA, Vakalov DS, Bernyukevich TV, Gvozdenko AA, Nagdalian AA, Blinov AV. Synthesis and Characterization of Calcium Silicate Nanoparticles Stabilized with Amino Acids. MICROMACHINES 2023; 14:245. [PMID: 36837945 PMCID: PMC9967975 DOI: 10.3390/mi14020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
This work presents the development of a method for the synthesis of calcium silicate nanoparticles stabilized with essential amino acids. CaSiO3 nanoparticles were obtained through chemical precipitation. In the first stage, the optimal calcium-containing precursor was determined. The samples were examined using scanning electron microscopy. It was found that Ca(CH3COO)2 was the optimal calcium-containing precursor. Then, the phase composition of calcium silicate was studied using X-ray phase analysis. The results showed the presence of high-intensity bands in the diffractogram, which characterized the phase of the nanosized CaSiO3-wollastonite. In the next stage, the influence of the type of amino acid on the microstructure of calcium silicate was studied. The amnio acids studied were valine, L-leucine, L-isoleucine, L-methionine, L-threonine, L-lysine, L-phenylalanine, and L-tryptophan. The analysis of the SEM micrographs showed that the addition of amino acids did not significantly affect the morphology of the CaSiO3 samples. The surface of the CaSiO3 samples, both without a stabilizer and with amino acids, was represented by irregularly shaped aggregates consisting of nanoparticles with a diameter of 50-400 nm. Further, in order to determine the optimal amino acid to use to stabilize nanoparticles, computerized quantum chemical modeling was carried out. Analysis of the data obtained showed that the most energetically favorable interaction was the CaSiO3-L-methionine configuration, where the interaction occurs through the amino group of the amino acid; the energy value of which was -2058.497 kcal/mol. To confirm the simulation results, the samples were examined using IR spectroscopy. An analysis of the results showed that the interaction of calcium silicate with L-methionine occurs via the formation of a bond through the NH3+ group of the amino acid.
Collapse
Affiliation(s)
- Anastasiya A. Blinova
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | | | - Asiyat R. Guseynova
- Faculty of Dentistry, Derzhavin Tambov State University, 392008 Tambov, Russia
| | - David G. Maglakelidze
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Tatiana A. Ilyaeva
- Faculty of Medicine, Stavropol State Medical University, 355017 Stavropol, Russia
| | - Batradz A. Gusov
- Faculty of Dentistry, North Ossetian State Medical University, 362025 Vladikavkaz, Russia
| | | | - Mari M. Pirumian
- Medical and Preventive Faculty, Rostov State Medical University, 344022 Rostov-on-Don, Russia
| | - Maxim A. Taravanov
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Maxim A. Pirogov
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Dmitriy S. Vakalov
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | | | - Alexey A. Gvozdenko
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Andrey A. Nagdalian
- Laboratory of Food and Industrial Biotechnology, North Caucasus Federal University, 355017 Stavropol, Russia
| | - Andrey V. Blinov
- Department of Physics and Technology of Nanostructures and Materials, Physical and Technical Faculty, North Caucasus Federal University, 355017 Stavropol, Russia
| |
Collapse
|
17
|
Abstract
Amino acid metabolism regulates essential cellular functions, not only by fueling protein synthesis, but also by supporting the biogenesis of nucleotides, redox factors and lipids. Amino acids are also involved in tricarboxylic acid cycle anaplerosis, epigenetic modifications, next to synthesis of neurotransmitters and hormones. As such, amino acids contribute to a broad range of cellular processes such as proliferation, matrix synthesis and intercellular communication, which are all critical for skeletal cell functioning. Here we summarize recent work elucidating how amino acid metabolism supports and regulates skeletal cell function during bone growth and homeostasis, as well as during skeletal disease. The most extensively studied amino acid is glutamine, and osteoblasts and chondrocytes rely heavily on this non-essential amino acid during for their functioning and differentiation. Regulated by lineage-specific transcription factors such as SOX9 and osteoanabolic agents such as parathyroid hormone or WNT, glutamine metabolism has a wide range of metabolic roles, as it fuels anabolic processes by producing nucleotides and non-essential amino acids, maintains redox balance by generating the antioxidant glutathione and regulates cell-specific gene expression via epigenetic mechanisms. We also describe how other amino acids affect skeletal cell functions, although further work is needed to fully understand their effect. The increasing number of studies using stable isotope labelling in several skeletal cell types at various stages of differentiation, together with conditional inactivation of amino acid transporters or enzymes in mouse models, will allow us to obtain a more complete picture of amino acid metabolism in skeletal cells.
Collapse
Affiliation(s)
| | | | - Steve Stegen
- Corresponding author at: Clinical and Experimental Endocrinology, KU Leuven, O&N1bis, Herestraat 49 box 902, 3000 Leuven, Belgium.
| |
Collapse
|
18
|
Jiménez JA, Lawlor ER, Lyssiotis CA. Amino acid metabolism in primary bone sarcomas. Front Oncol 2022; 12:1001318. [PMID: 36276057 PMCID: PMC9581121 DOI: 10.3389/fonc.2022.1001318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/19/2022] [Indexed: 12/30/2022] Open
Abstract
Primary bone sarcomas, including osteosarcoma (OS) and Ewing sarcoma (ES), are aggressive tumors with peak incidence in childhood and adolescence. The intense standard treatment for these patients consists of combined surgery and/or radiation and maximal doses of chemotherapy; a regimen that has not seen improvement in decades. Like other tumor types, ES and OS are characterized by dysregulated cellular metabolism and a rewiring of metabolic pathways to support the biosynthetic demands of malignant growth. Not only are cancer cells characterized by Warburg metabolism, or aerobic glycolysis, but emerging work has revealed a dependence on amino acid metabolism. Aside from incorporation into proteins, amino acids serve critical functions in redox balance, energy homeostasis, and epigenetic maintenance. In this review, we summarize current studies describing the amino acid metabolic requirements of primary bone sarcomas, focusing on OS and ES, and compare these dependencies in the normal bone and malignant tumor contexts. We also examine insights that can be gleaned from other cancers to better understand differential metabolic susceptibilities between primary and metastatic tumor microenvironments. Lastly, we discuss potential metabolic vulnerabilities that may be exploited therapeutically and provide better-targeted treatments to improve the current standard of care.
Collapse
Affiliation(s)
- Jennifer A. Jiménez
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Elizabeth R. Lawlor
- Department of Pediatrics, University of Washington, Seattle, WA, United States,Seattle Children’s Research Institute, Seattle, WA, United States,*Correspondence: Elizabeth R. Lawlor, ; Costas A. Lyssiotis,
| | - Costas A. Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States,Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI, United States,*Correspondence: Elizabeth R. Lawlor, ; Costas A. Lyssiotis,
| |
Collapse
|
19
|
Iwahashi S, Lyu J, Tokumura K, Osumi R, Hiraiwa M, Kubo T, Horie T, Demura S, Kawakami N, Saito T, Park G, Fukasawa K, Iezaki T, Suzuki A, Tomizawa A, Ochi H, Hojo H, Ohba S, Hinoi E. Conditional inactivation of the L-type amino acid transporter LAT1 in chondrocytes models idiopathic scoliosis in mice. J Cell Physiol 2022; 237:4292-4302. [PMID: 36161979 DOI: 10.1002/jcp.30883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/08/2022]
Abstract
Scoliosis, usually diagnosed in childhood and early adolescence, is an abnormal lateral curvature of the spine. L-type amino acid transporter 1 (LAT1), encoded by solute carrier transporter 7a5 (Slc7a5), plays a crucial role in amino acid sensing and signaling in specific cell types. We previously demonstrated the pivotal role of LAT1 on bone homeostasis in mice, and the expression of LAT1/SLC7A5 in vertebral cartilage of pediatric scoliosis patients; however, its role in chondrocytes on spinal homeostasis and implications regarding the underlying mechanisms during the onset and progression of scoliosis, remain unknown. Here, we identified LAT1 in mouse chondrocytes as an important regulator of postnatal spinal homeostasis. Conditional inactivation of LAT1 in chondrocytes resulted in a postnatal-onset severe thoracic scoliosis at the early adolescent stage with normal embryonic spinal development. Histological analyses revealed that Slc7a5 deletion in chondrocytes led to general disorganization of chondrocytes in the vertebral growth plate, along with an increase in apoptosis and a decrease in cell proliferation. Furthermore, loss of Slc7a5 in chondrocytes activated the general amino acid control (GAAC) pathway but inactivated the mechanistic target of rapamycin complex 1 (mTORC1) pathway in the vertebrae. The spinal deformity in Slc7a5-deficient mice was corrected by genetic inactivation of the GAAC pathway, but not by genetic activation of the mTORC1 pathway. These findings suggest that the LAT1-GAAC pathway in chondrocytes plays a critical role in the maintenance of proper spinal homeostasis by modulating cell proliferation and survivability.
Collapse
Affiliation(s)
- Sayuki Iwahashi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Jiajun Lyu
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Kazuya Tokumura
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Ryoma Osumi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Manami Hiraiwa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Takuya Kubo
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Tetsuhiro Horie
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Satoru Demura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Noriaki Kawakami
- Department of Orthopaedic Surgery, Ichinomiya Nishi Hospital, Ichinomiya, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Gyujin Park
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Kazuya Fukasawa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Takashi Iezaki
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Akane Suzuki
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Akane Tomizawa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Hiroki Ochi
- Department of Rehabilitation for Motor Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Saitama, Japan
| | - Hironori Hojo
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Shinsuke Ohba
- Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Eiichi Hinoi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan.,United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
20
|
Demura S, Hinoi E, Kawakami N, Handa M, Yokogawa N, Hiraiwa M, Kato S, Shinmura K, Shimizu T, Oku N, Annen R, Kobayashi M, Yamada Y, Nagatani S, Iezaki T, Taniguchi Y, Tsuchiya H. The L-type Amino Acid Transporter (LAT1) Expression in Patients with Scoliosis. Spine Surg Relat Res 2022; 6:402-407. [PMID: 36051676 PMCID: PMC9381085 DOI: 10.22603/ssrr.2021-0189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/11/2021] [Indexed: 01/04/2023] Open
Abstract
Introduction Amino acid transporters are transmembrane proteins that are known to mediate the transfer of amino acids. As one of the amino acid transporters, LAT1, which is encoded by Slc7a5, mediates the cellular uptake of the essential amino acids. Recently, most studies have focused on examining the relationship between LAT1 and skeletal formation in terms of development. However, little is known regarding the clinical features of LAT1 in the cartilage, which might result in the development of skeletal deformities such as scoliosis. Thus, the aim of this study was to investigate the expression of L-type amino acid transporter 1 (LAT1) and its solute carrier transporter 7a5 (Slc7a5) in patients with pediatric scoliosis and to compare with the relationship between LAT1 and Slc7a5 expression and their clinical features. Methods We have prospectively recruited 56 patients who underwent corrective spinal fusion for scoliosis. The patients comprised 40 girls and 16 boys, with a mean age of 13.1 years at the time of surgery. There were 34 idiopathic scoliosis (IS) patients, whereas 22 were congenital scoliosis (CS) patients. During the surgery, an epiphyseal part of the spinous process at apical vertebra was harvested; then, LAT1 and Slc7a5 expressions in the cartilage were evaluated. Results As per our findings, LAT1 expression was observed in the cartilage in 60.7% (34 out of 56) of the patients. LAT1 expression in IS patients was 76%, which were statistically higher compared to 36% in CS patients. When compared with LAT1 expression, no statistical difference was noted in terms of age, gender, body mass index (BMI), Cobb angle, and Risser grade. Meanwhile, the mean Slc7a5 expression in IS patients was determined to be significantly higher than that in CS patients. No significant correlation was observed between Slc7a5 expression and age, BMI, and Cobb angle. Conclusions LAT1 and Slc7a5 expression in IS and CS patients showed significant differences. These expressions were found to be not correlated with age, stature, and severity of the deformity.
Collapse
Affiliation(s)
- Satoru Demura
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Eiichi Hinoi
- Department of Bioactive Molecules, Gifu Pharmaceutical University, Gifu, Japan.,United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Noriaki Kawakami
- Department of Orthopaedic Surgery, Ichinomiya Nishi Hospital, Aichi, Japan
| | - Makoto Handa
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Noriaki Yokogawa
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Manami Hiraiwa
- Department of Bioactive Molecules, Gifu Pharmaceutical University, Gifu, Japan
| | - Satoshi Kato
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Kazuya Shinmura
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Takaki Shimizu
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Norihiro Oku
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Ryohei Annen
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Motoya Kobayashi
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Yohei Yamada
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Satoshi Nagatani
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| | - Takashi Iezaki
- Department of Bioactive Molecules, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Taniguchi
- Department of Orthopaedic Surgery, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery Graduate School of Medical Sciences Kanazawa University, Kanazawa, Japan
| |
Collapse
|
21
|
Xu Y, Wang Z, Wang Y, Huang Q, Ren C, Sun L, Wang Q, Li M, Liu H, Li Z, Zhang K, Ma T, Lu Y. Identification of differentially expressed autophagy genes associated with osteogenic differentiation in human bone marrow mesenchymal stem cells. Am J Transl Res 2022; 14:5326-5342. [PMID: 36105058 PMCID: PMC9452348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 07/05/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Mesenchymal stem cells derived from human tissues have been widely used for tissue regeneration because of their strong self-renewal capacity and multi-potential properties. Autophagy plays a vital role in maintaining bone homeostasis. However, the mechanism underlying this role for autophagy in the osteogenic differentiation of mesenchymal stem cells remains to be elucidated. METHODS Two microarray datasets were downloaded from the GEO database. Fourteen bone marrow mesenchymal stem cell samples comprising control and induction groups were selected to identify differentially expressed autophagy-related genes via multiple bioinformatics approaches, followed by functional analysis. Interactions among differentially expressed autophagy genes, miRNAs, and transcription factors were analyzed and visualized using Cytoscape software. The association between hub differentially expressed genes and autophagy was validated by qRT-PCR. RESULTS Ten autophagy-related genes (including VPS8, NDRG4, and CYBB) were identified as osteogenic hub genes. Correlation analysis revealed that CYBB was highly correlated with the sensitivity to multiple drugs, such as imexon, megestrol acetate, and isotretinoin. The regulatory network displayed a complex connection among miRNAs, transcription factors, and differentially expressed autophagy genes. Friends' analysis showed that NDRG4 was highly closely related to other hub genes (P < 0.05). Furthermore, NDRG4 expression was downregulated in the induction group (P < 0.01). NDRG4 was significantly correlated with infiltrating immune cells, including monocytes, eosinophils, type 17 T helper cells, neutrophils, activated CD8 T cells, and immature B cells. Levels of the 10 autophagy-related genes (including VPS8, NDRG4, and CYBB) were successfully validated based on in vitro experiments. CONCLUSION We identified candidate molecules to further investigate their functions in osteogenesis, providing novel insights into the role of autophagy in mesenchymal stem cell differentiation.
Collapse
Affiliation(s)
- Yibo Xu
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
- Bioinspired Engineering and Biomechanics Center (BEBC), School of Life Science and Technology, Xi′an Jiaotong UniversityXi’an 710049, Shaan’xi Province, China
| | - Zhimeng Wang
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
| | - Yakang Wang
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
| | - Qiang Huang
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
| | - Cheng Ren
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
- Bioinspired Engineering and Biomechanics Center (BEBC), School of Life Science and Technology, Xi′an Jiaotong UniversityXi’an 710049, Shaan’xi Province, China
| | - Liang Sun
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
| | - Qian Wang
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
| | - Ming Li
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
| | - Hongliang Liu
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
| | - Zhong Li
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
| | - Kun Zhang
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
| | - Teng Ma
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
- Bioinspired Engineering and Biomechanics Center (BEBC), School of Life Science and Technology, Xi′an Jiaotong UniversityXi’an 710049, Shaan’xi Province, China
| | - Yao Lu
- Department of Orthopaedic Surgery, Honghui Hospital, Xi’an Jiaotong UniversityXi’an 710054, Shaan’xi Province, China
- Bioinspired Engineering and Biomechanics Center (BEBC), School of Life Science and Technology, Xi′an Jiaotong UniversityXi’an 710049, Shaan’xi Province, China
| |
Collapse
|
22
|
Yamada T, Fukasawa K, Horie T, Kadota T, Lyu J, Tokumura K, Ochiai S, Iwahashi S, Suzuki A, Park G, Ueda R, Yamamoto M, Kitao T, Shirahase H, Ochi H, Sato S, Iezaki T, Hinoi E. The role of CDK8 in mesenchymal stem cells in controlling osteoclastogenesis and bone homeostasis. Stem Cell Reports 2022; 17:1576-1588. [PMID: 35777359 PMCID: PMC9287674 DOI: 10.1016/j.stemcr.2022.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 01/02/2023] Open
Abstract
Bone marrow mesenchymal stem cells (MSCs) are critical regulators of postnatal bone homeostasis. Osteoporosis is characterized by bone volume and strength deterioration, partly due to MSC dysfunction. Cyclin-dependent kinase 8 (CDK8) belongs to the transcription-related CDK family. Here, CDK8 in MSCs was identified as important for bone homeostasis. CDK8 level was increased in aged MSCs along with the association with aging-related signals. Mouse genetic studies revealed that CDK8 in MSCs plays a crucial role in bone resorption and homeostasis. Mechanistically, CDK8 in MSCs extrinsically controls osteoclastogenesis through the signal transducer and transcription 1 (STAT1)-receptor activator of the nuclear factor κ Β ligand (RANKL) axis. Moreover, aged MSCs have high osteoclastogenesis-supporting activity, partly through a CDK8-dependent manner. Finally, pharmacological inhibition of CDK8 effectively repressed MSC-dependent osteoclastogenesis and prevented ovariectomy-induced osteoclastic activation and bone loss. These findings highlight that the CDK8-STAT1-RANKL axis in MSCs could play a crucial role in bone resorption and homeostasis. Aging increases CDK8 expression level in MSCs and their progeny CDK8 in MSCs plays a crucial role in bone resorption and homeostasis CDK8 in MSCs extrinsically controls osteoclastogenesis through STAT1/RANKL axis CDK8 inhibitor prevents ovariectomy-induced osteoclastic activation and bone loss
Collapse
Affiliation(s)
- Takanori Yamada
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Kazuya Fukasawa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tetsuhiro Horie
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Takuya Kadota
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan; Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Kyoto, Japan
| | - Jiajun Lyu
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Kazuya Tokumura
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Shinsuke Ochiai
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Sayuki Iwahashi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Akane Suzuki
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Gyujin Park
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Rie Ueda
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Megumi Yamamoto
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Kyoto, Japan
| | - Tatsuya Kitao
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Kyoto, Japan
| | - Hiroaki Shirahase
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Kyoto, Japan
| | - Hiroki Ochi
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, Graduate School, Tokyo 113-8510, Japan
| | - Shingo Sato
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Takashi Iezaki
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Eiichi Hinoi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1196, Japan.
| |
Collapse
|
23
|
Go M, Shin E, Jang SY, Nam M, Hwang GS, Lee SY. BCAT1 promotes osteoclast maturation by regulating branched-chain amino acid metabolism. Exp Mol Med 2022; 54:825-833. [PMID: 35760874 PMCID: PMC9256685 DOI: 10.1038/s12276-022-00775-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/15/2022] [Accepted: 03/06/2022] [Indexed: 12/11/2022] Open
Abstract
Branched-chain aminotransferase 1 (BCAT1) transfers the amine group on branched-chain amino acids (BCAAs) to alpha-ketoglutarate. This generates glutamate along with alpha-keto acids that are eventually oxidized to provide the cell with energy. BCAT1 thus plays a critical role in sustaining BCAA concentrations and availability as an energy source. Osteoclasts have high metabolic needs during differentiation. When we assessed the levels of amino acids in bone marrow macrophages (BMMs) that were undergoing receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast differentiation, we found that the BCAA levels steadily increase during this process. In vitro analyses then showed that all three BCAAs but especially valine were needed for osteoclast maturation. Moreover, selective inhibition of BCAT1 with gabapentin significantly reduced osteoclast maturation. Expression of enzymatically dead BCAT1 also abrogated osteoclast maturation. Importantly, gabapentin inhibited lipopolysaccharide (LPS)-induced bone loss of calvaria in mice. These findings suggest that BCAT1 could serve as a therapeutic target that dampens osteoclast formation.
Collapse
Affiliation(s)
- Miyeon Go
- Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Eunji Shin
- Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seo Young Jang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03759, Republic of Korea.,Department of Chemistry & Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Miso Nam
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03759, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03759, Republic of Korea. .,Department of Chemistry & Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea.
| | - Soo Young Lee
- Department of Life Science, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
24
|
Therapeutic Potential for Intractable Asthma by Targeting L-Type Amino Acid Transporter 1. Biomolecules 2022; 12:biom12040553. [PMID: 35454142 PMCID: PMC9029068 DOI: 10.3390/biom12040553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 11/30/2022] Open
Abstract
Bronchial asthma is a chronic disease characterized by airway inflammation, obstruction, and hyperresponsiveness. CD4+ T cells, particularly T helper (Th) 2 cells, and their specific cytokines are important mediators in asthma pathogenesis. However, it has been established that Th subsets, other than Th2, as well as various cell types, including innate lymphoid cells (ILCs), significantly contribute to the development of allergic inflammation. These cells require facilitated amino acid uptake to ensure their full function upon activation. Emerging studies have suggested the potential of pharmacological inhibition of amino acid transporters to inhibit T cell activation and the application of this strategy for treating immunological and inflammatory disorders. In the present review, we explore the possibility of targeting L-type amino acid transporter (LAT) as a novel therapeutic approach for bronchial asthma, including its steroid-resistant endotypes.
Collapse
|
25
|
Hagen CM, Roth E, Graf TR, Verrey F, Graf R, Gupta A, Pellegrini G, Poncet N, Camargo SMR. Loss of LAT1 sex-dependently delays recovery after caerulein-induced acute pancreatitis. World J Gastroenterol 2022; 28:1024-1054. [PMID: 35431492 PMCID: PMC8968515 DOI: 10.3748/wjg.v28.i10.1024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/08/2021] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The expression of amino acid transporters is known to vary during acute pancreatitis (AP) except for LAT1 (slc7a5), the expression of which remains stable. LAT1 supports cell growth by importing leucine and thereby stimulates mammalian target of rapamycin (mTOR) activity, a phenomenon often observed in cancer cells. The mechanisms by which LAT1 influences physiological and pathophysiological processes and affects disease progression in the pancreas are not yet known.
AIM To evaluate the role of LAT1 in the development of and recovery from AP.
METHODS AP was induced with caerulein (cae) injections in female and male mice expressing LAT1 or after its knockout (LAT1 Cre/LoxP). The development of the initial AP injury and its recovery were followed for seven days after cae injections by daily measuring body weight, assessing microscopical tissue architecture, mRNA and protein expression, protein synthesis, and enzyme activity levels, as well as by testing the recruitment of immune cells by FACS and ELISA.
RESULTS The initial injury, evaluated by measurements of plasma amylase, lipase, and trypsin activity, as well as the gene expression of dedifferentiation markers, did not differ between the groups. However, early metabolic adaptations that support regeneration at later stages were blunted in LAT1 knockout mice. Especially in females, we observed less mTOR reactivation and dysfunctional autophagy. The later regeneration phase was clearly delayed in female LAT1 knockout mice, which did not regain normal expression of the pancreas-specific differentiation markers recombining binding protein suppressor of hairless-like protein (rbpjl) and basic helix-loop-helix family member A15 (mist1). Amylase mRNA and protein levels remained lower, and, strikingly, female LAT1 knockout mice presented signs of fibrosis lasting until day seven. In contrast, pancreas morphology had returned to normal in wild-type littermates.
CONCLUSION LAT1 supports the regeneration of acinar cells after AP. Female mice lacking LAT1 exhibited more pronounced alterations than male mice, indicating a sexual dimorphism of amino acid metabolism.
Collapse
Affiliation(s)
- Cristina M Hagen
- Institute of Physiology, University of Zurich, Zurich 8057, ZH, Switzerland
| | - Eva Roth
- Institute of Physiology, University of Zurich, Zurich 8057, ZH, Switzerland
| | - Theresia Reding Graf
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, Zurich University Hospital, Zurich 8091, ZH, Switzerland
| | - François Verrey
- Institute of Physiology, University of Zurich, Zurich 8057, ZH, Switzerland
| | - Rolf Graf
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, Zurich University Hospital, Zurich 8091, ZH, Switzerland
| | - Anurag Gupta
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, Zurich University Hospital, Zurich 8091, ZH, Switzerland
| | - Giovanni Pellegrini
- Institute of Veterinary Pathology, University of Zurich, Zurich 8057, ZH, Switzerland
| | - Nadège Poncet
- Institute of Physiology, University of Zurich, Zurich 8057, ZH, Switzerland
| | | |
Collapse
|
26
|
|
27
|
Ni F, Zhang T, Xiao W, Dong H, Gao J, Liu Y, Li J. IL-18-Mediated SLC7A5 Overexpression Enhances Osteogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells via the c-MYC Pathway. Front Cell Dev Biol 2021; 9:748831. [PMID: 34977008 PMCID: PMC8718798 DOI: 10.3389/fcell.2021.748831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: To investigate the role of IL-18 in the regulation of osteogenic differentiation in human bone marrow mesenchymal stem cells (hBMSCs). Methods: To assess whether IL-18 affects the osteogenic differentiation of hBMSCs through the c-MYC/SLC7A5 axis, IL-18 dose-response and time-course experiments were performed to evaluate its impact on osteogenic differentiation. To confirm osteogenic differentiation, alizarin red staining calcium measurement were performed. RT-qPCR and western blotting were used to determine the expression levels of bone-specific markers ALP, RUNX2, and BMP2, as well as those of SLC7A5 and c-MYC. Furthermore, SLC7A5 and c-MYC expression was evaluated via immunofluorescence. To elucidate the roles of SLC7A5 and c-MYC in osteoblast differentiation, cells were transfected with SLC7A5 or c-MYC siRNAs, or treated with the SLC7A5-specific inhibitor JPH203 and c-MYC-specific inhibitor 10058-F4, and the expression of SLC7A5, c-MYC, and bone-specific markers ALP, RUNX2, and BMP2 was assessed. Results: Our results demonstrated that IL-18 increased calcium deposition in hBMSCs, and upregulated the expression of SLC7A5, c-MYC, ALP, RUNX2, and BMP2. Silencing of SLC7A5 or c-MYC using siRNA reduced the expression of ALP, RUNX2, and BMP2, while IL-18 treatment partially reversed the inhibitory effect of siRNA. Similar results were obtained by treating hBMSCs with SLC7A5 and c-MYC specific inhibitors, leading to significant reduction of the osteogenesis effect of IL-18 on hBMSCs. Conclusion: In conclusion, our results indicate that IL-18 promotes the osteogenic differentiation of hBMSCs via the SLC7A5/c-MYC pathway and, therefore, may play an important role in fracture healing. These findings will provide new treatment strategies for delayed fracture healing after splenectomy.
Collapse
Affiliation(s)
- Feifei Ni
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Zhang
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanan Xiao
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hong Dong
- Liaoning Qifu Stem Cell Biotechnology Co, Ltd, Shenyang, China
| | - Jian Gao
- Liaoning Qifu Stem Cell Biotechnology Co, Ltd, Shenyang, China
| | - YaFeng Liu
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianjun Li
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Jianjun Li,
| |
Collapse
|
28
|
Hahn AK, Batushansky A, Rawle RA, Prado Lopes EB, June RK, Griffin TM. Effects of long-term exercise and a high-fat diet on synovial fluid metabolomics and joint structural phenotypes in mice: an integrated network analysis. Osteoarthritis Cartilage 2021; 29:1549-1563. [PMID: 34461226 PMCID: PMC8542629 DOI: 10.1016/j.joca.2021.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/18/2021] [Accepted: 08/04/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To explore how systemic factors that modify knee osteoarthritis risk are connected to 'whole-joint' structural changes by evaluating the effects of high-fat diet and wheel running exercise on synovial fluid (SF) metabolomics. METHODS Male mice were fed a defined control or high-fat (60% kcal fat) diet from 6 to 52 weeks of age, and half the animals were housed with running wheels from 26 to 52 weeks of age (n = 9-13 per group). Joint tissue structure and osteoarthritis pathology were evaluated by histology and micro-computed tomography. Systemic metabolic and inflammatory changes were evaluated by body composition, glucose tolerance testing, and serum biomarkers. SF metabolites were analyzed by high performance-liquid chromatography mass spectrometry. We built correlation-based network models to evaluate the connectivity between systemic and local metabolic biomarkers and osteoarthritis structural pathology within each experimental group. RESULTS High-fat diet caused moderate osteoarthritis, including cartilage pathology, synovitis and increased subchondral bone density. In contrast, voluntary exercise had a negligible effect on these joint structure components. 1,412 SF metabolite features were detected, with high-fat sedentary mice being the most distinct. Diet and activity uniquely altered SF metabolites attributed to amino acids, lipids, and steroids. Notably, high-fat diet increased network connections to systemic biomarkers such as interleukin-1β and glucose intolerance. In contrast, exercise increased local joint-level network connections, especially among subchondral bone features and SF metabolites. CONCLUSION Network mapping showed that obesity strengthened SF metabolite links to blood glucose and inflammation, whereas exercise strengthened SF metabolite links to subchondral bone structure.
Collapse
Affiliation(s)
- A K Hahn
- Molecular Biosciences Program, Montana State University, Bozeman, MT, 59717, USA; Department of Cell Biology & Neuroscience, Montana State University, Bozeman, MT, 59717, USA; Department of Biological and Environmental Sciences, Carroll College, Helena, MT, 59625, USA
| | - A Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104, USA
| | - R A Rawle
- Molecular Biosciences Program, Montana State University, Bozeman, MT, 59717, USA; Department of Microbiology & Immunology, Montana State University, Bozeman, MT, 59717, USA
| | - E B Prado Lopes
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104, USA
| | - R K June
- Molecular Biosciences Program, Montana State University, Bozeman, MT, 59717, USA; Department of Cell Biology & Neuroscience, Montana State University, Bozeman, MT, 59717, USA; Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, USA.
| | - T M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104, USA; Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
29
|
Suzuki A, Iwata J. Amino acid metabolism and autophagy in skeletal development and homeostasis. Bone 2021; 146:115881. [PMID: 33578033 PMCID: PMC8462526 DOI: 10.1016/j.bone.2021.115881] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/29/2020] [Accepted: 02/06/2021] [Indexed: 02/06/2023]
Abstract
Bone is an active organ that is continuously remodeled throughout life via formation and resorption; therefore, a fine-tuned bone (re)modeling is crucial for bone homeostasis and is closely connected with energy metabolism. Amino acids are essential for various cellular functions as well as an energy source, and their synthesis and catabolism (e.g., metabolism of carbohydrates and fatty acids) are regulated through numerous enzymatic cascades. In addition, the intracellular levels of amino acids are maintained by autophagy, a cellular recycling system for proteins and organelles; under nutrient deprivation conditions, autophagy is strongly induced to compensate for cellular demands and to restore the amino acid pool. Metabolites derived from amino acids are known to be precursors of bioactive molecules such as second messengers and neurotransmitters, which control various cellular processes, including cell proliferation, differentiation, and homeostasis. Thus, amino acid metabolism and autophagy are tightly and reciprocally regulated in our bodies. This review discusses the current knowledge and potential links between bone diseases and deficiencies in amino acid metabolism and autophagy.
Collapse
Affiliation(s)
- Akiko Suzuki
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
30
|
Ribet ABP, Ng PY, Pavlos NJ. Membrane Transport Proteins in Osteoclasts: The Ins and Outs. Front Cell Dev Biol 2021; 9:644986. [PMID: 33718388 PMCID: PMC7952445 DOI: 10.3389/fcell.2021.644986] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
During bone resorption, the osteoclast must sustain an extraordinarily low pH environment, withstand immense ionic pressures, and coordinate nutrient and waste exchange across its membrane to sustain its unique structural and functional polarity. To achieve this, osteoclasts are equipped with an elaborate set of membrane transport proteins (pumps, transporters and channels) that serve as molecular ‘gatekeepers’ to regulate the bilateral exchange of ions, amino acids, metabolites and macromolecules across the ruffled border and basolateral domains. Whereas the importance of the vacuolar-ATPase proton pump and chloride voltage-gated channel 7 in osteoclasts has long been established, comparatively little is known about the contributions of other membrane transport proteins, including those categorized as secondary active transporters. In this Special Issue review, we provide a contemporary update on the ‘ins and outs’ of membrane transport proteins implicated in osteoclast differentiation, function and bone homeostasis and discuss their therapeutic potential for the treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Amy B P Ribet
- Bone Biology and Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Pei Ying Ng
- Bone Biology and Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Nathan J Pavlos
- Bone Biology and Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|