1
|
Shen X, Zhang F, Tang C, Soković M, Mišić D, Xu H, Ye Y, Liu J. Advances in Sampling and Analytical Techniques for Single-Cell Metabolomics: Exploring Cellular Heterogeneity. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2025; 39:e10045. [PMID: 40223194 DOI: 10.1002/rcm.10045] [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: 01/07/2025] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
Single-cell metabolomics is an emerging and powerful technology that uncovers intercellular heterogeneity and reveals microenvironmental dynamics in both physiological and pathological conditions. This technology enables detailed observations of cellular interactions, providing valuable insights into processes such as aging, immune responses, and disease development. Despite significant advances, the need for detailed discussions on sampling and analytical methods in single-cell metabolomics continues to grow, with increasing focus on selecting the most suitable techniques for diverse research objectives. This review addresses these challenges by exploring key sampling and analytical strategies used in single-cell metabolomics. We focus on three main approaches: the capture and isolation of specific cell types, the precise aspiration of individual cells, and in situ mass spectrometry imaging. These methods are critically assessed to highlight strategies for achieving accurate metabolite detection at the single-cell level across diverse research applications.
Collapse
Affiliation(s)
- Xinxin Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fangyuan Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Chunping Tang
- China-Serbia "Belt and Road" Joint Laboratory for Natural Products and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Marina Soković
- China-Serbia "Belt and Road" Joint Laboratory for Natural Products and Drug Discovery, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Danijela Mišić
- China-Serbia "Belt and Road" Joint Laboratory for Natural Products and Drug Discovery, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Ye
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- China-Serbia "Belt and Road" Joint Laboratory for Natural Products and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jia Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
2
|
Lyu C, Tang Y, Shang FF. In situ mass spectrometry imaging reveals metabolite alterations in gray and white matter after spinal cord injury. Exp Ther Med 2025; 29:117. [PMID: 40297619 PMCID: PMC12035510 DOI: 10.3892/etm.2025.12867] [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: 11/04/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
It is known that spinal cord injury (SCI) causes metabolic disorders, such as disrupted lipid and amino acid metabolism. However, the in situ changes of metabolites in the spinal cord remain elusive. In the present study, adult Sprague-Dawley rats underwent spinal cord transection surgery. Ambient air flow-assisted desorption electrospray ionization mass spectrometry imaging was performed to identify the localization of metabolites in the white and gray matter and the ventral and dorsal horns within the spinal cord sections. The results revealed that 42 metabolites were specifically increased in the gray matter, and 90 metabolites were increased in the white matter compared with that in the sham group. In the ventral and dorsal horns, 86 and 103 metabolites, respectively, exhibited specific increases after injury. By contrast, numerous metabolites, especially lipids, were significantly decreased after SCI. Phosphatidylserine and phosphatidylethanolamine were mainly decreased in the gray matter, while cholesterol and ceramide were mainly decreased in the white matter. Specifically, phosphatidylethanolamine was detected at low levels in the dorsal horn following injury. However, the phosphatidylserine level decreased in the ventral horn. The functional enrichment of these metabolites via Kyoto Encyclopedia of Genes and Genomes analysis further confirmed the profile differences in the white and gray matter, as well as in the ventral and dorsal horns after SCI. These results provide valuable information on the metabolite profiles across various anatomical regions of the spinal cord following SCI, which may support the development of precise treatment strategies for patients.
Collapse
Affiliation(s)
- Chenghao Lyu
- International Medical College and College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yujing Tang
- International Medical College and College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
- College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, Heilongjiang 163319, P.R. China
| | - Fei-Fei Shang
- International Medical College and College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
3
|
Liu W, Hu X, Bao Z, Li Y, Zhang J, Yang S, Huang Y, Wang R, Wu J, Xu X, Sang Q, Di W, Lu H, Yin X, Qian K. Serum metabolic fingerprints encode functional biomarkers for ovarian cancer diagnosis: a large-scale cohort study. EBioMedicine 2025; 115:105706. [PMID: 40273469 PMCID: PMC12051638 DOI: 10.1016/j.ebiom.2025.105706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Ovarian cancer (OC) ranks as the most lethal gynaecological malignancy worldwide, with early diagnosis being crucial yet challenging. Current diagnostic methods like transvaginal ultrasound and blood biomarkers show limited sensitivity/specificity. This study aimed to identify and validate serum metabolic biomarkers for OC diagnosis using the largest cohort reported to date. METHODS We constructed a large-scale OC-associated cohort of 1432 subjects, including 662 OC, 563 benign ovarian disease, and 207 healthy control subjects, across retrospective (n = 1073) and set-aside validation (n = 359) cohorts. Serum metabolic fingerprints (SMFs) were recorded using nanoparticle-enhanced laser desorption/ionization mass spectrometry (NELDI-MS). A diagnostic panel was developed through machine learning of SMFs in the discovery cohort and validated in independent verification and set-aside validation cohorts. The identified metabolic biomarkers were further validated using liquid chromatography MS and their biological functions were assessed in OC cell lines. FINDINGS We identified a metabolic biomarker panel including glucose, histidine, pyrrole-2-carboxylic acid, and dihydrothymine. This panel achieved consistent areas under the curve (AUCs) of 0.87-0.89 for distinguishing between malignant and benign ovarian masses across all cohorts, and improved to AUCs of 0.95-0.99 when combined with risk of ovarian malignancy algorithm (ROMA). In vitro validation provided initial biological context for the metabolic alterations observed in our diagnostic panel. INTERPRETATION Our study established a reliable serum metabolic biomarker panel for OC diagnosis with potential clinical translations. The NELDI-MS based approach offers advantages of fast analytical speed (∼30 s/sample) and low cost (∼2-3 dollars/sample), making it suitable for large-scale clinical applications. FUNDING MOST (2021YFA0910100), NSFC (82421001, 823B2050, 824B2059, and 82173077), Medical-Engineering Joint Funds of Shanghai Jiao Tong University (YG2021GD02, YG2024ZD07, and YG2023ZD08), Shanghai Science and Technology Committee Project (23JC1403000), Shanghai Institutions of Higher Learning (2021-01-07-00-02-E00083), Shanghai Jiao Tong University Inner Mongolia Research Institute (2022XYJG0001-01-16), Sichuan Provincial Department of Science and Technology (2024YFHZ0176), Innovation Research Plan by the Shanghai Municipal Education Commission (ZXWF082101), Innovative Research Team of High-Level Local Universities in Shanghai (SHSMU-ZDCX20210700), Basic-Clinical Collaborative Innovation Project from Shanghai Immune Therapy Institute, Guangdong Basic and Applied Basic Research Foundation (2024A1515013255).
Collapse
Affiliation(s)
- Wanshan Liu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China
| | - Xiaoxiao Hu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China
| | - Zhouzhou Bao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China
| | - Yanyan Li
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China
| | - Juxiang Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China
| | - Shouzhi Yang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China
| | - Yida Huang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China
| | - Ruimin Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China
| | - Jiao Wu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China
| | - Xiaoyu Xu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China
| | - Qi Sang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China
| | - Wen Di
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China.
| | - Huaiwu Lu
- Department of Gynecologic Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| | - Xia Yin
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China.
| | - Kun Qian
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China; Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China.
| |
Collapse
|
4
|
Yang Q, Dai Y, Huang S, Liu B, Sun H, Zhou Y, Gong Y, Zhu F. MMEASE: enhanced analytical workflow for single-cell metabolomics. Nucleic Acids Res 2025:gkaf363. [PMID: 40308213 DOI: 10.1093/nar/gkaf363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/08/2025] [Accepted: 04/29/2025] [Indexed: 05/02/2025] Open
Abstract
Metabolomics is essential for providing an overview of what chemical processes are taking place. A clear shift from bulk metabolomics to single-cell metabolomics (SCM) is observed in current research, and an integral workflow enabling the analysis of SCM data is therefore in great demand. However, no such workflow has been available to date. Herein, MMEASE, previously designed for analyzing bulk metabolomic data, was therefore updated to its 2.0 version by developing the first comprehensive and in-depth workflow analyzing SCM data. First, it provided all sequential steps of modern SCM research (from SCM data processing, to cellular heterogeneity analysis, then to high-resolution metabolite annotation, and finally to cell-based biological interpretation). Second, compared with the existing tools, MMEASE 2.0 was superior by incorporating the widest variety of methods at every step of the SCM analyses. The originality and functionality of our MMEASE were extensively validated and explicitly described by case studies on benchmark data. All in all, MMEASE 2.0 was unique in accomplishing comprehensive and in-depth analyses of SCM data, which could be considered as an indispensable complement to the existing tools. Now, the latest version of MMEASE is freely accessible by all users at: https://idrblab.org/mmease/.
Collapse
Affiliation(s)
- Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yangbo Dai
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Shijie Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bing Liu
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Huaicheng Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuan Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yaguo Gong
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macao 999078, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
5
|
Liu Y, Zhu H, Zhang Q, Zhao Y. Clonal Analysis of Peripheral Blood T Cells in patients with Hashimoto's Thyroiditis at Different Stages using TCR Sequencing. Immunobiology 2025; 230:152890. [PMID: 40199058 DOI: 10.1016/j.imbio.2025.152890] [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: 09/21/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Hashimoto's Thyroiditis (HT) is a prevalent autoimmune disorder characterized by progressive thyroid damage mediated by T cells. The clonal diversity and expansion of T cells play a critical role in the pathogenesis of HT, but detailed insights into these characteristics at various disease stages are lacking. OBJECTIVE To analyze and compare the clonal diversity and T cell receptor (TCR) repertoire in peripheral blood T cells across different stages of Hashimoto's Thyroiditis using TCR sequencing. METHODS Peripheral blood samples from 19 HT patients at different disease stages and 4 healthy controls were collected. TCR sequencing was performed to assess T cell diversity and clonal expansion. Statistical analyses, including Shannon's entropy and Simpson's index, were employed to compare TCR diversity. Additionally, differential VJ gene usage and amino acid sequence diversity were analyzed. RESULTS The results revealed significant differences in TCR diversity between HT patients and healthy controls, with HT patients showing lower diversity. Notably, the frequency of hyperexpanded clones was higher in HT patients. Specific VJ genes exhibited differential usage patterns across disease stages, and a set of unique amino acid sequences was identified in each stage. CONCLUSION TCR sequencing highlights distinct clonal T cell expansions and shifts in VJ gene usage in HT patients, providing insights into the immune mechanisms driving disease progression.
Collapse
Affiliation(s)
- Yufeng Liu
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Huachao Zhu
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Qing Zhang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yanru Zhao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
6
|
Hu Q, Zhao W, Zhao Y, Li R, Zeng Y, Feng S, Di W, Shu W, Lou W, Wan J, Wang Y. Hollow Mesoporous Carbon Nanospheres/Ni Hybrids Aid in Metabolic Encoding for COVID-19 Recovery Assessment in Mothers and Fetuses. Anal Chem 2025; 97:6126-6135. [PMID: 40066735 DOI: 10.1021/acs.analchem.4c06790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Metabolite analysis of body fluids is an advanced method for disease diagnosis and status assessment. Laser desorption/ionization-mass spectrometry (LDI-MS) has been widely employed for metabolic analysis due to the fast detection speed and simple sample pretreatment. Here, we designed and synthesized hollow mesoporous carbon nanospheres anchored with Ni (HMCSs/Ni) to simultaneously enhance the ionization and thermal desorption processes of the LDI process owing to their hollow and mesoporous structure, large surface area, and abundant Ni-N bonds. Based on HMCSs/Ni, we built an LDI-MS platform that can be used for metabolic information extraction and achieved the rapid detection (about seconds per sample) of metabolic fingerprints in trace serum samples (∼0.1 μL) without complicated preprocessing procedures. Then, we conducted serum metabolic screening in a cohort of COVID-19-recovered pregnant women. The optimized machine learning model could distinguish recovered pregnant women from uninfected pregnant women based on metabolic features with an AUC value of 0.901. In addition, the model indicates that maternal COVID-19 infection does not significantly affect the metabolic fingerprints of the fetuses. Overall, our work shows the prospect of HMCSs/Ni-assisted LDI-MS in disease recovery assessment and metabolite analysis.
Collapse
Affiliation(s)
- Quan Hu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, P. R. China
| | - Weixiu Zhao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, China
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yinbing Zhao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, P. R. China
| | - Rongxin Li
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, P. R. China
| | - Yu Zeng
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, P. R. China
| | - Shuhuan Feng
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, China
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wen Di
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, China
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Weikang Shu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, P. R. China
| | - Weihua Lou
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, China
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jingjing Wan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, P. R. China
| | - You Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai 200127, China
- State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
7
|
Clarke HA, Hawkinson TR, Shedlock CJ, Medina T, Ribas RA, Wu L, Liu Z, Ma X, Xia Y, Huang Y, He X, Chang JE, Young LEA, Juras JA, Buoncristiani MD, James AN, Rushin A, Merritt ME, Mestas A, Lamb JF, Manauis EC, Austin GL, Chen L, Singh PK, Bian J, Vander Kooi CW, Evers BM, Brainson CF, Allison DB, Gentry MS, Sun RC. Glycogen drives tumour initiation and progression in lung adenocarcinoma. Nat Metab 2025:10.1038/s42255-025-01243-8. [PMID: 40069440 DOI: 10.1038/s42255-025-01243-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/12/2025] [Indexed: 03/17/2025]
Abstract
Lung adenocarcinoma (LUAD) is an aggressive cancer defined by oncogenic drivers and metabolic reprogramming. Here we leverage next-generation spatial screens to identify glycogen as a critical and previously underexplored oncogenic metabolite. High-throughput spatial analysis of human LUAD samples revealed that glycogen accumulation correlates with increased tumour grade and poor survival. Furthermore, we assessed the effect of increasing glycogen levels on LUAD via dietary intervention or via a genetic model. Approaches that increased glycogen levels provided compelling evidence that elevated glycogen substantially accelerates tumour progression, driving the formation of higher-grade tumours, while the genetic ablation of glycogen synthase effectively suppressed tumour growth. To further establish the connection between glycogen and cellular metabolism, we developed a multiplexed spatial technique to simultaneously assess glycogen and cellular metabolites, uncovering a direct relationship between glycogen levels and elevated central carbon metabolites essential for tumour growth. Our findings support the conclusion that glycogen accumulation drives LUAD cancer progression and provide a framework for integrating spatial metabolomics with translational models to uncover metabolic drivers of cancer.
Collapse
Affiliation(s)
- Harrison A Clarke
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Tara R Hawkinson
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Cameron J Shedlock
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Terrymar Medina
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Roberto A Ribas
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Lei Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Zizhen Liu
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Xin Ma
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Biostatistics College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yi Xia
- Department of Biostatistics College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yu Huang
- Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA
- Regenstrief Institute, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indianapolis, IN, USA
| | - Xing He
- Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA
- Regenstrief Institute, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indianapolis, IN, USA
| | - Josephine E Chang
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Lyndsay E A Young
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jelena A Juras
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | | | - Alexis N James
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Anna Rushin
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Matthew E Merritt
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Annette Mestas
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jessica F Lamb
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Elena C Manauis
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Grant L Austin
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Li Chen
- Department of Biostatistics College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Pankaj K Singh
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Jiang Bian
- Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA
- Regenstrief Institute, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indianapolis, IN, USA
| | - Craig W Vander Kooi
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - B Mark Evers
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Christine F Brainson
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Derek B Allison
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Matthew S Gentry
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA.
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA.
| | - Ramon C Sun
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA.
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA.
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
8
|
Li Z, Zhang S, Xiao Q, Shui S, Dong P, Jiang Y, Chen Y, Lan F, Peng Y, Ying B, Wu Y. Energy-Confinement 3D Flower-Shaped Cages for AI-Driven Decoding of Metabolic Fingerprints in Cardiovascular Disease Diagnosis. ACS NANO 2025; 19:6180-6194. [PMID: 39918943 DOI: 10.1021/acsnano.4c14656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Rapid and accurate detection plays a critical role in improving the survival and prognosis of patients with cardiovascular disease, but traditional detection methods are far from ideal for those with suspected conditions. Metabolite analysis based on nanomatrix-assisted laser desorption/ionization mass spectrometry (NMALDI-MS) is considered to be a promising technique for disease diagnosis. However, the performance of core nanomatrixes has limited its clinical application. In this study, we constructed 3D flower-shaped cages based on controllable structured metal-organic frameworks and iron oxide nanoparticles with low thermal conductivity and significant photothermal effects. The elongation of the incident light path through multilayer reflection significantly enhances the effective light absorption area of the nanomatrixes. Concurrently, the alternating layered structure confines the thermal energy, reducing thermal losses. Moreover, the 3D structure increases affinity sites, expanding the detection coverage. This approach effectively enhances the laser ionization and thermal desorption efficiency during the LDI process. We applied this technology to analyze the serum metabolomes of patients with myocardial infarction, heart failure, and heart failure combined with myocardial infarction, achieving cost-effective, high-throughput, highly accurate, and user-friendly detection of cardiovascular diseases. Subsequently, deep analysis of detected serum fingerprints via artificial intelligence models screens potential metabolic biomarkers, providing a new paradigm for the accurate diagnosis of cardiovascular diseases.
Collapse
Affiliation(s)
- Zhiyu Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Shuyu Zhang
- Machine Intelligence Lab, College of Computer Science, Sichuan University, Chengdu 610064, China
| | - Qianfeng Xiao
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Shaoxuan Shui
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Pingli Dong
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yujia Jiang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuanyuan Chen
- Machine Intelligence Lab, College of Computer Science, Sichuan University, Chengdu 610064, China
| | - Fang Lan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yong Peng
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
9
|
Yang C, Chen H, Wu Y, Shen X, Liu H, Liu T, Shen X, Xue R, Sun N, Deng C. Deep Learning-Enabled Rapid Metabolic Decoding of Small Extracellular Vesicles via Dual-Use Mass Spectroscopy Chip Array. Anal Chem 2025; 97:271-280. [PMID: 39711466 DOI: 10.1021/acs.analchem.4c04106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The increasing focus of small extracellular vesicles (sEVs) in liquid biopsy has created a significant demand for streamlined improvements in sEV isolation methods, efficient collection of high-quality sEV data, and powerful rapid analysis of large data sets. Herein, we develop a high-throughput dual-use mass spectroscopic chip array (DUMSCA) for the rapid isolation and detection of plasma sEVs. The DUMSCA realizes more than a 50% increase in speed compared to traditional method and confirms proficiency in robust storage, reuse, high-efficiency desorption/ionization, and metabolite quantification. With the collected metabolic data matrix of sEVs, a deep learning model achieves high-performance diagnosis of Crohn's disease. Furthermore, discovered biomarkers by feature sparsification and tandem mass spectrometry experiments also exhibited remarkable performance in diagnosis. This work demonstrates the rapidity and validity of DUMSCA for disease diagnosis, enabling the diagnosis of diseases without the necessity for prior knowledge and providing a high-throughput technology for sEV-based liquid biopsy that will empower its vigorous development.
Collapse
Affiliation(s)
- Chenyu Yang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Department of Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - He Chen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yun Wu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Department of Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Xiangguo Shen
- Department of Gastroenterology and Hepatology, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch Fudan University), Shanghai 200940, China
| | - Hongchun Liu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Gastroenterology and Hepatology, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch Fudan University), Shanghai 200940, China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chunhui Deng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Department of Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| |
Collapse
|
10
|
Watt SM, Roubelakis MG. Deciphering the Complexities of Adult Human Steady State and Stress-Induced Hematopoiesis: Progress and Challenges. Int J Mol Sci 2025; 26:671. [PMID: 39859383 PMCID: PMC11766050 DOI: 10.3390/ijms26020671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Human hematopoietic stem cells (HSCs) have traditionally been viewed as self-renewing, multipotent cells with enormous potential in sustaining essential steady state blood and immune cell production throughout life. Indeed, around 86% (1011-1012) of new cells generated daily in a healthy young human adult are of hematopoietic origin. Therapeutically, human HSCs have contributed to over 1.5 million hematopoietic cell transplants (HCTs) globally, making this the most successful regenerative therapy to date. We will commence this review by briefly highlighting selected key achievements (from 1868 to the end of the 20th century) that have contributed to this accomplishment. Much of our knowledge of hematopoiesis is based on small animal models that, despite their enormous importance, do not always recapitulate human hematopoiesis. Given this, we will critically review the progress and challenges faced in identifying adult human HSCs and tracing their lineage differentiation trajectories, referring to murine studies as needed. Moving forward and given that human hematopoiesis is dynamic and can readily adjust to a variety of stressors, we will then discuss recent research advances contributing to understanding (i) which HSPCs maintain daily steady state human hematopoiesis, (ii) where these are located, and (iii) which mechanisms come into play when homeostatic hematopoiesis switches to stress-induced or emergency hematopoiesis.
Collapse
Affiliation(s)
- Suzanne M. Watt
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9BQ, UK
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5005, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5001, Australia
| | - Maria G. Roubelakis
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece;
- Cell and Gene Therapy Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
| |
Collapse
|
11
|
Comazzetto S, Cassidy DL, DeVilbiss AW, Jeffery EC, Ottesen BR, Reyes AR, Paul A, Bansal S, Xie SZ, Muh S, Mathews TP, Chen B, Zhao Z, Morrison SJ. Ascorbate deficiency increases quiescence and self-renewal in hematopoietic stem cells and multipotent progenitors. Blood 2025; 145:114-126. [PMID: 39437548 PMCID: PMC11738029 DOI: 10.1182/blood.2024024769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/07/2024] [Accepted: 09/07/2024] [Indexed: 10/25/2024] Open
Abstract
ABSTRACT Ascorbate (vitamin C) limits hematopoietic stem cell (HSC) function and suppresses leukemia development, partly by promoting the function of the Tet2 tumor suppressor. In humans, ascorbate is obtained from the diet, whereas in mice, it is synthesized in the liver. In this study, we show that deletion of the Slc23a2 ascorbate transporter from hematopoietic cells depleted ascorbate to undetectable levels in HSCs and multipotent hematopoietic progenitors (MPPs) without altering the plasma ascorbate levels. Slc23a2 deficiency increased HSC reconstituting potential and self-renewal potential upon transplantation into irradiated mice. Slc23a2 deficiency also increased the reconstituting and self-renewal potentials of MPPs, conferring the ability to reconstitute irradiated mice long term. Slc23a2-deficient HSCs and MPPs divided much less frequently than control HSCs and MPPs. Increased self-renewal and reconstituting potential were observed particularly in quiescent Slc23a2-deficient HSCs and MPPs. The effect of Slc23a2 deficiency on MPP self-renewal was not mediated by reduced Tet2 function. Ascorbate thus regulates quiescence and restricts self-renewal potential in HSCs and MPPs such that ascorbate deficiency confers MPPs with long-term self-renewal potential.
Collapse
Affiliation(s)
- Stefano Comazzetto
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Daniel L Cassidy
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Andrew W DeVilbiss
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Elise C Jeffery
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Bethany R Ottesen
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Amanda R Reyes
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Animesh Paul
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Suraj Bansal
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Stephanie Z Xie
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sarah Muh
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Thomas P Mathews
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Brandon Chen
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Zhiyu Zhao
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Sean J Morrison
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
12
|
Ruffinatto L, Groult Y, Iacono J, Sarrazin S, de Laval B. Hematopoietic stem cell a reservoir of innate immune memory. Front Immunol 2024; 15:1491729. [PMID: 39720722 PMCID: PMC11666435 DOI: 10.3389/fimmu.2024.1491729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/30/2024] [Indexed: 12/26/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are a rare, long-lived and multipotent population that give rise to majority of blood cells and some tissue-resident immune cells. There is growing evidence that inflammatory stimuli can trigger persistent reprogramming in HSCs that enhances or inhibits the cellular functions of these HSCs and their progeny in response to subsequent infections. This newly discovered property makes HSCs a reservoir for innate immune memory. The molecular mechanisms underlying innate immune memory in HSCs are similar to those observed in innate immune cells, although their full elucidation is still pending. In this review, we examine the current state of knowledge on how an inflammatory response leads to reprogramming of HSCs. Understanding the full spectrum of consequences of reshaping early hematopoiesis is critical for assessing the potential benefits and risks under physiological and pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Bérengère de Laval
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut
National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
13
|
Herrera J, Bensussen A, García-Gómez ML, Garay-Arroyo A, Álvarez-Buylla ER. A system-level model reveals that transcriptional stochasticity is required for hematopoietic stem cell differentiation. NPJ Syst Biol Appl 2024; 10:145. [PMID: 39639033 PMCID: PMC11621455 DOI: 10.1038/s41540-024-00469-8] [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: 05/09/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
HSCs differentiation has been difficult to study experimentally due to the high number of components and interactions involved, as well as the impact of diverse physiological conditions. From a 200-node network, that was grounded on experimental data, we derived a 21-node regulatory network by collapsing linear pathways and retaining the functional feedback loops. This regulatory network core integrates key nodes and interactions underlying HSCs differentiation, including transcription factors, metabolic, and redox signaling pathways. We used Boolean, continuous, and stochastic dynamic models to simulate the hypoxic conditions of the HSCs niche, as well as the patterns and temporal sequences of HSCs transitions and differentiation. Our findings indicate that HSCs differentiation is a plastic process in which cell fates can transdifferentiate among themselves. Additionally, we found that cell heterogeneity is fundamental for HSCs differentiation. Lastly, we found that oxygen activates ROS production, inhibiting quiescence and promoting growth and differentiation pathways of HSCs.
Collapse
Affiliation(s)
- Joel Herrera
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Antonio Bensussen
- Departamento de Control Automático, Cinvestav-IPN, Ciudad de México, México
| | - Mónica L García-Gómez
- Theoretical Biology, Institute of Biodynamics and Biocomplexity; Experimental and Computational Plant Development, Institute of Environmental Biology, Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Adriana Garay-Arroyo
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Elena R Álvarez-Buylla
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, México.
| |
Collapse
|
14
|
Pan S, Yin L, Liu J, Tong J, Wang Z, Zhao J, Liu X, Chen Y, Miao J, Zhou Y, Zeng S, Xu T. Metabolomics-driven approaches for identifying therapeutic targets in drug discovery. MedComm (Beijing) 2024; 5:e792. [PMID: 39534557 PMCID: PMC11555024 DOI: 10.1002/mco2.792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Identification of therapeutic targets can directly elucidate the mechanism and effect of drug therapy, which is a central step in drug development. The disconnect between protein targets and phenotypes under complex mechanisms hampers comprehensive target understanding. Metabolomics, as a systems biology tool that captures phenotypic changes induced by exogenous compounds, has emerged as a valuable approach for target identification. A comprehensive overview was provided in this review to illustrate the principles and advantages of metabolomics, delving into the application of metabolomics in target identification. This review outlines various metabolomics-based methods, such as dose-response metabolomics, stable isotope-resolved metabolomics, and multiomics, which identify key enzymes and metabolic pathways affected by exogenous substances through dose-dependent metabolite-drug interactions. Emerging techniques, including single-cell metabolomics, artificial intelligence, and mass spectrometry imaging, are also explored for their potential to enhance target discovery. The review emphasizes metabolomics' critical role in advancing our understanding of disease mechanisms and accelerating targeted drug development, while acknowledging current challenges in the field.
Collapse
Affiliation(s)
- Shanshan Pan
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Luan Yin
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Jie Liu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Jie Tong
- Department of Radiology and Biomedical ImagingPET CenterYale School of MedicineNew HavenConnecticutUSA
| | - Zichuan Wang
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Jiahui Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Xuesong Liu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouZhejiangChina
| | - Yong Chen
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouZhejiangChina
| | - Jing Miao
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Yuan Zhou
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Su Zeng
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Tengfei Xu
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
15
|
Man CH, Li C, Xu X, Zhao M. Metabolic regulation in normal and leukemic stem cells. Trends Pharmacol Sci 2024; 45:919-930. [PMID: 39306527 DOI: 10.1016/j.tips.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 10/06/2024]
Abstract
Hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs) are crucial for ensuring hematopoietic homeostasis and driving leukemia progression, respectively. Recent research has revealed that metabolic adaptations significantly regulate the function and survival of these stem cells. In this review, we provide an overview of how metabolic pathways regulate oxidative and proteostatic stresses in HSCs during homeostasis and aging. Furthermore, we highlight targetable metabolic pathways and explore their interactions with epigenetics and the microenvironment in addressing the chemoresistance and immune evasion capacities of LSCs. The metabolic differences between HSCs and LSCs have profound implications for therapeutic strategies.
Collapse
Affiliation(s)
- Cheuk-Him Man
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Changzheng Li
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xi Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510030, China
| | - Meng Zhao
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
16
|
Liu Y, Wang R, Zhang C, Huang L, Chen J, Zeng Y, Chen H, Wang G, Qian K, Huang P. Automated Diagnosis and Phenotyping of Tuberculosis Using Serum Metabolic Fingerprints. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406233. [PMID: 39159075 PMCID: PMC11497029 DOI: 10.1002/advs.202406233] [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: 06/05/2024] [Revised: 07/23/2024] [Indexed: 08/21/2024]
Abstract
Tuberculosis (TB) stands as the second most fatal infectious disease after COVID-19, the effective treatment of which depends on accurate diagnosis and phenotyping. Metabolomics provides valuable insights into the identification of differential metabolites for disease diagnosis and phenotyping. However, TB diagnosis and phenotyping remain great challenges due to the lack of a satisfactory metabolic approach. Here, a metabolomics-based diagnostic method for rapid TB detection is reported. Serum metabolic fingerprints are examined via an automated nanoparticle-enhanced laser desorption/ionization mass spectrometry platform outstanding by its rapid detection speed (measured in seconds), minimal sample consumption (in nanoliters), and cost-effectiveness (approximately $3). A panel of 14 m z-1 features is identified as biomarkers for TB diagnosis and a panel of 4 m z-1 features for TB phenotyping. Based on the acquired biomarkers, TB metabolic models are constructed through advanced machine learning algorithms. The robust metabolic model yields a 97.8% (95% confidence interval (CI), 0.964-0.986) area under the curve (AUC) in TB diagnosis and an 85.7% (95% CI, 0.806-0.891) AUC in phenotyping. In this study, serum metabolic biomarker panels are revealed and develop an accurate metabolic tool with desirable diagnostic performance for TB diagnosis and phenotyping, which may expedite the effective implementation of the end-TB strategy.
Collapse
Affiliation(s)
- Yajing Liu
- Department of Ultrasound in MedicineThe Second Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Ruimin Wang
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Chao Zhang
- Department of Ultrasound in MedicineThe Second Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Lin Huang
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Jifan Chen
- Department of Ultrasound in MedicineThe Second Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Yiqing Zeng
- Department of Ultrasound in MedicineThe Second Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Hongjian Chen
- Post‐Doctoral Research CenterZhejiang SUKEAN Pharmaceutical Co., LtdHangzhou311225P. R. China
| | - Guowei Wang
- Department of Ultrasound in MedicineThe Second Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Kun Qian
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Pintong Huang
- Department of Ultrasound in MedicineThe Second Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhou310009P. R. China
- Research Center for Life Science and Human HealthBinjiang Institute of Zhejiang UniversityHangzhou310053P. R. China
| |
Collapse
|
17
|
Xiong M, Xiu Y, Long J, Zhao X, Wang Q, Yang H, Yu H, Bian L, Ju Y, Yin H, Hou Q, Liang F, Liu N, Chen F, Fan R, Sun Y, Zeng Y. Proteomics reveals dynamic metabolic changes in human hematopoietic stem progenitor cells from fetal to adulthood. Stem Cell Res Ther 2024; 15:303. [PMID: 39278906 PMCID: PMC11403967 DOI: 10.1186/s13287-024-03930-x] [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: 07/23/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Hematopoietic stem progenitor cells (HSPCs) undergo phenotypical and functional changes during their emergence and development. Although the molecular programs governing the development of human hematopoietic stem cells (HSCs) have been investigated broadly, the relationships between dynamic metabolic alterations and their functions remain poorly characterized. METHODS In this study, we comprehensively described the proteomics of HSPCs in the human fetal liver (FL), umbilical cord blood (UCB), and adult bone marrow (aBM). The metabolic state of human HSPCs was assessed via a Seahorse assay, RT‒PCR, and flow cytometry-based metabolic-related analysis. To investigate whether perturbing glutathione metabolism affects reactive oxygen species (ROS) production, the metabolic state, and the expansion of human HSPCs, HSPCs were treated with buthionine sulfoximine (BSO), an inhibitor of glutathione synthetase, and N-acetyl-L-cysteine (NAC). RESULTS We investigated the metabolomic landscape of human HSPCs from the fetal, perinatal, and adult developmental stages by in-depth quantitative proteomics and predicted a metabolic switch from the oxidative state to the glycolytic state during human HSPC development. Seahorse assays, mitochondrial activity, ROS level, glucose uptake, and protein synthesis rate analysis supported our findings. In addition, immune-related pathways and antigen presentation were upregulated in UCB or aBM HSPCs, indicating their functional maturation upon development. Glutathione-related metabolic perturbations resulted in distinct responses in human HSPCs and progenitors. Furthermore, the molecular and immunophenotypic differences between human HSPCs at different developmental stages were revealed at the protein level for the first time. CONCLUSION The metabolic landscape of human HSPCs at three developmental stages (FL, UCB, and aBM), combined with proteomics and functional validations, substantially extends our understanding of HSC metabolic regulation. These findings provide valuable resources for understanding human HSC function and development during fetal and adult life.
Collapse
Affiliation(s)
- Mingfang Xiong
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Yanyu Xiu
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Juan Long
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Xiao Zhao
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Qianqian Wang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Haoyu Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Hang Yu
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Lihong Bian
- Department of Gynecology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yan Ju
- Department of Gynecology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Hongyu Yin
- Department of Gynecology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Qingxiang Hou
- Department of Obstetrics and Gynecology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Fei Liang
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Nan Liu
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Fudong Chen
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Ruiwen Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Yuying Sun
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| | - Yang Zeng
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China.
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China.
| |
Collapse
|
18
|
Chen X, Yu W, Zhao Y, Ji Y, Qi Z, Guan Y, Wan J, Hao Y. Diagnosis of epilepsy by machine learning of high-performance plasma metabolic fingerprinting. Talanta 2024; 277:126328. [PMID: 38824860 DOI: 10.1016/j.talanta.2024.126328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/18/2024] [Accepted: 05/25/2024] [Indexed: 06/04/2024]
Abstract
Epilepsy is a chronic neurological disorder that causes a major threat to public health and the burden of disease worldwide. High-performance diagnostic tools for epilepsy need to be developed to improve diagnostic accuracy and efficiency while still missing. Herein, we utilized nanoparticle-enhanced laser desorption/ionization mass spectrometry (NELDI MS) to acquire plasma metabolic fingerprints (PMFs) from epileptic and healthy individuals for timely and accurate screening of epilepsy. The NELDI MS enabled high detection speed (∼30 s per sample), high throughput (up to 384 samples per run), and favorable reproducibility (coefficients of variation <15 %), acquiring high-performed PMFs. We next constructed an epilepsy diagnostic model by machine learning of PMFs, achieving desirable diagnostic capability with the area under the curve (AUC) value of 0.941 for the validation set. Furthermore, four metabolites were identified as a diagnostic biomarker panel for epilepsy, with an AUC value of 0.812-0.860. Our approach provides a high-performed and high-throughput platform for epileptic diagnostics, promoting the development of metabolic diagnostic tools in precision medicine.
Collapse
Affiliation(s)
- Xiaonan Chen
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, PR China
| | - Wendi Yu
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Yinbing Zhao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, PR China
| | - Yuxi Ji
- School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, PR China
| | - Ziheng Qi
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, PR China
| | - Yangtai Guan
- Department of Neurology, Punan Branch of Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200125, PR China.
| | - Jingjing Wan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, PR China.
| | - Yong Hao
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China.
| |
Collapse
|
19
|
Morganti C, Bonora M, Ito K. Metabolism and HSC fate: what NADPH is made for. Trends Cell Biol 2024:S0962-8924(24)00141-7. [PMID: 39054107 PMCID: PMC11757803 DOI: 10.1016/j.tcb.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Mitochondrial metabolism plays a central role in the regulation of hematopoietic stem cell (HSC) biology. Mitochondrial fatty acid oxidation (FAO) is pivotal in controlling HSC self-renewal and differentiation. Herein, we discuss recent evidence suggesting that NADPH generated in the mitochondria can influence the fate of HSCs. Although NADPH has multiple functions, HSCs show high levels of NADPH that are preferentially used for cholesterol biosynthesis. Endogenous cholesterol supports the biogenesis of extracellular vesicles (EVs), which are essential for maintaining HSC properties. We also highlight the significance of EVs in hematopoiesis through autocrine signaling. Elucidating the mitochondrial NADPH-cholesterol axis as part of the metabolic requirements of healthy HSCs will facilitate the development of new therapies for hematological disorders.
Collapse
Affiliation(s)
- Claudia Morganti
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| | - Massimo Bonora
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| |
Collapse
|
20
|
Chen X, Wang Y, Pei C, Li R, Shu W, Qi Z, Zhao Y, Wang Y, Lin Y, Zhao L, Peng D, Wan J. Vacancy-Driven High-Performance Metabolic Assay for Diagnosis and Therapeutic Evaluation of Depression. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312755. [PMID: 38692290 DOI: 10.1002/adma.202312755] [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: 11/27/2023] [Revised: 03/31/2024] [Indexed: 05/03/2024]
Abstract
Depression is one of the most common mental illnesses and is a well-known risk factor for suicide, characterized by low overall efficacy (<50%) and high relapse rate (40%). A rapid and objective approach for screening and prognosis of depression is highly desirable but still awaits further development. Herein, a high-performance metabolite-based assay to aid the diagnosis and therapeutic evaluation of depression by developing a vacancy-engineered cobalt oxide (Vo-Co3O4) assisted laser desorption/ionization mass spectrometer platform is presented. The easy-prepared nanoparticles with optimal vacancy achieve a considerable signal enhancement, characterized by favorable charge transfer and increased photothermal conversion. The optimized Vo-Co3O4 allows for a direct and robust record of plasma metabolic fingerprints (PMFs). Through machine learning of PMFs, high-performance depression diagnosis is achieved, with the areas under the curve (AUC) of 0.941-0.980 and an accuracy of over 92%. Furthermore, a simplified diagnostic panel for depression is established, with a desirable AUC value of 0.933. Finally, proline levels are quantified in a follow-up cohort of depressive patients, highlighting the potential of metabolite quantification in the therapeutic evaluation of depression. This work promotes the progression of advanced matrixes and brings insights into the management of depression.
Collapse
Affiliation(s)
- Xiaonan Chen
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Yun Wang
- Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Congcong Pei
- School of Chemistry, Zhengzhou University, Zhengzhou, 450001, P. R. China
- Center of Advanced Analysis and Gene Sequencing, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Rongxin Li
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Weikang Shu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Ziheng Qi
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Yinbing Zhao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Yanhui Wang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Yingying Lin
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Liang Zhao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Daihui Peng
- Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Jingjing Wan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| |
Collapse
|
21
|
Zhong ZJ, Ling J, Yao ZP, Liu LF, Zheng JY, Xin GZ. Targeted Quantification of Glutathione/Arginine Redox Metabolism Based on a Novel Paired Mass Spectrometry Probe Approach for the Functional Assessment of Redox Status. Anal Chem 2024; 96:9885-9893. [PMID: 38848670 DOI: 10.1021/acs.analchem.4c00732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Glutathione (GSH) redox control and arginine metabolism are critical in regulating the physiological response to injury and oxidative stress. Quantification assessment of the GSH/arginine redox metabolism supports monitoring metabolic pathway shifts during pathological processes and their linkages to redox regulation. However, assessing the redox status of organisms with complex matrices is challenging, and single redox molecule analysis may not be accurate for interrogating the redox status in cells and in vivo. Herein, guided by a paired derivatization strategy, we present a new ultraperformance liquid chromatography tandem mass spectrometry (UPLC-MS/MS)-based approach for the functional assessment of biological redox status. Two structurally analogous probes, 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate (AQC) and newly synthesized 2-methyl-6-aminoquinolyl-N-hydroxysuccinimidyl carbamate (MeAQC), were set for paired derivatization. The developed approach was successfully applied to LPS-stimulated RAW 264.7 cells and HDM-induced asthma mice to obtain quantitative information on GSH/arginine redox metabolism. The results suggest that the redox status was remarkably altered upon LPS and HDM stimulation. We expect that this approach will be of good use in a clinical biomarker assay and potential drug screening associated with redox metabolism, oxidative damage, and redox signaling.
Collapse
Affiliation(s)
- Zhu-Jun Zhong
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jing Ling
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhong-Ping Yao
- State Key Laboratory of Chemical Biology and Drug Discovery, Research Centre for Chinese Medicine Innovation, Food Safety and Technology Research Centre and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, China
| | - Li-Fang Liu
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Yi Zheng
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Gui-Zhong Xin
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
22
|
Ding C, Zhu Y, Huo Z, Yang S, Zhou Y, Yiming A, Chen W, Liu S, Qian K, Huang L. Pt/NiFe-LDH hybrids for quantification and qualification of polyphenols. Mater Today Bio 2024; 26:101047. [PMID: 38638703 PMCID: PMC11025000 DOI: 10.1016/j.mtbio.2024.101047] [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: 02/01/2024] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024] Open
Abstract
Polyphenols with antioxidant properties are of significant interest in medical and pharmaceutical applications. Given the diverse range of activities of polyphenols in vivo, accurate detection of these compounds plays a crucial role in nutritional surveillance and pharmaceutical development. Yet, the efficient quantitation of polyphenol contents and qualification of monomer compositions present a notable challenge when studying polyphenol bioavailability. In this study, platinum-modified nickel-iron layered double hydroxide (Pt/NiFe-LDH hybrids) were designed to mimic peroxidases for colorimetric analysis and act as enhanced matrices for laser desorption/ionization mass spectrometry (LDI MS) to quantify and qualify polyphenols. The hybrids exhibited an enzymatic activity of 33.472 U/mg for colorimetric assays, facilitating the rapid and direct quantitation of total tea polyphenols within approximately 1 min. Additionally, the heterogeneous structure and exposed hydroxyl groups on the hybrid surface contributed to photoelectric enhancement and in-situ enrichment of polyphenols in LDI MS. This study introduces an innovative approach to detect polyphenols using advanced materials, potentially inspiring the future development and applications of other photoactive nanomaterials.
Collapse
Affiliation(s)
- Chunmeng Ding
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Yuexing Zhu
- Second Military Medical University, Changhai Hospital, Department of Lab Diagnostics, Shanghai, 200433, P. R. China
| | - Zhiyuan Huo
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Shouzhi Yang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Yan Zhou
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Ayizekeranmu Yiming
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Wei Chen
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Shanrong Liu
- Second Military Medical University, Changhai Hospital, Department of Lab Diagnostics, Shanghai, 200433, P. R. China
| | - Kun Qian
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Lin Huang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| |
Collapse
|
23
|
Hilovsky D, Hartsell J, Young JD, Liu X. Stable Isotope Tracing Analysis in Cancer Research: Advancements and Challenges in Identifying Dysregulated Cancer Metabolism and Treatment Strategies. Metabolites 2024; 14:318. [PMID: 38921453 PMCID: PMC11205609 DOI: 10.3390/metabo14060318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Metabolic reprogramming is a hallmark of cancer, driving the development of therapies targeting cancer metabolism. Stable isotope tracing has emerged as a widely adopted tool for monitoring cancer metabolism both in vitro and in vivo. Advances in instrumentation and the development of new tracers, metabolite databases, and data analysis tools have expanded the scope of cancer metabolism studies across these scales. In this review, we explore the latest advancements in metabolic analysis, spanning from experimental design in stable isotope-labeling metabolomics to sophisticated data analysis techniques. We highlight successful applications in cancer research, particularly focusing on ongoing clinical trials utilizing stable isotope tracing to characterize disease progression, treatment responses, and potential mechanisms of resistance to anticancer therapies. Furthermore, we outline key challenges and discuss potential strategies to address them, aiming to enhance our understanding of the biochemical basis of cancer metabolism.
Collapse
Affiliation(s)
- Dalton Hilovsky
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695, USA; (D.H.); (J.H.)
| | - Joshua Hartsell
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695, USA; (D.H.); (J.H.)
| | - Jamey D. Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37212, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37212, USA
| | - Xiaojing Liu
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695, USA; (D.H.); (J.H.)
| |
Collapse
|
24
|
Li Z, Peng W, Zhou J, Shui S, Liu Y, Li T, Zhan X, Chen Y, Lan F, Ying B, Wu Y. Multidimensional Interactive Cascading Nanochips for Detection of Multiple Liver Diseases via Precise Metabolite Profiling. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312799. [PMID: 38263756 DOI: 10.1002/adma.202312799] [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: 11/27/2023] [Revised: 01/11/2024] [Indexed: 01/25/2024]
Abstract
It is challenging to detect and differentiate multiple diseases with high complexity/similarity from the same organ. Metabolic analysis based on nanomatrix-assisted laser desorption/ionization mass spectrometry (NMALDI-MS) is a promising platform for disease diagnosis, while the enhanced property of its core nanomatrix materials has plenty of room for improvement. Herein, a multidimensional interactive cascade nanochip composed of iron oxide nanoparticles (FeNPs)/MXene/gold nanoparticles (AuNPs), IMG, is reported for serum metabolic profiling to achieve high-throughput detection of multiple liver diseases. MXene serves as a multi-binding site and an electron-hole source for ionization during NMALDI-MS analysis. Introduction of AuNPs with surface plasmon resonance (SPR) properties facilitates surface charge accumulation and rapid energy conversion. FeNPs are integrated into the MXene/Au nanocomposite to sharply reduce the thermal conductivity of the nanochip with negligible heat loss for strong thermally-driven desorption, and construct a multi-interaction proton transport pathway with MXene and AuNPs for strong ionization. Analysis of these enhanced serum fingerprint signals detected from the IMG nanochip through a neural network model results in differentiation of multiple liver diseases via a single pass and revelation of potential metabolic biomarkers. The promising method can rapidly and accurately screen various liver diseases, thus allowing timely treatment of liver diseases.
Collapse
Affiliation(s)
- Zhiyu Li
- National Engineering Research Center for Biomaterials, School of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Weili Peng
- Machine Intelligence Lab, College of Computer Science, Sichuan University, Chengdu, 610064, China
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Shaoxuan Shui
- National Engineering Research Center for Biomaterials, School of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Yicheng Liu
- National Engineering Research Center for Biomaterials, School of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Tan Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Xiaohui Zhan
- National Engineering Research Center for Biomaterials, School of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Yuanyuan Chen
- Machine Intelligence Lab, College of Computer Science, Sichuan University, Chengdu, 610064, China
| | - Fang Lan
- National Engineering Research Center for Biomaterials, School of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, School of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
25
|
Xu J, Fei P, Simon DW, Morowitz MJ, Mehta PA, Du W. Crosstalk between DNA Damage Repair and Metabolic Regulation in Hematopoietic Stem Cells. Cells 2024; 13:733. [PMID: 38727270 PMCID: PMC11083014 DOI: 10.3390/cells13090733] [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: 03/23/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
Self-renewal and differentiation are two characteristics of hematopoietic stem cells (HSCs). Under steady physiological conditions, most primitive HSCs remain quiescent in the bone marrow (BM). They respond to different stimuli to refresh the blood system. The transition from quiescence to activation is accompanied by major changes in metabolism, a fundamental cellular process in living organisms that produces or consumes energy. Cellular metabolism is now considered to be a key regulator of HSC maintenance. Interestingly, HSCs possess a distinct metabolic profile with a preference for glycolysis rather than oxidative phosphorylation (OXPHOS) for energy production. Byproducts from the cellular metabolism can also damage DNA. To counteract such insults, mammalian cells have evolved a complex and efficient DNA damage repair (DDR) system to eliminate various DNA lesions and guard genomic stability. Given the enormous regenerative potential coupled with the lifetime persistence of HSCs, tight control of HSC genome stability is essential. The intersection of DDR and the HSC metabolism has recently emerged as an area of intense research interest, unraveling the profound connections between genomic stability and cellular energetics. In this brief review, we delve into the interplay between DDR deficiency and the metabolic reprogramming of HSCs, shedding light on the dynamic relationship that governs the fate and functionality of these remarkable stem cells. Understanding the crosstalk between DDR and the cellular metabolism will open a new avenue of research designed to target these interacting pathways for improving HSC function and treating hematologic disorders.
Collapse
Affiliation(s)
- Jian Xu
- Division of Hematology and Oncology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Peiwen Fei
- Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI 96812, USA
| | - Dennis W. Simon
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael J. Morowitz
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Parinda A. Mehta
- Division of Blood and Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Wei Du
- Division of Hematology and Oncology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
26
|
Comazzetto S, Cassidy DL, DeVilbiss AW, Jeffery EC, Ottesen BR, Reyes AR, Muh S, Mathews TP, Chen B, Zhao Z, Morrison SJ. Ascorbate depletion increases quiescence and self-renewal potential in hematopoietic stem cells and multipotent progenitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587574. [PMID: 38617357 PMCID: PMC11014518 DOI: 10.1101/2024.04.01.587574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Ascorbate (vitamin C) limits hematopoietic stem cell (HSC) function and suppresses leukemia development by promoting the function of the Tet2 tumor suppressor. In humans, ascorbate is obtained from the diet while in mice it is synthesized in the liver. In this study, we show that deletion of the Slc23a2 ascorbate transporter severely depleted ascorbate from hematopoietic cells. Slc23a2 deficiency increased HSC reconstituting potential and self-renewal potential upon transplantation into irradiated mice. Slc23a2 deficiency also increased the reconstituting and self-renewal potential of multipotent hematopoietic progenitors (MPPs), conferring the ability to long-term reconstitute irradiated mice. Slc23a2-deficient HSCs and MPPs divided much less frequently than control HSCs and MPPs. Increased self-renewal and reconstituting potential were observed particularly in quiescent Slc23a2-deficient HSCs and MPPs. The effect of Slc23a2 deficiency on MPP self-renewal was not mediated by reduced Tet2 function. Ascorbate thus regulates quiescence and restricts self-renewal potential in HSCs and MPPs such that ascorbate depletion confers MPPs with long-term self-renewal potential.
Collapse
Affiliation(s)
- Stefano Comazzetto
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel L. Cassidy
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrew W. DeVilbiss
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elise C. Jeffery
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bethany R. Ottesen
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Amanda R. Reyes
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sarah Muh
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas P. Mathews
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Brandon Chen
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean J. Morrison
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
27
|
Ai Y, Peng K, Li C, Zhang J, Wang G, Wang B, Huang E. Assessment of Reference Genes Stability in Cortical Bone of Obese and Diabetic Mice. Diabetes Metab Syndr Obes 2024; 17:1081-1091. [PMID: 38455760 PMCID: PMC10917645 DOI: 10.2147/dmso.s453458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/15/2024] [Indexed: 03/09/2024] Open
Abstract
Introduction Bone, a pivotal structural organ, is susceptible to disorders with profound health implications. The investigation of gene expression in bone tissue is imperative, particularly within the context of metabolic diseases such as obesity and diabetes that augment the susceptibility to bone fractures. The objective of this study is to identify a set of internal control genes for the analysis of gene expression. Methods This study employs reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) to assess gene expression in bone tissue. We selected fourteen housekeeping genes and assessed their stability in the cortical bone of mouse models for obesity and diabetes using four well-established algorithms (GeNorm, BestKeeper, NormFinder, and the comparative Delta Ct method). Results and Conclusion We identified Rpl13a as the mostly stably expressed reference gene in cortical bone tissue from mouse models of obesity and diabetes (db/db), while Gapdh was found to be the most stable reference gene in another diabetes model, KKAy mice. Additionally, Ef1a, Ppia, Rplp0, and Rpl22 were identified as alternative genes suitable for normalizing gene expression in cortical bone from obesity and diabetes mouse models. These findings enhance RT-qPCR accuracy and reliability, offering a strategic guide to select reference gene for studying bone tissue gene expression in metabolic disorders.
Collapse
Affiliation(s)
- Yuanli Ai
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Kun Peng
- Chongqing Medical and Pharmaceutical College, Chongqing, People’s Republic of China
| | - Chunli Li
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jun Zhang
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Gang Wang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Bin Wang
- Institute of Life Sciences, College of Basic Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Enyi Huang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education College of Stomatology, College of Stomatology, Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|