1
|
Gorman BL, Shafer CC, Ragi N, Sharma K, Neumann EK, Anderton CR. Imaging and spatially resolved mass spectrometry applications in nephrology. Nat Rev Nephrol 2025; 21:399-416. [PMID: 40148534 DOI: 10.1038/s41581-025-00946-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
The application of spatially resolved mass spectrometry (MS) and MS imaging approaches for studying biomolecular processes in the kidney is rapidly growing. These powerful methods, which enable label-free and multiplexed detection of many molecular classes across omics domains (including metabolites, drugs, proteins and protein post-translational modifications), are beginning to reveal new molecular insights related to kidney health and disease. The complexity of the kidney often necessitates multiple scales of analysis for interrogating biofluids, whole organs, functional tissue units, single cells and subcellular compartments. Various MS methods can generate omics data across these spatial domains and facilitate both basic science and pathological assessment of the kidney. Optimal processes related to sample preparation and handling for different MS applications are rapidly evolving. Emerging technology and methods, improvement of spatial resolution, broader molecular characterization, multimodal and multiomics approaches and the use of machine learning and artificial intelligence approaches promise to make these applications even more valuable in the field of nephology. Overall, spatially resolved MS and MS imaging methods have the potential to fill much of the omics gap in systems biology analysis of the kidney and provide functional outputs that cannot be obtained using genomics and transcriptomic methods.
Collapse
Affiliation(s)
- Brittney L Gorman
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Catelynn C Shafer
- Department of Chemistry, University of California, Davis, Davis, CA, 95695, USA
| | - Nagarjunachary Ragi
- Center for Precision Medicine, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Kumar Sharma
- Center for Precision Medicine, The University of Texas Health San Antonio, San Antonio, TX, USA
- Division of Nephrology, Department of Medicine, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Elizabeth K Neumann
- Department of Chemistry, University of California, Davis, Davis, CA, 95695, USA
| | - Christopher R Anderton
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
2
|
Kumar R, Romano JD, Ritchie MD. Network-based analyses of multiomics data in biomedicine. BioData Min 2025; 18:37. [PMID: 40426270 PMCID: PMC12117783 DOI: 10.1186/s13040-025-00452-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Network representations of data are designed to encode relationships between concepts as sets of edges between nodes. Human biology is inherently complex and is represented by data that often exists in a hierarchical nature. One canonical example is the relationship that exists within and between various -omics datasets, including genomics, transcriptomics, and proteomics, among others. Encoding such data in a network-based or graph-based representation allows the explicit incorporation of such relationships into various biomedical big data tasks, including (but not limited to) disease subtyping, interaction prediction, biomarker identification, and patient classification. This review will present various existing approaches in using network representations and analysis of data in multiomics in the framework of deep learning and machine learning approaches, subdivided into supervised and unsupervised approaches, to identify benefits and drawbacks of various approaches as well as the possible next steps for the field.
Collapse
Affiliation(s)
- Rachit Kumar
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph D Romano
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Division of Informatics, Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Parvez RK, Kim DK, Csipán RL, Guo J, Zeng Z, Zhang CC, Li Z, McMahon AP. Dmrt2 and Hmx2 direct intercalated cell diversity in the mammalian kidney through antagonistic and supporting regulatory processes. Proc Natl Acad Sci U S A 2025; 122:e2418471122. [PMID: 40354537 PMCID: PMC12107187 DOI: 10.1073/pnas.2418471122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/15/2025] [Indexed: 05/14/2025] Open
Abstract
Intercalated cells (ICs) in the mammalian kidney regulate circulatory pH through IC subtype-restricted actions of bicarbonate transporters: pH is elevated by Slc4a1 restricted to type A-ICs (A-ICs) and depressed by Slc26a4 in type B-IC (B-ICs). NonA-nonB-ICs (nA/nB-ICs) also produce Slc26a4 though their function is unclear. Though both nephron and ureteric progenitor lineages generate A-ICs, the former also generates nA/nB-ICs and the latter B-ICs. Lineage and cell type restricted transporter gene expression in the mouse and human kidney is preceded by expression of the transcriptional regulators Dmrt2/DMRT2 in A-ICs, and either, or both, Hmx2/HMX2 and Hmx3/HMX3 in B- and nA/nB ICs. CRISPR/Cas9-directed removal of Dmrt2 and the linked Hmx2/Hmx3 genes resulted in IC-subtype switching. A-ICs adopted an Hmx2+/Slc26a4+ B-IC cell fate on Dmrt2 removal while B-ICs initiated a Dmrt2+/Slc4a1+ A-IC program on Hmx2/Hmx3 removal. Triple knockout of Dmrt2, Hmx2, and Hmx3 resulted in hybrid ICs expressing both Slc4a1 and Slc26a4. Thus, restricted expression of these regulators is essential for specifying IC subtypes. To explore these mechanisms, Hmx2 and Dmrt2 were activated ectopically in ureteric organoid cultures. Introduction of Foxi1-a pan determinant of ICs-activated early Dmrt2+ A-IC development while cointroduction of Hmx2 silenced Foxi1-dependent Dmrt2 expression and led to an upregulation of Slc26a4. In contrast, coexpression of Foxi1 and Dmrt2 upregulated Slc4a1. These data support a model in which mutually repressive interactions between Dmrt2 and Hmx2/3 establish distinct IC identities and ongoing activity of these factors supports gene regulatory programs specific to each IC subtype.
Collapse
Affiliation(s)
- Riana K. Parvez
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA90033
| | - Doh Kyung Kim
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA90033
| | - Réka L. Csipán
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA90033
| | - Jinjin Guo
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA90033
| | - Zipeng Zeng
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA90033
- University of Southern California/University Kidney Research Organization Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Chennan C. Zhang
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA90033
- University of Southern California/University Kidney Research Organization Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Zhongwei Li
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA90033
- University of Southern California/University Kidney Research Organization Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Andrew P. McMahon
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA90033
| |
Collapse
|
4
|
Fung AA, Li Z, Boote C, Markov P, Gaut JP, Jain S, Shi L. Label-free multimodal optical biopsy reveals biomolecular and morphological features of diabetic kidney tissue in 2D and 3D. Nat Commun 2025; 16:4509. [PMID: 40374604 PMCID: PMC12081717 DOI: 10.1038/s41467-025-59163-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/14/2025] [Indexed: 05/17/2025] Open
Abstract
Kidney disease, the ninth leading cause of death in the United States, suffers from poor diagnostic efficiency (10%). Traditional biopsies use molecular reagents to enhance diagnostic power but are limited by overlapping spatial and chromatic signals, product quality variability, and additional processing. To address these challenges without disrupting routine diagnostics, we implement label-free imaging modalities-stimulated Raman scattering (SRS), second harmonic generation (SHG), and two-photon fluorescence (TPF)-within a single setup. We identify morphological, lipidomic, and metabolic biomarkers in control and diabetic kidney samples at subcellular resolution. Label-free Stimulated Raman Histology (SRH) reveals distinct collagen morphology, mesangial-glomerular volumes, lipid saturation, redox status, and lipid-protein concentrations previously unrecognized in kidney diseases. Using the same tissue section enhances diagnostic value without compromising limited tissue. These multimodal biomarkers broadly deepen the understanding of kidney disease progression by integrating lipidomic, fibrotic, and metabolic data.
Collapse
Affiliation(s)
- Anthony A Fung
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Zhi Li
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Craig Boote
- School of Optometry & Vision Sciences, Cardiff University, Cardiff, UK
| | | | - Joseph P Gaut
- Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, MO, USA
| | - Sanjay Jain
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| | - Lingyan Shi
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
5
|
Zhu Z, Wang Y, Qi Z, Hu W, Zhang X, Wagner SK, Wang Y, Ran AR, Ong J, Waisberg E, Masalkhi M, Suh A, Tham YC, Cheung CY, Yang X, Yu H, Ge Z, Wang W, Sheng B, Liu Y, Lee AG, Denniston AK, Wijngaarden PV, Keane PA, Cheng CY, He M, Wong TY. Oculomics: Current concepts and evidence. Prog Retin Eye Res 2025; 106:101350. [PMID: 40049544 DOI: 10.1016/j.preteyeres.2025.101350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
The eye provides novel insights into general health, as well as pathogenesis and development of systemic diseases. In the past decade, growing evidence has demonstrated that the eye's structure and function mirror multiple systemic health conditions, especially in cardiovascular diseases, neurodegenerative disorders, and kidney impairments. This has given rise to the field of oculomics-the application of ophthalmic biomarkers to understand mechanisms, detect and predict disease. The development of this field has been accelerated by three major advances: 1) the availability and widespread clinical adoption of high-resolution and non-invasive ophthalmic imaging ("hardware"); 2) the availability of large studies to interrogate associations ("big data"); 3) the development of novel analytical methods, including artificial intelligence (AI) ("software"). Oculomics offers an opportunity to enhance our understanding of the interplay between the eye and the body, while supporting development of innovative diagnostic, prognostic, and therapeutic tools. These advances have been further accelerated by developments in AI, coupled with large-scale linkage datasets linking ocular imaging data with systemic health data. Oculomics also enables the detection, screening, diagnosis, and monitoring of many systemic health conditions. Furthermore, oculomics with AI allows prediction of the risk of systemic diseases, enabling risk stratification, opening up new avenues for prevention or individualized risk prediction and prevention, facilitating personalized medicine. In this review, we summarise current concepts and evidence in the field of oculomics, highlighting the progress that has been made, remaining challenges, and the opportunities for future research.
Collapse
Affiliation(s)
- Zhuoting Zhu
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, VIC, Australia; Department of Surgery (Ophthalmology), University of Melbourne, Melbourne, VIC, Australia.
| | - Yueye Wang
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Ziyi Qi
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, VIC, Australia; Department of Surgery (Ophthalmology), University of Melbourne, Melbourne, VIC, Australia; Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai, China
| | - Wenyi Hu
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, VIC, Australia; Department of Surgery (Ophthalmology), University of Melbourne, Melbourne, VIC, Australia
| | - Xiayin Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Siegfried K Wagner
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, UK; Institute of Ophthalmology, University College London, London, UK
| | - Yujie Wang
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, VIC, Australia; Department of Surgery (Ophthalmology), University of Melbourne, Melbourne, VIC, Australia
| | - An Ran Ran
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, USA
| | - Ethan Waisberg
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mouayad Masalkhi
- University College Dublin School of Medicine, Belfield, Dublin, Ireland
| | - Alex Suh
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Yih Chung Tham
- Department of Ophthalmology and Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Carol Y Cheung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaohong Yang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zongyuan Ge
- Monash e-Research Center, Faculty of Engineering, Airdoc Research, Nvidia AI Technology Research Center, Monash University, Melbourne, VIC, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Bin Sheng
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Liu
- Google Research, Mountain View, CA, USA
| | - Andrew G Lee
- Center for Space Medicine and the Department of Ophthalmology, Baylor College of Medicine, Houston, USA; Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, USA; The Houston Methodist Research Institute, Houston Methodist Hospital, Houston, USA; Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine, New York, USA; Department of Ophthalmology, University of Texas Medical Branch, Galveston, USA; University of Texas MD Anderson Cancer Center, Houston, USA; Texas A&M College of Medicine, Bryan, USA; Department of Ophthalmology, The University of Iowa Hospitals and Clinics, Iowa City, USA
| | - Alastair K Denniston
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, UK; Institute of Ophthalmology, University College London, London, UK; National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre (BRC), University Hospital Birmingham and University of Birmingham, Birmingham, UK; University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK; Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK
| | - Peter van Wijngaarden
- Centre for Eye Research Australia, Ophthalmology, University of Melbourne, Melbourne, VIC, Australia; Department of Surgery (Ophthalmology), University of Melbourne, Melbourne, VIC, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Pearse A Keane
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, UK; Institute of Ophthalmology, University College London, London, UK
| | - Ching-Yu Cheng
- Department of Ophthalmology and Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Mingguang He
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China; Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Kowloon, Hong Kong, China; Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, China
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua Medicine, Tsinghua University, Beijing, China.
| |
Collapse
|
6
|
Li W, Li W, Ren L, Zhao W, Zhou Y, Li X, Tu P, Liu W, Song Y. Online extraction-LC-MS/MS is an alternative imaging tool for spatial-resolved metabolomics: Mint leaf as a pilot study. Food Chem 2025; 473:143069. [PMID: 39879757 DOI: 10.1016/j.foodchem.2025.143069] [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: 07/03/2024] [Revised: 01/11/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
An attempt was made here to a complemental analytical tool for classical MSI approach. OLE-LC-MS/MS imaging was proposed to plot the spatial-resolved metabolome through deploying mint leaf as a proof-of-concept. A dried leaf underwent chemical composition characterization using OLE-LC-Qtof-MS. Another dried leaf was cut into small pieces, and all pieces were successively packed into a suitable cartridge to undergo OLE-LC-SRM measurements. Fifty-two compounds were observed and identified. Special attention was paid onto isomeric identification using fragment ion intensity ranking style, e.g., 3-O-caffeoylquinic acid vs. 4-O-caffeoylquinic acid. Thereof, 23 abundant ones were involved for relatively quantitative analysis. Quantitative settings were optimized using online ER-MS program. Following spatial metabolome imaging, regioselective distributions were observed for most concerned metabolites. Particularly, isomer-specific occurrences were observed for luteolin-7-O-glucuronide and luteolin-3'-O-glucuronide. Together, OLE-LC-MS/MS is alternative for spatial metabolome imaging due to the advantages at isomeric separation, identification confidence, and quantitative accuracy.
Collapse
Affiliation(s)
- Wei Li
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China,; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Wenzheng Li
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China,; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Luyao Ren
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China,; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Wenhui Zhao
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China,; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Yuxuan Zhou
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China,; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Xiaoyun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China,; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Wenjing Liu
- School of Pharmacy, Henan University of Chinese Medicine, Jinshui East Road, Zhengdong New District, Zhengzhou 450046, China..
| | - Yuelin Song
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102401, China,.
| |
Collapse
|
7
|
Bhatia D, Srivastava SP. Editorial: Diabetic kidney disease: routes to drug development, pharmacology and underlying molecular mechanisms, volume II. Front Pharmacol 2025; 16:1609100. [PMID: 40351438 PMCID: PMC12061879 DOI: 10.3389/fphar.2025.1609100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025] Open
Affiliation(s)
- Divya Bhatia
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | | |
Collapse
|
8
|
Aihara S, Muto Y. Single-cell epigenetics and multiomics analysis in kidney research. Clin Exp Nephrol 2025:10.1007/s10157-025-02679-8. [PMID: 40281349 DOI: 10.1007/s10157-025-02679-8] [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: 01/02/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025]
Abstract
The rapid evolution of single-cell sequencing technologies has significantly advanced our knowledge of cellular heterogeneity and the underlying molecular basis in healthy and diseased kidneys. While single-cell transcriptomic analysis excels in characterizing cell states in the heterogeneous population, the complex regulatory mechanisms governing the gene expressions are difficult to decipher using transcriptomic data alone. Single-cell sequencing technology has recently extended to include epigenome and other modalities, allowing single-cell multiomics analysis. Especially, the integrative analysis of epigenome and transcriptome dissects the cell-specific, gene-regulatory mechanisms driving cellular heterogeneity. An increasing number of single-cell multimodal atlases are being generated in nephrology research, offering novel insights into cellular diversity and the underpinning epigenetic regulation. This ongoing paradigm shift in kidney research accelerates the identification of new biomarkers and potential therapeutic targets, promoting clinical translation. In this era of transformative nephrology research, the basic knowledge of single-cell sequencing analysis and multiomics approach is valuable not only for basic science researchers but for all nephrologists. This review overview single-cell analysis, with a focus on emerging epigenomic and multiomics approaches and their application to kidney research.
Collapse
Affiliation(s)
- Seishi Aihara
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5901 Forest Park Rd., Dallas, TX, 75390, USA
| | - Yoshiharu Muto
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5901 Forest Park Rd., Dallas, TX, 75390, USA.
| |
Collapse
|
9
|
Velu PP, Abhari RE, Henderson NC. Spatial genomics: Mapping the landscape of fibrosis. Sci Transl Med 2025; 17:eadm6783. [PMID: 40203082 DOI: 10.1126/scitranslmed.adm6783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
Organ fibrosis causes major morbidity and mortality worldwide. Treatments for fibrosis are limited, with organ transplantation being the only cure. Here, we review how various state-of-the-art spatial genomics approaches are being deployed to interrogate fibrosis across multiple organs, providing exciting insights into fibrotic disease pathogenesis. These include the detailed topographical annotation of pathogenic cell populations and states, detection of transcriptomic perturbations in morphologically normal tissue, characterization of fibrotic and homeostatic niches and their cellular constituents, and in situ interrogation of ligand-receptor interactions within these microenvironments. Together, these powerful readouts enable detailed analysis of fibrosis evolution across time and space.
Collapse
Affiliation(s)
- Prasad Palani Velu
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Roxanna E Abhari
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Neil C Henderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4UU, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 1QY, UK
| |
Collapse
|
10
|
Marquez J, O’Sullivan L, Squire AE, Ryan GL, Debiec KE, Amies Oelschlager AM, Adam MP. Case Report: a novel variant in WT1 leads to focal segmental glomerulosclerosis and uterovaginal anomalies through exon skipping. FRONTIERS IN NEPHROLOGY 2025; 5:1542475. [PMID: 40235736 PMCID: PMC11997443 DOI: 10.3389/fneph.2025.1542475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/28/2025] [Indexed: 04/17/2025]
Abstract
Background Podocytopathies are a varied set of renal diseases in which podocytes are unable to perform their typical filtration function within the glomerulus. This typically leads to edema, proteinuria, and hypoalbuminemia early in life. Among podocytopathies, focal segmental glomerulosclerosis (FSGS) is characterized by histology demonstrating segmental and focal sclerosis of the glomerular tuft. FSGS affects an estimated 1-20 per one million individuals and leads to significant morbidity and mortality related to renal failure. While FSGS can be attributed to many causes, such as drug reactions and infections, underlying pathogenic genetic variants play an increasingly well-recognized role in this disease. Case A 38-year-old 46,XX female patient of self-reported Cambodian ancestry was evaluated due to her history of atypical uterovaginal morphology. She had a history of hypertension and nephrotic range proteinuria that was diagnosed early in adulthood. A kidney biopsy at that time revealed FSGS. Following worsening renal function and subsequent end-stage renal disease (ESRD), she underwent a kidney transplant at 33 years of age. After kidney transplant, she presented with hematocolpos and was found to have distal vaginal atresia and an arcuate uterus. She underwent vaginoplasty and then had regular menses. She was noted to have persistently elevated follicle stimulating hormone levels, consistent with primary ovarian insufficiency, but with normal anti-Müllerian hormone levels. Assessment of her family history was suggestive of other individuals in her family with similar renal disease and uterine differences. Genetic analysis identified a WT1 variant (c.1338A>C; p. =) of uncertain significance that is also present in her similarly affected mother. To help clarify the potential impact of this variant, we completed a mini-gene assay to detect in vitro splicing changes in the presence of the WT1 variant sequence uncovered in this individual. This demonstrated resultant aberrant splicing that further supports the pathogenicity of the uncovered variant for this individual. Conclusions To our knowledge, this represents the first case of a podocytopathy with co-occurring uterovaginal anomalies due to exon skipping in WT1. The patient exhibited a severe course of chronic kidney dysfunction requiring a kidney transplant. Clinical RNA sequencing to clarify variants impacting splicing remains challenging due to tissue- specific gene expression for genes such as WT1, thus, research-based assays may be beneficial to understand the consequence of rare or previously uncharacterized variants.
Collapse
Affiliation(s)
- Jonathan Marquez
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Lauren O’Sullivan
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Audrey E. Squire
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Ginny L. Ryan
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Washington, Seattle, WA, United States
| | - Katherine E. Debiec
- Department of Obstetrics and Gynecology, Division of Pediatric and Adolescent Gynecology, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Anne-Marie Amies Oelschlager
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Washington, Seattle, WA, United States
| | - Margaret P. Adam
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| |
Collapse
|
11
|
Li H, Bao S, Farzad N, Qin X, Fung AA, Zhang D, Bai Z, Tao B, Fan R. Spatially resolved genome-wide joint profiling of epigenome and transcriptome with spatial-ATAC-RNA-seq and spatial-CUT&Tag-RNA-seq. Nat Protoc 2025:10.1038/s41596-025-01145-9. [PMID: 40119005 DOI: 10.1038/s41596-025-01145-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/15/2024] [Indexed: 03/24/2025]
Abstract
The epigenome of a cell is tightly correlated with gene transcription, which controls cell identity and diverse biological activities. Recent advances in spatial technologies have improved our understanding of tissue heterogeneity by analyzing transcriptomics or epigenomics with spatial information preserved, but have been mainly restricted to one molecular layer at a time. Here we present procedures for two spatially resolved sequencing methods, spatial-ATAC-RNA-seq and spatial-CUT&Tag-RNA-seq, that co-profile transcriptome and epigenome genome wide. In both methods, transcriptomic readouts are generated through tissue fixation, permeabilization and in situ reverse transcription. In spatial-ATAC-RNA-seq, Tn5 transposase is used to probe accessible chromatin, and in spatial-CUT&Tag-RNA-seq, the tissue is incubated with primary antibodies that target histone modifications, followed by Protein A-fused Tn5-induced tagmentation. Both methods leverage a microfluidic device that delivers two sets of oligonucleotide barcodes to generate a two-dimensional mosaic of tissue pixels at near single-cell resolution. A spatial-ATAC-RNA-seq or spatial-CUT&Tag-RNA-seq library can be generated in 3-5 d, allowing researchers to simultaneously investigate the transcriptomic landscape and epigenomic landscape of an intact tissue section. This protocol is an extension of our previous spatially resolved epigenome sequencing protocol and provides opportunities in multimodal profiling.
Collapse
Affiliation(s)
- Haikuo Li
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Shuozhen Bao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Negin Farzad
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xiaoyu Qin
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Anthony A Fung
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Di Zhang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Bo Tao
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
- Yale Stem Cell Center and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Research on Aging (Y-Age), Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
12
|
Zheng Z, Qiao X, Yin J, Kong J, Han W, Qin J, Meng F, Tian G, Feng X. Advancements in omics technologies: Molecular mechanisms of acute lung injury and acute respiratory distress syndrome (Review). Int J Mol Med 2025; 55:38. [PMID: 39749711 PMCID: PMC11722059 DOI: 10.3892/ijmm.2024.5479] [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: 09/06/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an inflammatory response arising from lung and systemic injury with diverse causes and associated with high rates of morbidity and mortality. To date, no fully effective pharmacological therapies have been established and the relevant underlying mechanisms warrant elucidation, which may be facilitated by multi‑omics technology. The present review summarizes the application of multi‑omics technology in identifying novel diagnostic markers and therapeutic strategies of ALI/ARDS as well as its pathogenesis.
Collapse
Affiliation(s)
- Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junjie Kong
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Wanqing Han
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jing Qin
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Fanda Meng
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, P.R. China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
13
|
Zhu Z, Cao Y, Jian Y, Hu H, Yang Q, Hao Y, Jiang H, Luo Z, Yang X, Li W, Hu J, Liu H, Liang W, Ding G, Chen Z. CerS6 links ceramide metabolism to innate immune responses in diabetic kidney disease. Nat Commun 2025; 16:1528. [PMID: 39934147 PMCID: PMC11814332 DOI: 10.1038/s41467-025-56891-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: 06/06/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
Ectopic lipid deposition, mitochondrial injury, and inflammatory responses contribute to the development of diabetic kidney disease (DKD); however, the mechanistic link between these processes remains unclear. In this study, we demonstrate that the ceramide synthase 6 (CerS6) is primarily localized in podocytes of the glomeruli and is upregulated in two different models of diabetic mice. Podocyte-specific CerS6 knockout ameliorates glomerular injury and inflammatory responses in male diabetic mice and in male mice with adriamycin-induced nephropathy. In contrast, podocyte-specific overexpression of CerS6 sufficiently induces proteinuria. Mechanistically, CerS6-derived ceramide (d18:1/16:0) can bind to the mitochondrial channel protein VDAC1 at Glu59 residue, initiating mitochondrial DNA (mtDNA) leakage, activating the cGAS-STING signaling pathway, and ultimately promoting an immune-inflammatory response in the kidney. Importantly, CERS6 expression is increased in podocytes from kidney biopsies of patients with DKD and focal segmental glomerulosclerosis (FSGS), and the expression level of CERS6 is correlated negatively with glomerular filtration rate and positively with proteinuria. Thus, our findings suggest that targeting CerS6 may be a potential therapeutic strategy for proteinuric kidney diseases.
Collapse
Affiliation(s)
- Zijing Zhu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Yun Cao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Yonghong Jian
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Qian Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Yiqun Hao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Houhui Jiang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Zilv Luo
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Xueyan Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Weiwei Li
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Hongyan Liu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China.
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China.
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, China.
- Hubei Clinical Research Center of Kidney Disease, Wuhan, China.
| |
Collapse
|
14
|
van den Berg CW, Dumas SJ, Little MH, Rabelink TJ. Challenges in maturation and integration of kidney organoids for stem cell-based renal replacement therapy. Kidney Int 2025; 107:262-270. [PMID: 39571903 DOI: 10.1016/j.kint.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 12/10/2024]
Abstract
Human pluripotent stem cell-derived kidney organoids hold promise for future applications in regenerative medicine. However, significant biological hurdles need to be overcome to enable their use as a transplantable stem cell-derived therapeutic graft. Current kidney organoid protocols do not recapitulate a complete integrated developing kidney, but embryonic kidney transplantations have provided clues for advancing maturation and functionality of kidney organoids. Transplantation, subsequent vascularization, and blood perfusion of kidney organoids improve nephron patterning and maturation, suggesting a role for angiocrine factors as well as metabolic wiring in these processes. Transplanted organoids exhibit filtration, but the resulting filtrate has no apparent exit path for excretion. Improved in vitro patterning of kidney organoids may be required such that a more structurally correct tissue is formed before transplant. Here we review current progress with transplantation of kidney organoids, as well as their engraftment and integration, and identify the key obstacles to therapeutic success and how these might be achieved.
Collapse
Affiliation(s)
- Cathelijne W van den Berg
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, the Netherlands.
| | - Sébastien J Dumas
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, the Netherlands
| | - Melissa H Little
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Ton J Rabelink
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
15
|
Li S, Susztak K. Mitochondrial dysfunction has a central role in diabetic kidney disease. Nat Rev Nephrol 2025; 21:77-78. [PMID: 39681602 DOI: 10.1038/s41581-024-00919-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Affiliation(s)
- Shen Li
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Penn/CHOP Kidney Innovation Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Demirci H, Bahena-Lopez J, Smorodchenko A, Su XT, Nelson J, Yang CL, Curry J, Duan XP, Wang WH, Sharkovska Y, Liu R, Yilmaz DE, Quintanova C, Emberly K, Emery B, Himmerkus N, Bleich M, Ellison DH, Bachmann S. Distinct cell types along thick ascending limb express pathways for monovalent and divalent cation transport. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633282. [PMID: 39896580 PMCID: PMC11785040 DOI: 10.1101/2025.01.16.633282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Kidney thick ascending limb cells reabsorb sodium, potassium, calcium, and magnesium and contribute to urinary concentration. These cells are typically viewed as of a single type that recycles potassium across the apical membrane and generates a lumen-positive transepithelial voltage driving calcium and magnesium reabsorption, although variability in potassium channel expression has been reported. Additionally, recent transcriptomic analyses suggest that different cell types exist along this segment, but classifications have varied and have not led to a new consensus model. We used immunolocalization, electrophysiology and enriched single nucleus RNA-Seq to identify thick ascending limb cell types in rat, mouse and human. We identified three major TAL cell types defined by expression of potassium channels and claudins. One has apical potassium channels, low basolateral potassium conductance, and is bordered by a sodium-permeable claudin. A second lacks apical potassium channels, has high basolateral potassium conductance and is bordered by calcium- and magnesium-permeable claudins. A third type also lacks apical potassium channels and has a high basolateral potassium conductance, but these cells are ringed by sodium-permeable claudins. The recognition of diverse cell types resolves longstanding questions about how solute transport can be modulated selectively and how disruption of these cells leads to human disease.
Collapse
Affiliation(s)
- Hasan Demirci
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Cell- and Neurobiology, Charité-Universitätsmedizin Berlin, 10117 Berlin Germany
| | - Jessica Bahena-Lopez
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, Oregon
| | | | - Xiao-Tong Su
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, Oregon
| | - Jonathan Nelson
- Division of Nephrology & Hypertension, USC Keck School of Medicine, Los Angeles, CA
| | - Chao-Ling Yang
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, Oregon
| | - Joshua Curry
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, Oregon
| | - Xin-Peng Duan
- Department of Physiology, Xuzhou Medical University, 221004 Xuzhou, China
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, New York
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, New York
| | - Yuliya Sharkovska
- Klinik für Pädiatrie, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Ruisheng Liu
- Department of Molecular Pharmacology & Physiology, University of South Florida, Tampa, FL
| | - Duygu Elif Yilmaz
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Catarina Quintanova
- Institute of Physiology, Christian-Albrechts-University, 24118 Kiel, Germany
| | - Katie Emberly
- Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR
| | - Ben Emery
- Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR
| | - Nina Himmerkus
- Institute of Physiology, Christian-Albrechts-University, 24118 Kiel, Germany
| | - Markus Bleich
- Institute of Physiology, Christian-Albrechts-University, 24118 Kiel, Germany
| | - David H. Ellison
- Division of Hypertension and Nephrology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, Oregon
| | - Sebastian Bachmann
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Cell- and Neurobiology, Charité-Universitätsmedizin Berlin, 10117 Berlin Germany
| |
Collapse
|
17
|
Crotta Asis A, Asaro A, D'Angelo G. Single cell lipid biology. Trends Cell Biol 2025:S0962-8924(24)00255-1. [PMID: 39814618 DOI: 10.1016/j.tcb.2024.12.002] [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: 08/19/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
Lipids are major cell constituents endowed with astonishing structural diversity. The pathways responsible for the assembly and disposal of different lipid species are energetically demanding, and genes encoding lipid metabolic factors and lipid-related proteins comprise a sizable fraction of our coding genome. Despite the importance of lipids, the biological significance of lipid structural diversity remains largely obscure. Recent technological developments have enabled extensive lipid analysis at the single cell level, revealing unexpected cell-cell variability in lipid composition. This new evidence suggests that lipid diversity is exploited in multicellularity and that lipids have a role in the establishment and maintenance of cell identity. In this review, we highlight the emerging concepts and technologies in single cell lipid analysis and the implications of this research for future studies.
Collapse
Affiliation(s)
- Agostina Crotta Asis
- Institute of Bioengineering (IBI) and Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Antonino Asaro
- Institute of Bioengineering (IBI) and Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Giovanni D'Angelo
- Institute of Bioengineering (IBI) and Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
18
|
Lopes MB, Coletti R, Duranton F, Glorieux G, Jaimes Campos MA, Klein J, Ley M, Perco P, Sampri A, Tur-Sinai A. The Omics-Driven Machine Learning Path to Cost-Effective Precision Medicine in Chronic Kidney Disease. Proteomics 2025:e202400108. [PMID: 39790049 DOI: 10.1002/pmic.202400108] [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: 11/07/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Chronic kidney disease (CKD) poses a significant and growing global health challenge, making early detection and slowing disease progression essential for improving patient outcomes. Traditional diagnostic methods such as glomerular filtration rate and proteinuria are insufficient to capture the complexity of CKD. In contrast, omics technologies have shed light on the molecular mechanisms of CKD, helping to identify biomarkers for disease assessment and management. Artificial intelligence (AI) and machine learning (ML) could transform CKD care, enabling biomarker discovery for early diagnosis and risk prediction, and personalized treatment. By integrating multi-omics datasets, AI can provide real-time, patient-specific insights, improve decision support, and optimize cost efficiency by early detection and avoidance of unnecessary treatments. Multidisciplinary collaborations and sophisticated ML methods are essential to advance diagnostic and therapeutic strategies in CKD. This review presents a comprehensive overview of the pipeline for translating CKD omics data into personalized treatment, covering recent advances in omics research, the role of ML in CKD, and the critical need for clinical validation of AI-driven discoveries to ensure their efficacy, relevance, and cost-effectiveness in patient care.
Collapse
Affiliation(s)
- Marta B Lopes
- Center for Mathematics and Applications (NOVA Math), NOVA School of Science and Technology (NOVA FCT), Caparica, Portugal
- UNIDEMI, Department of Mechanical and Industrial Engineering, NOVA School of Science and Technology (NOVA FCT), Caparica, Portugal
| | - Roberta Coletti
- Center for Mathematics and Applications (NOVA Math), NOVA School of Science and Technology (NOVA FCT), Caparica, Portugal
| | | | - Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Unit, Ghent University Hospital, Gent, Belgium
| | - Mayra Alejandra Jaimes Campos
- Department of Biomarker Research, Mosaiques Diagnostics GmbH, Hannover, Germany
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Aachen, Germany
| | - Julie Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM), Institute of Cardiovascular and Metabolic Disease, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Matthias Ley
- Delta4 GmbH, Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University Vienna, Vienna, Austria
| | - Paul Perco
- Delta4 GmbH, Vienna, Austria
- Department of Internal Medicine IV, Medical University Innsbruck, Innsbruck, Austria
| | - Alexia Sampri
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
19
|
Zhang R, Dai F, Deng S, Zeng Y, Wang J, Liu G. Reprogramming of Glucose Metabolism for Revisiting Hepatocellular Carcinoma Resistance to Transcatheter Hepatic Arterial Chemoembolization. Chembiochem 2025; 26:e202400719. [PMID: 39501124 DOI: 10.1002/cbic.202400719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/04/2024] [Indexed: 11/24/2024]
Abstract
Hepatocellular carcinoma (HCC) is recognized globally as one of the most lethal tumors, presenting a significant menace to patients' lives owing to its exceptional aggressiveness and tendency to recur. Transcatheter hepatic arterial chemoembolization (TACE) therapy, as a first-line treatment option for patients with advanced HCC, has been proven effective. However, it is disheartening that nearly 40 % of patients exhibit resistance to this therapy. Consequently, this review delves into the metabolic aspects of glucose metabolism to explore the underlying mechanisms behind TACE treatment resistance and to propose potentially fruitful therapeutic strategies. The ultimate objective is to present novel insights for the development of personalized treatment methods targeting HCC.
Collapse
Affiliation(s)
- Ruijie Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Fan Dai
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Songhan Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Yun Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Jinyang Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
20
|
Janosevic D, De Luca T, Eadon MT. The Kidney Precision Medicine Project and Single-Cell Biology of the Injured Proximal Tubule. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:7-22. [PMID: 39332674 PMCID: PMC11686451 DOI: 10.1016/j.ajpath.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) has led to major advances in our understanding of proximal tubule subtypes in health and disease. The proximal tubule serves essential functions in overall homeostasis, but pathologic or physiological perturbations can affect its transcriptomic signature and corresponding tasks. These alterations in proximal tubular cells are often described within a scRNA-seq atlas as cell states, which are pathophysiological subclassifications based on molecular and morphologic changes in a cell's response to that injury compared with its native state. This review describes the major cell states defined in the Kidney Precision Medicine Project's scRNA-seq atlas. It then identifies the overlap between the Kidney Precision Medicine Project and other seminal works that may use different nomenclature or cluster proximal tubule cells at different resolutions to define cell state subtypes. The goal is for the reader to understand the key transcriptomic markers of important cellular injury and regeneration processes across this highly dynamic and evolving field.
Collapse
Affiliation(s)
- Danielle Janosevic
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Thomas De Luca
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Michael T Eadon
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
21
|
Yoshikawa T, Yanagita M. Single-Cell Analysis Provides New Insights into the Roles of Tertiary Lymphoid Structures and Immune Cell Infiltration in Kidney Injury and Chronic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:40-54. [PMID: 39097168 DOI: 10.1016/j.ajpath.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 08/05/2024]
Abstract
Chronic kidney disease (CKD) is a global health concern with high morbidity and mortality. Acute kidney injury (AKI) is a pivotal risk factor for the progression of CKD, and the rate of AKI-to-CKD progression increases with aging. Intrarenal inflammation is a fundamental mechanism underlying AKI-to-CKD progression. Tertiary lymphoid structures (TLSs), ectopic lymphoid aggregates formed in nonlymphoid organs, develop in aged injured kidneys, but not in young kidneys, with prolonged inflammation and maladaptive repair, which potentially exacerbates AKI-to-CKD progression in aged individuals. Dysregulated immune responses are involved in the pathogenesis of various kidney diseases, such as IgA nephropathy, lupus nephritis, and diabetic kidney diseases, thereby deteriorating kidney function. TLSs also develop in several kidney diseases, including transplanted kidneys and renal cell carcinoma. However, the precise immunologic mechanisms driving AKI-to-CKD progression and development of these kidney diseases remain unclear, which hinders the development of novel therapeutic approaches. This review aims to describe recent findings from single-cell analysis of cellular heterogeneity and complex interactions among immune and renal parenchymal cells, which potentially contribute to the pathogenesis of AKI-to-CKD progression and other kidney diseases, highlighting the mechanisms of formation and pathogenic roles of TLSs in aged injured kidneys.
Collapse
Affiliation(s)
- Takahisa Yoshikawa
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan.
| |
Collapse
|
22
|
Baysoy A, Tian X, Zhang F, Renauer P, Bai Z, Shi H, Li H, Tao B, Yang M, Enninful A, Gao F, Wang G, Zhang W, Tran T, Patterson NH, Bao S, Dong C, Xin S, Zhong M, Rankin S, Guy C, Wang Y, Connelly JP, Pruett-Miller SM, Chi H, Chen S, Fan R. Spatially Resolved in vivo CRISPR Screen Sequencing via Perturb-DBiT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624106. [PMID: 39605490 PMCID: PMC11601513 DOI: 10.1101/2024.11.18.624106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Perturb-seq enabled the profiling of transcriptional effects of genetic perturbations in single cells but lacks the ability to examine the impact on tissue environments. We present Perturb-DBiT for simultaneous co-sequencing of spatial transcriptome and guide RNAs (gRNAs) on the same tissue section for in vivo CRISPR screen with genome-scale gRNA libraries, offering a comprehensive understanding of how genetic modifications affect cellular behavior and tissue architecture. This platform supports a variety of delivery vectors, gRNA library sizes, and tissue preparations, along with two distinct gRNA capture methods, making it adaptable to a wide range of experimental setups. In applying Perturb-DBiT, we conducted un-biased knockouts of tens of genes or at genome-wide scale across three cancer models. We mapped all gRNAs in individual colonies and corresponding transcriptomes in a human cancer metastatic colonization model, revealing clonal dynamics and cooperation. We also examined the effect of genetic perturbation on the tumor immune microenvironment in an immune-competent syngeneic model, uncovering differential and synergistic perturbations in promoting immune infiltration or suppression in tumors. Perturb-DBiT allows for simultaneously evaluating the impact of each knockout on tumor initiation, development, metastasis, histopathology, and immune landscape. Ultimately, it not only broadens the scope of genetic inquiry, but also lays the groundwork for developing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Alev Baysoy
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- These authors contributed equally
| | - Xiaolong Tian
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- These authors contributed equally
| | - Feifei Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- These authors contributed equally
| | - Paul Renauer
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- These authors contributed equally
| | - Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Hao Shi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Haikuo Li
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Bo Tao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Mingyu Yang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Archibald Enninful
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Fu Gao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Guangchuan Wang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | - Shuozhen Bao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Chuanpeng Dong
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Shan Xin
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Mei Zhong
- Department of Cell Biology, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Sherri Rankin
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yan Wang
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jon P. Connelly
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | | | - Hongbo Chi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Systems Biology Institute, Integrated Science & Technology Center, West Haven, CT, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA
- Human and Translational Immunology, Yale University School of Medicine, New Haven, CT 06520, USA
- Lead contact
| |
Collapse
|
23
|
Rroji M, Spasovski G. Omics Studies in CKD: Diagnostic Opportunities and Therapeutic Potential. Proteomics 2024:e202400151. [PMID: 39523931 DOI: 10.1002/pmic.202400151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Omics technologies have significantly advanced the prediction and therapeutic approaches for chronic kidney disease (CKD) by providing comprehensive molecular insights. This is a review of the current state and future prospects of integrating biomarkers into the clinical practice for CKD, aiming to improve patient outcomes by targeted therapeutic interventions. In fact, the integration of genomic, transcriptomic, proteomic, and metabolomic data has enhanced our understanding of CKD pathogenesis and identified novel biomarkers for an early diagnosis and targeted treatment. Advanced computational methods and artificial intelligence (AI) have further refined multi-omics data analysis, leading to more accurate prediction models for disease progression and therapeutic responses. These developments highlight the potential to improve CKD patient care with a precise and individualized treatment plan .
Collapse
Affiliation(s)
- Merita Rroji
- Faculty of Medicine, Department of Nephrology, University of Medicine Tirana, Tirana, Albania
| | - Goce Spasovski
- Medical Faculty, Department of Nephrology, University of Skopje, Skopje, North Macedonia
| |
Collapse
|
24
|
Hansen J, Siddiq MM, He JC, Iyengar R. Integrating Metabolomics and Transcriptomics to Characterize Differential Functional Capabilities of Kidney Proximal Tubule Cell Subtypes. Semin Nephrol 2024; 44:151577. [PMID: 40180882 DOI: 10.1016/j.semnephrol.2025.151577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
The coupling between energy metabolism and transport processes is a key feature that defines the functional capability of proximal tubule cells. Recent studies using metabolomics and transcriptomics provide insights into the relationships between changes in single-cell transcriptomic profiles and energy metabolism during kidney development and in disease states. In this review, we describe insights from these studies and how mapping of metabolites to functional pathways within cells enables these insights. We also describe our analyses of fatty acid metabolism pathways from single-cell transcriptomic data obtained by the Kidney Precision Medicine Project, which indicate that proximal tubule cell subtypes can be divided into two major groups with high and low levels of mRNAs for fatty acid (beta) oxidation enzymes. On average, patients with CKD have higher levels of cells with low fatty acid oxidation capability. These cells also have lower levels of sodium transporters. Within each group of proximal tubule cell subtypes there is considerable variability between individual patients. Integrating these data with metabolomics analyses can provide insights into how the differential metabolic capabilities of proximal tubule cells are related to disease features in individual patients. Identifying such relationships can lead to development of precision medicine approaches in nephrology.
Collapse
Affiliation(s)
- Jens Hansen
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mustafa M Siddiq
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY; James J Peter Department of Veterans Affairs Medical Center, New York, NY
| | - Ravi Iyengar
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
25
|
Min X, Zhao Y, Yu M, Zhang W, Jiang X, Guo K, Wang X, Huang J, Li T, Sun L, He J. Spatially resolved metabolomics: From metabolite mapping to function visualising. Clin Transl Med 2024; 14:e70031. [PMID: 39456123 PMCID: PMC11511672 DOI: 10.1002/ctm2.70031] [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: 05/05/2024] [Revised: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 10/28/2024] Open
Abstract
Mass spectrometry imaging (MSI)-based spatially resolved metabolomics addresses the limitations inherent in traditional liquid chromatography-tandem mass spectrometry (LC-MS)-based metabolomics, particularly the loss of spatial context within heterogeneous tissues. MSI not only enhances our understanding of disease aetiology but also aids in the identification of biomarkers and the assessment of drug toxicity and therapeutic efficacy by converting invisible metabolites and biological networks into visually rendered image data. In this comprehensive review, we illuminate the key advancements in MSI-driven spatially resolved metabolomics over the past few years. We first outline recent innovations in preprocessing methodologies and MSI instrumentation that improve the sensitivity and comprehensiveness of metabolite detection. We then delve into the progress made in functional visualization techniques, which enhance the precision of metabolite identification and annotation. Ultimately, we discuss the significant potential applications of spatially resolved metabolomics technology in translational medicine and drug development, offering new perspectives for future research and clinical translation. HIGHLIGHTS: MSI-driven spatial metabolomics preserves metabolite spatial information, enhancing disease analysis and biomarker discovery. Advances in MSI technology improve detection sensitivity and accuracy, expanding bioanalytical applications. Enhanced visualization techniques refine metabolite identification and spatial distribution analysis. Integration of MSI with AI promises to advance precision medicine and accelerate drug development.
Collapse
Affiliation(s)
- Xinyue Min
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yiran Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Meng Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wenchao Zhang
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinyi Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Kaijing Guo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiangyi Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jianpeng Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Tong Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lixin Sun
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Jiuming He
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- NMPA Key Laboratory of safety research and evaluation of Innovative Drug, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
26
|
Zhang G, Reeves WB. Spatial Metabolomics in Acute Kidney Injury. Semin Nephrol 2024; 44:151580. [PMID: 40221281 DOI: 10.1016/j.semnephrol.2025.151580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Acute kidney injury (AKI) is a common condition linked to increased morbidity, mortality, and substantial health care costs both in the United States and globally. Early diagnosis, prompt intervention, and effective therapeutic management of AKI are vital for improving patient outcomes. Recent advancements in renal imaging and omics technologies have provided new perspectives and deeper insights into kidney injury while also presenting challenges in developing a comprehensive cellular and molecular atlas of the condition. This review focuses on the application of mass spectrometry imaging-based spatial metabolomics in studying ischemia- and toxin-induced AKI in animal models and human patients. Spatial metabolomics offers a deeper understanding of the pathophysiological connections between various processes, such as dysregulated lipid metabolism and the shift from the tricarboxylic acid cycle to glycolytic flux, which contribute to functional impairment and structural damage in AKI. Continued research in renal multimodal imaging and omics is essential to further our understanding of kidney injury from diagnostic, mechanistic, and therapeutic perspectives.
Collapse
Affiliation(s)
- Guanshi Zhang
- Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX; Audie L. Murphy Memorial VA Hospital, South Texas Veterans Health Care System, San Antonio, TX.
| | - W Brian Reeves
- Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, TX
| |
Collapse
|
27
|
Fung AA, Li Z, Boote C, Markov P, Jain S, Shi L. Label-Free Optical Biopsy Reveals Biomolecular and Morphological Features of Diabetic Kidney Tissue in 2D and 3D. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.27.620507. [PMID: 39553929 PMCID: PMC11565847 DOI: 10.1101/2024.10.27.620507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Kidney disease, the ninth leading cause of death in the United States, has one of the poorest diagnostic efficiencies of only 10%1. Conventional diagnostic methods often rely on light microscopy analysis of 2D fixed tissue sections with limited molecular insight compared to omics studies. Targeting multiple features in a biopsy using molecular or chemical reagents can enhance molecular phenotyping but are limited by overlap of their spatial and chromatic properties, variations in quality of the products, limited multimodal nature and need additional tissue processing. To overcome these limitations and increase the breadth of molecular information available from tissue without an impact on routine diagnostic workup, we implemented label-free imaging modalities including stimulated Raman scattering (SRS) microscopy, second harmonic generation (SHG), and two photon fluorescence (TPF) into a single microscopy setup. We visualized and identified morphological, structural, lipidomic, and metabolic biomarkers of control and diabetic human kidney biopsy samples in 2D and 3D at a subcellular resolution. The label-free biomarkers, including collagen fiber morphology, mesangial-glomerular fractional volume, lipid saturation, redox status, and relative lipid and protein concentrations in the form of Stimulated Raman Histology (SRH), illustrate distinct features in kidney disease tissues not previously appreciated. The same tissue section can be used for routine diagnostic work up thus enhancing the power of cliniopathological insights obtainable without compromising already limited tissue. The additional multimodal biomarkers and metrics are broadly applicable and deepen our understanding of the progression of kidney diseases by integrating lipidomic, fibrotic, and metabolic data.
Collapse
Affiliation(s)
- Anthony A Fung
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA 92093
| | - Zhi Li
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA 92093
| | - Craig Boote
- School of Optometry & Vision Sciences, Cardiff University, Cardiff, UK CF24 4HQ
| | - Petar Markov
- EMBL c/o DESY, Notkestr. 85, Geb. 25a, Hamburg, Germany
| | - Sanjay Jain
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Lingyan Shi
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA 92093
| |
Collapse
|
28
|
Li H, Humphreys BD. Protocol for multimodal profiling of human kidneys with simultaneous high-throughput ATAC and RNA expression with sequencing. STAR Protoc 2024; 5:103049. [PMID: 38900631 PMCID: PMC11239685 DOI: 10.1016/j.xpro.2024.103049] [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: 03/05/2024] [Revised: 03/31/2024] [Accepted: 04/15/2024] [Indexed: 06/22/2024] Open
Abstract
Simultaneous high-throughput ATAC and RNA expression with sequencing (SHARE-seq) profiles transcriptomics and chromatin accessibility in the same cells at high throughput. Here, we present a protocol for multimodal profiling of human kidneys with SHARE-seq. We describe steps for processing fixed nuclei for SHARE-seq split-pool barcoding and library preparation. We also detail how to determine the optimal working concentration of Tn5 transposase for transposition and tagmentation. This protocol allows researchers to generate large-scale single-cell multiomics data at low reagent cost. For complete details on the use and execution of this protocol, please refer to Li et al.1.
Collapse
Affiliation(s)
- Haikuo Li
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63105, USA.
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63105, USA; Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO 63105, USA.
| |
Collapse
|
29
|
Li H, Li D, Humphreys BD. Chromatin conformation and histone modification profiling across human kidney anatomic regions. Sci Data 2024; 11:797. [PMID: 39025878 PMCID: PMC11258246 DOI: 10.1038/s41597-024-03648-8] [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: 03/18/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024] Open
Abstract
The three major anatomic regions of the human kidney include the cortex, medulla and papilla, with different functions and vulnerabilities to kidney diseases. Epigenetic mechanisms underlying these anatomic structures are incompletely understood. Here, we performed chromatin conformation capture with Hi-C and histone modification H3K4me3/H3K27me3 Cleavage Under Targets and Release Using Nuclease (CUT&RUN) sequencing on the kidney cortex, medulla and papilla dissected from one individual donor. Nuclear suspensions were generated from each region and split subjected to paired Hi-C and CUT&RUN sequencing. We evaluated the quality of next-generation sequencing data, Hi-C chromatin contact matrices and CUT&RUN peak calling. H3K4me3 and H3K27me3 histone modifications represent active and repressive gene transcription, respectively, and differences in chromatin conformation between kidney regions can be analyzed with this dataset. All raw and processed data files are publicly available, allowing researchers to survey the epigenetic landscape across regional human kidney anatomy.
Collapse
Affiliation(s)
- Haikuo Li
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Dian Li
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
30
|
Li H, Humphreys BD. Spatially resolved metabolomic dataset of distinct human kidney anatomic regions. Data Brief 2024; 54:110431. [PMID: 38708307 PMCID: PMC11067325 DOI: 10.1016/j.dib.2024.110431] [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: 03/22/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Cortex, medulla and papilla are three major human kidney anatomic structures and they harbour unique metabolic functions, but the underlying metabolomic profiles are largely unknown at spatial resolution. Here, we generated a spatially resolved metabolomics dataset on human kidney cortex, medulla and papilla tissues dissected from the same donor. Matrix-Assisted Laser Desorption/Ionization-Imaging Mass Spectrometry (MALDI-IMS) was used to detect metabolite species over mass-to-charge ratios of 50 -1500 for each section at a resolution of 10 × 10 µm2 pixel size. We present raw data matrix of each sample, feature annotations, raw AnnData merged from three samples and processed AnnData files after quality control, dimensional reduction and data integration, which contains a total of 170,459 spatially resolved metabolomes with 562 features detected. This dataset can be either visualized through an interactive browser or further analyzed to study metabolomic heterogeneity across regional human kidney anatomy.
Collapse
Affiliation(s)
- Haikuo Li
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Benjamin D. Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
31
|
Huan C, Li J, Li Y, Zhao S, Yang Q, Zhang Z, Li C, Li S, Guo Z, Yao J, Zhang W, Zhou L. Spatially Resolved Multiomics: Data Analysis from Monoomics to Multiomics. BME FRONTIERS 2024; 6:0084. [PMID: 39810754 PMCID: PMC11725630 DOI: 10.34133/bmef.0084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/05/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
Spatial monoomics has been recognized as a powerful tool for exploring life sciences. Recently, spatial multiomics has advanced considerably, which could contribute to clarifying many biological issues. Spatial monoomics techniques in epigenomics, genomics, transcriptomics, proteomics, and metabolomics can enhance our understanding of biological functions and cellular identities by simultaneously measuring tissue structures and biomolecule levels. Spatial monoomics technology has evolved from monoomics to spatial multiomics. Moreover, the spatial resolution, high-throughput detection capability, capture efficiency, and compatibility with various sample types of omics technology have considerably advanced. Despite the technological advances in this field, data analysis frameworks have stagnated. Current challenges include incomplete spatial monoomics data analysis pipeline, overly complex data analysis tasks, and few established spatial multiomics data analysis strategies. In this review, we systematically summarize recent developments of various spatial monoomics techniques and improvements in related data analysis pipeline. On the basis of the spatial multiomics technology, we propose a data integration strategy with cross-platform, cross-slice, and cross-modality. We summarize the potential applications of spatial monoomics technology, aiming to provide researchers and clinicians with a better understanding of how such applications have advanced. Spatial multiomics technology is expected to substantially impact biology and precision medicine through measurements of cellular tissue structures and the extraction of biomolecular features.
Collapse
Affiliation(s)
- Changxiang Huan
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230026, China
| | - Jinze Li
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
| | - Yingxue Li
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
| | - Shasha Zhao
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
| | - Qi Yang
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
| | - Zhiqi Zhang
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
| | - Chuanyu Li
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230026, China
| | - Shuli Li
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
| | - Zhen Guo
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230026, China
| | - Jia Yao
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230026, China
| | - Wei Zhang
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230026, China
| | - Lianqun Zhou
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology,
Chinese Academy of Sciences, Suzhou 215163, China
| |
Collapse
|