1
|
Cui X, Zhong Z, Xu S, Pan Y, Wang X, Zhang L, He A, Ye X, Cao H, Zhang W, Tian R. Ion exchange- and enrichment-based technology applied to large-scale plasma proteomic analysis of breast cancer neoadjuvant chemotherapy. J Chromatogr A 2025; 1750:465914. [PMID: 40188783 DOI: 10.1016/j.chroma.2025.465914] [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: 02/15/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
Mass spectrometry (MS) based proteomics provides unbiased quantification of all proteins in plasma, which can dynamically reflect individual health states in real time. However, large-scale proteomics studies are constrained by the excessive dynamic range of plasma proteome and low throughput. Herein, two kinds of magnetic metal-organic frameworks (MOFs) modified with ion exchange functional groups (denoted as MHP-UiO-66-SAX and MHP-HKUST-1-SCX) were designed and fabricated to exhibit large protein adsorption capability, which were combined with an automated Liquid-handling System, thus realizing in-depth, high-throughput and automated proteomics studies. The constructed workflow could automatically complete the sample preparation before MS within only six hours and nearly a thousand protein groups per sample could be quantified. In the cohort study of nearly one hundred breast cancer neoadjuvant chemotherapy (NC) plasma samples, two differentially expressed proteins previously reported as biomarkers were related with the pathological complete response (PCR) of the breast cancer, demonstrating the feasibility of the developed technology for preparing large-scale clinical samples and exhibiting the potential application in monitoring the effect of chemotherapy.
Collapse
Affiliation(s)
- Xiaozhen Cui
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhihua Zhong
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China; School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Sen Xu
- Shanghai Research Institute of Chemical Industry, Shanghai 200062, China; Department of Clinical Laboratory, Zhongshan Hospital, Fudan University, Shanghai 200032,China
| | - Yini Pan
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China; School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xi Wang
- The Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Luobin Zhang
- The Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen 518020, China
| | - An He
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xueting Ye
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China; The Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Hua Cao
- The Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen 518020, China.
| | - Weibing Zhang
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Ruijun Tian
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
2
|
Zhang D, Zhang H, Yang Y, Jin Y, Chen Y, Wu C. Advancing tissue analysis: Integrating mass tags with mass spectrometry imaging and immunohistochemistry. J Proteomics 2025; 316:105436. [PMID: 40180154 DOI: 10.1016/j.jprot.2025.105436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/28/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
In biological and biomedical research, it's a crucial task to detect or quantify proteins or proteomes accurately across multiple samples. Immunohistochemistry (IHC) and spatial proteomics based on mass spectrometry imaging (MSI) are used to detect proteins in tissue samples. IHC can detect precisely but has a limited throughput, whereas MSI can simultaneously visualize thousands of specific chemical components but hindered by detailed protein annotation. Thereby, the introduction of mass tags may be adopted to expand the potential for integrating MSI and IHC. By enriching optical information for IHC and enhancing MS signals, mass tags can boost the accuracy of qualitative, localization, and quantitative detection of specific proteins in tissue sections, thereby widening the scope of protein detection and annotation results. Consequently, more comprehensive information regarding biological processes and disease states can be obtained, which aids in understanding complex biological processes and disease mechanisms and provides additional perspectives for clinical diagnosis and treatment. In the current review, we aim to discuss the role of different mass tags (e.g., mass tags based on inorganic molecules and organic molecules) in the combined application of MSI and IHC.
Collapse
Affiliation(s)
- Dandan Zhang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Hairong Zhang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Yuexin Yang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Ying Jin
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yingjie Chen
- Xiamen Key Laboratory for Clinical Efficacy and Evidence-Based Research of Traditional Chinese Medicine, Xiamen University, Xiamen 361102, China.
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
3
|
Starodubtseva N, Poluektova A, Tokareva A, Kukaev E, Avdeeva A, Rimskaya E, Khodzayeva Z. Proteome-Based Maternal Plasma and Serum Biomarkers for Preeclampsia: A Systematic Review and Meta-Analysis. Life (Basel) 2025; 15:776. [PMID: 40430203 PMCID: PMC12113278 DOI: 10.3390/life15050776] [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: 04/04/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Proteomics has emerged as a transformative tool in biomedical research, enabling comprehensive characterization of protein profiles in complex biological systems. In preeclampsia (PE) research, quantitative proteomic analyses of plasma and serum have revealed critical insights into disease mechanisms and potential biomarkers. Through a systematic review of 17 studies (2009-2024), we identified 561 differentially expressed plasma/serum proteins (p < 0.05) in PE patients versus healthy controls, with 122 proteins consistently replicated across ≥2 independent studies. Stratified analysis by clinical subtype (early-vs. late-onset PE) demonstrated both concordant and divergent protein expression patterns, reflecting heterogeneity in PE pathophysiology, methodological variations (e.g., sample processing, proteomic platforms), and differences between discovery-phase and targeted validation studies. The trimester-specific biomarker panels proposed here offer a framework for future large-scale, multicenter validation. By integrating advanced proteomic technologies with standardized preanalytical and analytical protocols, these findings advance opportunities for early prediction (first-trimester biomarker signatures); mechanistic insight (complement system involvement); and personalized management (subtype-specific therapeutic targets). This work underscores the potential of proteomics to reshape PE research, from molecular discovery to clinical translation, ultimately improving outcomes for this leading cause of maternal and perinatal morbidity.
Collapse
Affiliation(s)
- Natalia Starodubtseva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Alina Poluektova
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Alisa Tokareva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Evgenii Kukaev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Anna Avdeeva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| | - Elena Rimskaya
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
- Lebedev Physical Institute, 119991 Moscow, Russia
| | - Zulfiya Khodzayeva
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.P.); (A.T.); (E.K.); (A.A.); (E.R.); (Z.K.)
| |
Collapse
|
4
|
Lin ZT, Jana B, Korupolu S, Kong Y, Liu G, Dong Y, Li Y, Zhang Q, Shou W, Upadhyay P, Wang Z, Ran Z, Wu MX. Wearable Photonic Device for Multiple Biomarker Sampling and Detection without Blood Draws. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2416240. [PMID: 40326959 DOI: 10.1002/adma.202416240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/25/2025] [Indexed: 05/07/2025]
Abstract
Needle-based blood draws or phlebotomy practice in clinics for centuries, often causing pain, discomfort, and inconvenience. Here, a wearable photonic device is presented by integrating a microlens array (MLA) and an optic microneedle array (OMNA) functionalized with immunobinding for safe and needle-free biomarker sampling and detection. The MLA-integrated OMNA amplifies and transmits LED light at 595 nm into skin through the OMNA, bypassing the light-absorbing melanin in the epidermal layer, and evenly distributing it in the capillary-enriched dermis independent of the skin colors. The 595 nm light is absorbed by hemoglobin (Hb) and oxygen-Hb within the capillaries, triggering thermal dilation of capillaries without damaging them or causing petechiae. The light illumination remarkably increases in the concentrations of various blood biomarkers in the skin through biomarker extravasation. These biomarkers bound specifically to the capture antibodies on OMNA with each microneedle covalently immobilized with one specific antibody. The OMNA is extensively modified to amplify the immunobinding signals and achieve sensitivity superior to that of enzyme-linked immunosorbent assay (ELISA) kits. As proof of concept, the functionality of the prototype for minimally invasive sampling and precise multiplexed blood biomarker detection in two mouse models is validated to quantify acute inflammation and specific antibody production.
Collapse
Affiliation(s)
- Zuan-Tao Lin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Biswabandhu Jana
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Sandeep Korupolu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yifei Kong
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Guishi Liu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yan Dong
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yongli Li
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Quanwei Zhang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Wan Shou
- Department of Mechanical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Prabhat Upadhyay
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhilong Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Zihan Ran
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
5
|
Mohammed R, El Sheikh M, Hashim NT, Chaitanya NC, Suleiman A. Comparative Analysis of Salivary Tumor Marker CA-125 Among Oral Squamous Cell Carcinoma Patients and Healthy Individuals. Dent J (Basel) 2025; 13:194. [PMID: 40422614 DOI: 10.3390/dj13050194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/24/2025] [Accepted: 04/27/2025] [Indexed: 05/28/2025] Open
Abstract
Background/Objectives: In Sudan, oral cancer is one of the top ten most common cancers, with OSCC representing the majority of the cases. To date, despite the fact that saliva can be collected simply and non-invasively, there is no approved salivary tumor marker for OSCC. This study aimed to investigate the reliability of salivary CA-125 as a tumor marker for OSCC by measuring and comparing its level among OSCC and healthy individuals as well as its level across different histopathological grades. Methods: A total of 100 subjects were enrolled; 50 were patients with OSCC, while the other 50 were matched healthy individuals. Non-stimulated whole saliva was collected before the administration of definitive treatment, and the concentration of salivary CA-125 was quantified using an automated immunoassay analyzer that employs a one-step sandwich fluorescent enzyme immunoassay (FEIA). Results: The level of salivary CA-125 was 342.65 U/mL in the cases group, which was significantly increased compared with 203.65 U/mL in the healthy controls (p = 0.017). Statistically significant differences in the level of salivary CA-125 among different histopathological grades were observed (p = 0.014). The sensitivity, specificity, accuracy, and positive and negative predictive values were 48%, 78%, 63%, 68.6%, and 60%, respectively. Conclusions: This study suggests that salivary CA-125 could serve as a potential tumor marker for OSCC. However, its clinical application requires further validation.
Collapse
Affiliation(s)
- Riham Mohammed
- RAK College of Dental Sciences, RAK Medical and Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates
| | - Mariam El Sheikh
- Faculty of Dentistry, University of Khartoum, Khartoum 11115, Sudan
| | - Nada Tawfig Hashim
- RAK College of Dental Sciences, RAK Medical and Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates
| | - Nallan Csk Chaitanya
- RAK College of Dental Sciences, RAK Medical and Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates
| | - Ahmed Suleiman
- Faculty of Dentistry, University of Khartoum, Khartoum 11115, Sudan
| |
Collapse
|
6
|
Liu M, Jiao F, He Y. Rapid classification of proteins suspended in an evaporated sessile drop. Talanta 2025; 294:128224. [PMID: 40311480 DOI: 10.1016/j.talanta.2025.128224] [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: 01/15/2025] [Revised: 04/20/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
Reliable and accessible technologies for biological sample analysis are crucial for rapid, accurate illness diagnosis in resource-limited settings. Conventional methods like mass spectrometry and enzyme-linked immunosorbent assay (ELISA) often face challenges of technological complexity and equipment immobility, leading to delayed diagnoses and elevated costs. This study introduces evaporative deposition patterns from sessile droplets as a simple, cost-effective approach for classifying proteins in protein-salt mixtures, leveraging quantitative texture analysis to achieve high diagnostic accuracy. Using mixtures of potassium chloride (KCl) or sodium chloride (NaCl) with Bovine Serum Albumin (BSA) or Ovalbumin (OVA) as model systems, we varied salt (0.25-2 wt%) and protein (0.5-1.5 wt%) concentrations, observing distinct deposition morphologies such as coffee rings, cruciform/dendritic crystals at high salt levels, and volcano-shaped /spoke-like patterns at low or high protein levels. A novel quantitative framework integrating radial density distributions, gray-level co-occurrence matrix (GLCM) texture features, and receiver operating characteristic (ROC) curves achieved a 96.74 % classification accuracy for NaCl-protein mixtures, surpassing traditional texture analysis in simplicity and efficacy. These findings demonstrate the potential of evaporative deposition analysis as a simple tool for distinguishing molecular mixtures, laying the groundwork for future diagnostic applications. Unlike prior qualitative studies, this work provides a robust, quantitative method for protein classification, advancing the understanding of protein-salt interactions.
Collapse
Affiliation(s)
- Maoxi Liu
- School of Chemical Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Feng Jiao
- School of Chemical Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Yongqing He
- Chongqing Key Laboratory of Micro-Nano System and Intelligent Transduction, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Chongqing, 400067, China.
| |
Collapse
|
7
|
Nishigaki A, Ishikawa H, Nishiguchi Y, Tachibana K, Kato N, Matsuda K, Mori Y, Matsuyama H, Matsuura K, Ii Y, Wakita H, Oikawa S, Tomimoto H, Shindo A. Alpha-1-acid glycoprotein as a potential serum biomarker for cerebral amyloid angiopathy. J Alzheimers Dis 2025:13872877251333802. [PMID: 40261388 DOI: 10.1177/13872877251333802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BackgroundCerebral amyloid angiopathy (CAA) is a form of cerebral small vessel disease (SVD) associated with Alzheimer's disease, intracerebral hemorrhage, and cognitive decline. Despite its clinical significance, no reliable serum biomarker exists for early diagnosis or monitoring of disease progression.ObjectiveThis study hypothesizes that α1-acid glycoprotein (α1-AGP) and other serum biomarkers can aid CAA diagnosis and assessment using gel-based mass spectrometry. A comparative analysis was performed to investigate associations between serum biomarkers and radiological scores.MethodsSerum proteins from individuals with probable or possible CAA (n = 10), classified using the modified Boston criteria, and age-matched controls (n = 10) were analyzed via two-dimensional differential gel electrophoresis (2D-DIGE) and matrix-assisted laser desorption/ionization time-of-flight tandem mass spectrometry (MALDI-TOF/TOF-MS). Candidate proteins were validated using enzyme-linked immunosorbent assay (ELISA). Outcome measures included biomarker diagnostic accuracy, assessed by receiver operating characteristic (ROC) curve analysis, and correlations between α1-AGP levels and CAA-SVD scores.ResultsFour proteins-hemopexin, complement C3, complement C9, and α1-AGP-were significantly elevated, while apolipoprotein A-1 was reduced in the CAA group. ELISA confirmed higher α1-AGP levels in individuals with CAA (p < 0.0001). ROC analysis demonstrated that α1-AGP could indicate the presence of CAA with a sensitivity and specificity of 1.00 (95%CI: 1.000, 1.000). Additionally, α1-AGP levels correlated with the CAA-SVD score (R² = 0.783).Conclusionsα1-AGP may serve as a novel serum biomarker for CAA. Larger cohorts and external validation are required to substantiate these findings and determine their clinical relevance.
Collapse
Affiliation(s)
- Akisato Nishigaki
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidehiro Ishikawa
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yamato Nishiguchi
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kei Tachibana
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Natsuko Kato
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kana Matsuda
- Department of Dementia Prevention and Therapeutics, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yurie Mori
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hirofumi Matsuyama
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Keita Matsuura
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yuichiro Ii
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hideaki Wakita
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Shinji Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
- Department of Dementia Prevention and Therapeutics, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
8
|
Chen Z, Peng T, Zhong M, Zhang Y, Zhang Y, Hou Q, Peng T, Yang X, Zhou H, Liu L, Han M, Tang H, He L, Li J, Niu H, Xu K. Integrated metabolomics and proteomics analysis in children with cerebral palsy exposed to botulinum toxin-A. Pediatr Res 2025:10.1038/s41390-025-04038-5. [PMID: 40247116 DOI: 10.1038/s41390-025-04038-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/13/2024] [Accepted: 03/16/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND We previously examined plasma metabolic changes before and after botulinum toxin-A injections of cerebral palsy (CP) children and showed that the glycine, serine and threonine metabolism may play a key role in neuritogenesis. This study analysed untargeted metabolomics combined with proteomics of plasma to discussed which substances are meaningfully changed, to what extent they affect the effects of action. METHODS Blood samples were collected from 91 children with spastic CP at 4 time points: pre-injection (T1), 1 month post-injection (T2), 3 months post-injection (T3) and 6 months post-injection (T4). Differentially changed metabolites and proteins were selected, and co-expression pathways were constructed to explore the key molecular processes. RESULTS A total of 674 proteins and 354 metabolites were identified. The differential metabolites were mainly involved in the linoleic acid metabolism, beta-Alanine metabolism, citrate cycle, pyruvate metabolism and glycolysis or gluconeogenesis. Differential proteins were primarily associated with glucose metabolism, lipid metabolism, immune and inflammation responses. Co-expression pathways showed that ECM-receptor interaction, complement and coagulation cascades, glycolysis or gluconeogenesis, pyruvate metabolism, and linoleic acid metabolism were the main pathways. CONSLUSIONS Our results indicated the botulinum toxin-A predominantly activated the glucose metabolism, lipid metabolism, and immune and inflammation responses, and energy metabolism changed significantly in this process. TRIAL REGISTRATION DETAILS ChiCTR2000033800, Research on the mechanism of botulinum toxin relieving spasticity in children with cerebral palsy. Approval No. 202023041. Registered 13 June 2020, http://www.chictr.org.cn/showproj.html?proj=52267 . IMPACT STATEMENT This is the first study that combined dynamic metabolomics and proteomics analysis to investigate the molecular changes in children with spastic cerebral palsy after botulinum toxin-A injections, which might provide a theoretical reference for the further subsequent study for targets to increase the efficacy and prolong the duration of botulinum toxin-A, and would be a valuable resource for the metabolomics and proteomics field in this group.
Collapse
Affiliation(s)
- Zhaofang Chen
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Tingting Peng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Mengru Zhong
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Yage Zhang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Yuan Zhang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, Shanghai, China
| | - Qingfen Hou
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, Guangzhou, China
| | - Tingting Peng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Xubo Yang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Hongyu Zhou
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Mingshan Han
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Jinling Li
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China
| | - Huiran Niu
- Genechem Biotechnology Co., Ltd, Shanghai, Shanghai, China
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangzhou, China.
| |
Collapse
|
9
|
Gadour E, Miutescu B, Hassan Z, Aljahdli ES, Raees K. Advancements in the diagnosis of biliopancreatic diseases: A comparative review and study on future insights. World J Gastrointest Endosc 2025; 17:103391. [PMID: 40291132 PMCID: PMC12019128 DOI: 10.4253/wjge.v17.i4.103391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/19/2025] [Accepted: 03/08/2025] [Indexed: 04/14/2025] Open
Abstract
Owing to the complex and often asymptomatic presentations, the diagnosis of biliopancreatic diseases, including pancreatic and biliary malignancies, remains challenging. Recent technological advancements have remarkably improved the diagnostic accuracy and patient outcomes in these diseases. This review explores key advancements in diagnostic modalities, including biomarkers, imaging techniques, and artificial intelligence (AI)-based technologies. Biomarkers, such as cancer antigen 19-9, KRAS mutations, and inflammatory markers, provide crucial insights into disease progression and treatment responses. Advanced imaging modalities include enhanced computed tomography (CT), positron emission tomography-CT, magnetic resonance cholangiopancreatography, and endoscopic ultrasound. AI integration in imaging and pathology has enhanced diagnostic precision through deep learning algorithms that analyze medical images, automate routine diagnostic tasks, and provide predictive analytics for personalized treatment strategies. The applications of these technologies are diverse, ranging from early cancer detection to therapeutic guidance and real-time imaging. Biomarker-based liquid biopsies and AI-assisted imaging tools are essential for non-invasive diagnostics and individualized patient management. Furthermore, AI-driven models are transforming disease stratification, thus enhancing risk assessment and decision-making. Future studies should explore standardizing biomarker validation, improving AI-driven diagnostics, and expanding the accessibility of advanced imaging technologies in resource-limited settings. The continued development of non-invasive diagnostic techniques and precision medicine approaches is crucial for optimizing the detection and management of biliopancreatic diseases. Collaborative efforts between clinicians, researchers, and industry stakeholders will be pivotal in applying these advancements in clinical practice.
Collapse
Affiliation(s)
- Eyad Gadour
- Multiorgan Transplant Centre of Excellence, Liver Transplantation Unit, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia
- Internal Medicine, Zamzam University College, School of Medicine, Khartoum 11113, Sudan
| | - Bogdan Miutescu
- Department of Gastroenterology and Hepatology, Victor Babes University of Medicine and Pharmacy, Timisoara 300041, Romania
- Advanced Regional Research Center in Gastroenterology and Hepatology, Victor Babes University of Medicine and Pharmacy, Timisoara 30041, Romania
| | - Zeinab Hassan
- Department of Internal Medicine, Stockport Hospitals NHS Foundation Trust, Manchester SK2 7JE, United Kingdom
| | - Emad S Aljahdli
- Gastroenterology Division, King Abdulaziz University, Faculty of Medicine, Jeddah 21589, Saudi Arabia
- Gastrointestinal Oncology Unit, King Abdulaziz University Hospital, Jeddah 22252, Saudi Arabia
| | - Khurram Raees
- Department of Gastroenterology and Hepatology, Royal Blackburn Hospital, Blackburn BB2 3HH, United Kingdom
| |
Collapse
|
10
|
Tretiak S, Maia TM, Van Haver D, Staes A, Devos S, Rijsselaere T, Goossens E, Van Immerseel F, Impens F, Antonissen G. Blood proteome profiling for biomarker discovery in broilers with necrotic enteritis. Sci Rep 2025; 15:12895. [PMID: 40234672 PMCID: PMC12000508 DOI: 10.1038/s41598-025-97783-w] [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: 03/08/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
Analysis of the blood proteome allows identification of proteins related to changes upon certain physiological conditions. The pathophysiology of necrotic enteritis (NE) has been extensively studied. While intestinal changes have been very well documented, data addressing NE-induced alterations in the blood proteome are scant, although these might have merit in diagnostics. In light of recent technological advancements in proteomics and pressing need for tools to access gut health, the current study employs mass-spectrometry (MS)-based proteomics to identify biomarkers for gastrointestinal health of chickens. Here, we report findings of an untargeted proteomics investigation conducted on blood plasma in chickens under NE challenge. Two MS-strategies were used for analysis: conventional data dependent acquisition coupled to standard nanoflow liquid chromatography (LC) (nano-DDA) and recently-developed data independent acquisition coupled to an Evosep One LC system (Evo-DIA). Despite superior completeness and quantification of the Evo-DIA-acquired data, high degree of agreement in identification and quantification was observed between both approaches. Additionally, we identified 15 differentially expressed proteins (shared by nano-DDA and Evo-DIA) that represent responses of animals to infection and may serve as potential biomarkers. Experimental validation through ELISA immunoassays and targeted MS for selected regulated proteins (CFD, HPS5, and MASP2) confirmed medium-to-high levels of inter-protein correlation. A GSEA analysis revealed enrichment in a number of processes related to adaptive and humoral immunity, immune activation and response in infected animals. Data are available via ProteomeXchange with identifiers PXD050461, PXD050473, and PXD061607.
Collapse
Affiliation(s)
- Svitlana Tretiak
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, B-9820, Belgium
- Impextraco NV, Wiekevorstsesteenweg 38, Heist-op-den-Berg, 2220, Belgium
| | - Teresa Mendes Maia
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, B-9052, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, B-9052, Belgium
- VIB Proteomics Core, Technologiepark-Zwijnaarde 75, B9052, Ghent, Belgium
| | - Delphi Van Haver
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, B-9052, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, B-9052, Belgium
- VIB Proteomics Core, Technologiepark-Zwijnaarde 75, B9052, Ghent, Belgium
| | - An Staes
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, B-9052, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, B-9052, Belgium
- VIB Proteomics Core, Technologiepark-Zwijnaarde 75, B9052, Ghent, Belgium
| | - Simon Devos
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, B-9052, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, B-9052, Belgium
- VIB Proteomics Core, Technologiepark-Zwijnaarde 75, B9052, Ghent, Belgium
| | - Tom Rijsselaere
- Impextraco NV, Wiekevorstsesteenweg 38, Heist-op-den-Berg, 2220, Belgium
| | - Evy Goossens
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, B-9820, Belgium
| | - Filip Van Immerseel
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, B-9820, Belgium
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, B-9052, Belgium.
- Department of Biomolecular Medicine, Ghent University, Ghent, B-9052, Belgium.
- VIB Proteomics Core, Technologiepark-Zwijnaarde 75, B9052, Ghent, Belgium.
| | - Gunther Antonissen
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, B-9820, Belgium.
| |
Collapse
|
11
|
Reasoner EA, Chan HJ, Aballo TJ, Plouff KJ, Noh S, Ge Y, Jin S. In Situ Metal-Organic Framework Growth in Serum Encapsulates and Depletes Abundant Proteins for Integrated Plasma Proteomics. ACS NANO 2025; 19:13968-13981. [PMID: 40168247 PMCID: PMC12047221 DOI: 10.1021/acsnano.4c18028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Protein biomarkers in human serum provide critical insights into various physiological conditions and diseases, enabling early diagnosis, prognosis, and personalized treatment. However, detecting low-abundance protein biomarkers is challenging due to the presence of highly abundant proteins that make up ∼99% of the plasma proteome. Here, we report the use of in situ metal-organic framework (MOF) growth in serum to effectively deplete highly abundant serum proteins for integrated proteomic analysis. Through biomolecule-mediated nucleation of a zeolitic imidazolate framework (ZIF-8), abundant plasma proteins are selectively encapsulated within ZIF-8 and removed from serum via centrifugation, leaving a depleted protein fraction in the supernatant. Bottom-up proteomics analysis confirmed significant depletion of the topmost abundant proteins, many at depletion levels exceeding 95%. Such depletion enabled the identification of 277 total proteins in the supernatant (uncaptured) fraction in a single-shot analysis, including 54 proteins that were only identified after depletion, 12 drug targets, and many potential disease biomarkers. Top-down proteomics characterization of the captured and uncaptured protein fractions at the proteoform-level confirmed this method is not biased toward any specific proteoform of individual proteins. These results demonstrate that in situ MOF growth can selectively and effectively deplete high-abundance proteins from serum in a simple, low cost, one-pot synthesis to enable integrated top-down and bottom-up proteomic analysis of serum protein biomarkers.
Collapse
Affiliation(s)
- Emily A. Reasoner
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Hsin-Ju Chan
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Timothy J. Aballo
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Kylie J. Plouff
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Seungwoo Noh
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Song Jin
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
12
|
Dimayacyac-Esleta BRT, Mira FD, Zarate LM, Porras BJO, Juntilla DLA, Suñga LBL, Pondevida VB, Naval SS, Sayo TMS, Luna HGC, Prieto EI. Discovery of Key Candidate Protein Biomarkers in Early-Stage Nonsmall Cell Lung Carcinoma through Quantitative Proteomics. J Proteome Res 2025; 24:1701-1714. [PMID: 40014793 DOI: 10.1021/acs.jproteome.4c00764] [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: 03/01/2025]
Abstract
Difficulties in early-stage diagnosis are among the factors contributing to the high mortality of nonsmall cell lung carcinoma (NSCLC) patients. Unfortunately, diagnostic biomarkers are currently lacking, limiting options in the clinic. To discover proteins that have potential for biomarker applications, we performed an in-depth quantitative proteomic analysis on a cohort of Filipino early-stage NSCLC lung adenocarcinoma (LUAD) patients. Differentially expressed proteins (DEPs) were obtained by using tandem mass tag (TMT) labeling and mass spectrometry (MS)-based quantitative proteomics. A total of 6240 quantified proteins were identified with 3155 significantly upregulated and 1248 significantly downregulated. Integration of the proteomic result with curated transcriptome data allowed the identification of 33 proteins with biomarker potential. This study also provided insights into relevant pathways in NSCLC LUAD, such as protein translation and metabolic pathways. Interestingly, all of the enzymes in the hexosamine biosynthetic pathway (HBP) are found to be upregulated, suggesting its important role in NSCLC LUAD. It is worthwhile to look at the potential of targeting the metabolic vulnerability of NSCLC LUAD as a new strategy in drug development. All MS data were deposited into ProteomeXchange with the identifier PXD050598.
Collapse
Affiliation(s)
| | - Ferdinand D Mira
- Institute of Chemistry, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Lorenzo M Zarate
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Ben Joshua O Porras
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Dave Laurence A Juntilla
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Lara Beatrice L Suñga
- Institute of Chemistry, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Venus B Pondevida
- Institute of Chemistry, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Sullian S Naval
- Lung Center of the Philippines, Metro Manila 1100, Philippines
| | | | | | - Eloise I Prieto
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Metro Manila 1101, Philippines
| |
Collapse
|
13
|
Yuan M, Liu A, Xu B, Sales K, Karnik S, Voelker T, Salha D, Zimmer J, O’Dell M, Gorityala S, Hays A, Lester T, Reynolds G, Tary-Lehmann M, Yu M, Roberge M, Malone K, Patel V, Love I, Lin J, Diaz M, Xu T, Garofolo W, McGregor J, Leskovar A, Kernstock R, Pellerin M, Brown M, Spytko A, Lowes S, Ambrose D, Dufield D, Kane C, Veeramachaneni R, Luna M, Warrino D, Dwivedi V, Xu A, Hyer E, Iles T, Majumdar R, Sikkema D, Thomas E, Carlsson A, Dakappagari N, Riccitelli N, Marco CD, Bouhajib M, Iordachescu A, Sanghvi M, Barton H, Lavelle A, Dompkowski E, Rundlett S, Matys K, Sangster T, Turksma A, Gu W, Liu J, Hoffpauir B, Rocha A, Pirro J, Bergeron J, O’Brien K, Fang X, Dong K, Yamashita J. Recommendations on biomarker assay validation (BAV) in tissues by GCC. Bioanalysis 2025; 17:429-438. [PMID: 40248960 PMCID: PMC12026151 DOI: 10.1080/17576180.2025.2471243] [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: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 04/19/2025] Open
Abstract
Biomarker analysis enables a deep understanding of physiological and biological processes and offers insights into pathological disease states and conditions. When measured in tissues, the spatial distribution of biomarkers may be evaluated. To meet regulatory and sponsor requirements, guidance on the approach to validation and the parameters to be evaluated is essential. The main goals of this GCC white paper are to disseminate the survey results discussed during the 16th& 17thGCC Closed Forums (2023 & 2024) and to provide recommendations from the GCC members on technical and regulatory considerations for the bioanalysis of biomarkers in tissues.
Collapse
Affiliation(s)
- Moucun Yuan
- PPD, part of Thermo Fisher Scientific, Richmond, VA, USA
| | | | - Bin Xu
- Accurant Biotech, Cranbury, NJ, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Mathilde Yu
- Cerba Research Canada Formally CIRION BioPharma Research Inc, a Cerba Research Company, Laval, QC, Canada
| | - Martin Roberge
- Cerba Research Canada Formally CIRION BioPharma Research Inc, a Cerba Research Company, Laval, QC, Canada
| | | | | | - Iain Love
- Charles River Laboratories, EdinburgTranent, UK
| | | | - Manisha Diaz
- Eurofins Viracor BioPharma Services, Lenexa, KS, USA
| | - Tao Xu
- Frontage Laboratories, Exton, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Allan Xu
- Keystone Bioanalytical, North Wales, PA, USA
| | | | | | | | | | | | | | - Naveen Dakappagari
- Navigate BioPharma Services, Inc. (A Novartis Subsidiary), Carlsbad, CA, USA
| | - Nathan Riccitelli
- Navigate BioPharma Services, Inc. (A Novartis Subsidiary), Carlsbad, CA, USA
| | | | | | | | | | - Hollie Barton
- PPD, part of Thermo Fisher Scientific, Richmond, VA, USA
| | - Amy Lavelle
- PPD, part of Thermo Fisher Scientific, Richmond, VA, USA
| | | | | | | | | | | | - Weihua Gu
- Shanghai Xihua Scientific Co, Shanghai, China
| | - Jia Liu
- Shanghai Xihua Scientific Co, Shanghai, China
| | | | | | | | | | | | | | - Kelly Dong
- United-Power Pharma Tech Co, Beijing, China
| | | |
Collapse
|
14
|
Luo Y, Liu X, Liu Y, Xu T, Zhang X. Enhancing biosensing of trace tau protein in clinical samples via emergence macroscopic directed aggregation. Biosens Bioelectron 2025; 273:117157. [PMID: 39824699 DOI: 10.1016/j.bios.2025.117157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/26/2024] [Accepted: 01/10/2025] [Indexed: 01/20/2025]
Abstract
Alzheimer's disease (AD) is an irreversible neurodegenerative disorder that poses a significant risk to human health and well-being. The high cost and invasiveness of neuroimaging and cerebrospinal fluid (CSF) analysis underscores the necessity for accessible early screening via blood samples. In this study, we developed an ultrasound-based strategy for emergent macroscopic that enhances the acoustic response enrichment of specific proteins by introducing functionalized microspheres. This customized macroscopic-directed aggregation method combines protein enrichment with specific fluorescent antibody recognition, enabling quantitative detection characterized by high sensitivity and specificity for tau proteins in clinical samples. In comparison with previously reported biosensing platforms, this strategy achieves indirectly driven clustering of tau proteins by integrating an acoustic resonant chamber, realizing facile and low-cost enrichment detection of protein without the need for amplification. Results demonstrate that this method exhibits a linear detection range from 1 pg/mL to 10 ng/mL, with a detection limit of 0.183 pg/mL. AD patients and healthy individuals were successfully distinguished with an accuracy of 90.9%. This ultrasonic biosensing strategy based on protein aggregation exhibits potential as a valuable tool for early screening of AD.
Collapse
Affiliation(s)
- Yong Luo
- Longgang Central Hospital of Shenzhen, Longgang Clinical Institute of Shantou University Medical College, Shenzhen, Guangdong, 518060, PR China; Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Xingyun Liu
- School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Yibiao Liu
- Longgang Central Hospital of Shenzhen, Longgang Clinical Institute of Shantou University Medical College, Shenzhen, Guangdong, 518060, PR China.
| | - Tailin Xu
- Synthetic Biology Research Center, Institute for Advanced Study (IAS), Shenzhen University, Shenzhen, Guangdong, 518060, PR China; School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, Guangdong, 518060, PR China.
| | - Xueji Zhang
- Synthetic Biology Research Center, Institute for Advanced Study (IAS), Shenzhen University, Shenzhen, Guangdong, 518060, PR China; School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, Guangdong, 518060, PR China.
| |
Collapse
|
15
|
Rosa-Fernandes L, Santiago VF, da Silva-Santos Y, Lopes TT, Peixoto EPM, Rodrigues SAM, Marinho CRF, Palmisano G, Epiphanio S. Serum Proteomics of Experimental Malaria-Associated ARDS Reveals a Regulation of Acute-Phase Response Proteins. J Immunol Res 2025; 2025:5642957. [PMID: 40160901 PMCID: PMC11955258 DOI: 10.1155/jimr/5642957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/07/2025] [Indexed: 04/02/2025] Open
Abstract
Malaria is a parasitic infectious disease considered a public health problem. Acute respiratory distress syndrome (ARDS) is a complication in malaria-infected individuals with a high mortality rate (80% to 100%) and can occur before, during, or after antimalarial drug treatment. Although inflammation and epithelial/endothelial injury pathways have been determined through these studies, specific circulating malaria-associated ARDS markers have not yet been established. We applied a quantitative mass spectrometry (MS)-based proteomic approach to identify altered molecular pathways in a mouse model of malaria-associated ARDS. Acute-phase response (APR) proteins were regulated in the ARDS group, suggesting their potential involvement in the development of the syndrome. They may serve as biomarkers when analyzed alongside other proteins that require further investigation. Additionally, the regulation of APR proteins in the ARDS group provides valuable insights into the pathophysiology of ARDS, contributing to a better understanding of the syndrome.
Collapse
Affiliation(s)
- Lívia Rosa-Fernandes
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Verônica Feijoli Santiago
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Yasmin da Silva-Santos
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Tissiane Tarosso Lopes
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Sabrina Epiphanio
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Huo C, Zhang C, Lu J, Su X, Qi X, Guo Y, Bao Y, Jia H, Cao G, Na R, Zhang W, Li X. A deep learning tissue classifier based on differential co-expression genes predicts the pregnancy outcomes of cattle†. Biol Reprod 2025; 112:550-562. [PMID: 39832283 DOI: 10.1093/biolre/ioaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/13/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
Economic losses in cattle farms are frequently associated with failed pregnancies. Some studies found that the transcriptomic profiles of blood and endometrial tissues in cattle with varying pregnancy outcomes display discrepancies even before artificial insemination (AI) or embryo transfer (ET). In the study, 330 samples from seven distinct sources and two tissue types were integrated and divided into two groups based on the ability to establish and maintain pregnancy after AI or ET: P (pregnant) and NP (nonpregnant). By analyzing gene co-variation and employing machine learning algorithms, the objective was to identify genes that could predict pregnancy outcomes in cattle. Initially, within each tissue type, the top 100 differentially co-expressed genes (DCEGs) were identified based on the analysis of changes in correlation coefficients and network topological structure. Subsequently, these genes were used in models trained by seven different machine learning algorithms. Overall, models trained on DCEGs exhibited superior predictive accuracy compared to those trained on an equivalent number of differential expression genes. Among them, the deep learning models based on differential co-expression genes in blood and endometrial tissue achieved prediction accuracies of 91.7% and 82.6%, respectively. Finally, the importance of DCEGs was ranked using SHapley Additive exPlanations (SHAP) and enrichment analysis, identifying key signaling pathways that influence pregnancy. In summary, this study identified a set of genes potentially affecting pregnancy by analyzing the overall co-variation of gene connections between multiple sources. These key genes facilitated the development of interpretable machine learning models that accurately predict pregnancy outcomes in cattle.
Collapse
Affiliation(s)
- Chenxi Huo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Chuanqiang Zhang
- Inner Mongolia SK·Xing Animal Breeding and Breeding Biotechnology Research Institute Co., Ltd, Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Jing Lu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaofeng Su
- Inner Mongolia Yili Industrial Group Co., Ltd, Hohhot, China
| | - Xiaoxia Qi
- Inner Mongolia SK·Xing Animal Breeding and Breeding Biotechnology Research Institute Co., Ltd, Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Yaqiang Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yanchun Bao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Hongxia Jia
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Guifang Cao
- Inner Mongolia SK·Xing Animal Breeding and Breeding Biotechnology Research Institute Co., Ltd, Hohhot, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Risu Na
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Engineering Research Center of Genomic Big Data for Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| | - Wenguang Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Engineering Research Center of Genomic Big Data for Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| | - Xihe Li
- Inner Mongolia SK·Xing Animal Breeding and Breeding Biotechnology Research Institute Co., Ltd, Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
- Inner Mongolia Yili Industrial Group Co., Ltd, Hohhot, China
| |
Collapse
|
17
|
Hong CT, Chung CC, Hsieh YC, Chan L. Plasma extracellular vesicle pathognomonic proteins as the biomarkers of the progression of Parkinson's disease. Biosci Trends 2025; 19:116-124. [PMID: 39924179 DOI: 10.5582/bst.2024.01369] [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: 02/11/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder for which reliable blood biomarkers to predict disease progression remain elusive. Plasma extracellular vesicles (EVs) have gained attention as a promising biomarker platform due to their stability and ability to cross the blood-brain barrier. This study explored the potential of EV-cargo proteins, specifically α-synuclein, tau, and β-amyloid, as biomarkers of PD progression. A cohort of 55 people with PD (PwP) and 58 healthy controls (HCs) underwent annual assessments of plasma EV proteins, cognition, and motor symptoms. EVs were isolated and validated using standardized methods, with pathognomonic proteins quantified via immunomagnetic reduction assays. Associations between biomarker changes and clinical symptom progression were analyzed. Over an average of 3.96 visits for PwP and 2.25 visits for HCs, PwP exhibited a distinct pattern of plasma EV protein changes linked to motor symptom progression, particularly in the Unified PD Rating Scale (UPDRS) part II score. Notably, changes in plasma EV α-synuclein levels were significantly correlated with changes in motor and cognitive symptoms, suggesting its central role in disease progression. These findings highlight the potential of plasma EV biomarkers, especially α-synuclein, as indicators of ongoing pathogenesis and as candidates for evaluating α-synuclein-targeted therapies in PD.
Collapse
Affiliation(s)
- Chien-Tai Hong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Chen-Chih Chung
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chen Hsieh
- Ph.D in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| |
Collapse
|
18
|
Pei J, Qiu H, Wang W, Wang Y, Wang M, Wang D, Li J, Qin Y. The Contribution and Perspectives of Proteomics to Epithelial Ovarian Cancer. Proteomics Clin Appl 2025; 19:e202300220. [PMID: 39865556 DOI: 10.1002/prca.202300220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/27/2024] [Accepted: 01/10/2025] [Indexed: 01/28/2025]
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy which mainly consists of serous, mucinous, clear cell, and endometrioid subtypes. Due to the lack of classic symptoms at an early stage, EOC usually presented as advanced tumors with local and/or distant metastasis. Although a large portion of EOC was initially platinum-sensitive, most patients would acquire resistance to common chemotherapeutic agents. These aforementioned issues lead to a challenge for clinical treatments and unsatisfying outcomes. Previous studies have demonstrated the genetic features of EOC are hard to target and the alterations at DNA and RNA levels are not fully represented at the protein expression profiles which made it more complex. In recent years, a panel of studies attempted to explore the key proteins involved in the development and progression of EOC using high-throughput proteomic technologies. We herein summarized them to provide a full view of this topic.
Collapse
Affiliation(s)
- Jiayu Pei
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Haifeng Qiu
- Department of Gynecology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wenjia Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yulu Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Min Wang
- Department of Gynecology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Dian Wang
- Department of Gynecology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yanru Qin
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
19
|
Yu H, Song J, Li J, Qi Y, Fan Z, Liu Q, Yu L, Song J, Dong H. Applications of Digital Enzyme-Linked Immunosorbent Assays in Ophthalmology. Cell Biochem Biophys 2025; 83:215-220. [PMID: 39333452 PMCID: PMC11870898 DOI: 10.1007/s12013-024-01515-2] [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] [Accepted: 08/28/2024] [Indexed: 09/29/2024]
Abstract
Digital enzyme-linked immunosorbent assays (dELISAs) very sensitively detect biomarkers that cannot be measured using traditional methods. The molecules are confined within a small volume, their counts accurately computed, and the results rapidly delivered. Digital ELISAs find many applications. In recent years, such ELISAs have become increasingly used to aid ophthalmological diagnoses and treatments, and have revolutionized the field. This article reviews the applications of dELISAs in clinical practice, especially in the sphere of ophthalmology.
Collapse
Affiliation(s)
- He Yu
- Department of Ophthalmology, the Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Jiaping Song
- Department of Clinical Laboratory, the Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Junrong Li
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Yuanyuan Qi
- Department of Ophthalmology, the Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Zhe Fan
- Department of General Surgery, the Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Qiming Liu
- Department of Ophthalmology, the Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Liang Yu
- Department of Ophthalmology, the Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Jian Song
- Department of Ophthalmology, the Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - He Dong
- Department of Ophthalmology, the Third People's Hospital of Dalian, Dalian Medical University, Dalian, China.
| |
Collapse
|
20
|
Liu Q, Wang M, Dai X, Li S, Guo H, Huang H, Xie Y, Xu C, Liu Y, Tan W. Extreme Tolerance of Nanoparticle-Protein Corona to Ultra-High Abundance Proteins Enhances the Depth of Serum Proteomics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413713. [PMID: 39840619 PMCID: PMC11923864 DOI: 10.1002/advs.202413713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/15/2024] [Indexed: 01/23/2025]
Abstract
The serum nanoparticle-protein corona (NPC) provides specific disease information, thus opening a new avenue for high-throughput in-depth proteomics to facilitate biomarker discovery. Yet, little is known about the interactions between NPs and proteins, and its role in enhanced depth of serum proteomics. Herein, a series of protein spike-in experiments are conducted to systematically investigate protein depletion and enrichment during NPC formation. Proteomic depth is serum concentration-dependent, and NPC exhibits powerful tolerance to ultra-high abundant proteins. In addition, protein-protein interactions (PPI), especially those involving albumin, play a pivotal role in promoting proteomic depth. Furthermore, a triple-protein assay is established to interrogate the relationship between protein binding affinity and concentration. NPC formation is a product of balancing binding affinity, concentration, and PPI. Overall, this study elucidates how NPs achieve protein depletion and enrichment for enhanced serum proteomic depth to gain a better understanding of NPC as an essential tool of proteome profiling.
Collapse
Affiliation(s)
- Qiqi Liu
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Mengjie Wang
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Xin Dai
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
- School of Molecular MedicineHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhouZhejiang310024China
| | - Shuangqin Li
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Haoxiang Guo
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Haozhe Huang
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Yueli Xie
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Chenlu Xu
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Yuan Liu
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
- School of Molecular MedicineHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhouZhejiang310024China
| | - Weihong Tan
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of Medicineand College of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityHangzhouShanghai200240China
| |
Collapse
|
21
|
Wei L, Guo ZG, Wei J, Zhou Y, Sun W, Han C. Changes in rat plasma proteomes during the first week after birth. Front Vet Sci 2025; 12:1440716. [PMID: 40070918 PMCID: PMC11894578 DOI: 10.3389/fvets.2025.1440716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Blood plasma is the most informative body fluid, containing large amounts of substances that are released by active secretion or leakage from tissues and cells. Therefore, plasma changes reflect the body state. To explore changes in plasma during the early life of Wistar-rats, the plasma proteomes of newborn and first-week rats were investigated using liquid chromatography-tandem mass spectrometry. A total of 639 proteins were identified at both developmental stages and 570 proteins were used for quantitative analysis. The plasma of first-week rats, compared to that in newborn rats using label-free quantification, showed that the levels of 42 proteins significantly increased while those of 17 proteins decreased. Plasma proteomic patterns at both developmental stages can be easily separated using differential protein cluster analysis. Using the Ingenuity Pathway analysis tool, some pathways including LXR/RXR Activation, DHCR24 Signaling Pathway, Acute Phase Response Signaling, and Detoxification of Reactive Oxygen Species were significantly enhanced. Over 10 categories related to the development and functions were enriched. Plasma proteomes of first-week rats were distinct from those of newborn rats. These changes would make it easier for newborn rats to survive. This is the first study using liquid chromatography-tandem mass spectrometry to investigate newborn rat plasma proteome changes, providing a basis and clues for studying animal development.
Collapse
Affiliation(s)
- Lilong Wei
- Clinical Laboratory Center, China-Japan Friendship Hospital, Beijing, China
| | - Zheng-guang Guo
- Core Facility of Instruments, School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Wei
- Department of Pharmacy, Clinical Research Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yun Zhou
- Clinical Laboratory Center, China-Japan Friendship Hospital, Beijing, China
| | - Wei Sun
- Core Facility of Instruments, School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Chengwu Han
- Clinical Laboratory Center, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
22
|
Arabfard M, Behmard E, Maghsoudloo M, Dadgar E, Parvin S, Bagheri H. Identifying candidate RNA-seq biomarkers for severity discrimination in chemical injuries: A machine learning and molecular dynamics approach. Int Immunopharmacol 2025; 148:114090. [PMID: 39847951 DOI: 10.1016/j.intimp.2025.114090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/11/2025] [Accepted: 01/11/2025] [Indexed: 01/25/2025]
Abstract
INTRODUCTION Biomarkers play a crucial role across various fields by providing insights into biological responses to interventions. High-throughput gene expression profiling technologies facilitate the discovery of data-driven biomarkers through extensive datasets. This study focuses on identifying biomarkers in gene expression data related to chemical injuries by mustard gas, covering a spectrum from healthy individuals to severe injuries. MATERIALS AND METHODS The study utilized RNA-Seq data comprising 52 expression data samples for 54,583 gene transcripts. These samples were categorized into four classes based on the GOLD classification for chemically injured individuals: Severe (n = 14), Moderate (n = 11), Mild (n = 16), and healthy controls (n = 11). Data preparation involved examining an Excel file created in the R programming environment using MLSeq and devtools packages. Feature selection was performed using Genetic Algorithm and Simulated Annealing, with Random Forest algorithm employed for classification. Ab initio methods ensured computational efficiency and result accuracy, while molecular dynamics simulation acted as a virtual experiment bridging the gap between experimental and theoretical experiences. RESULTS A total of 12 models were created, each introducing a list of differentially expressed genes as potential biomarkers. The performance of models varied across group comparisons, with the Genetic Algorithm generally outperforming Simulated Annealing in most cases. For the Severe vs. Moderate group, GA achieved the best performance with an accuracy of 94.38%, recall of 91.64%, and specificity of 97.10%. The results highlight the effectiveness of GA in most group comparisons, while SA performed better in specific cases involving Moderate and Mild groups. These biomarkers were evaluated against the gene expression data to assess their expression changes between different groups of chemically injured individuals. Four genes were selected based on level expression for further investigation: CXCR1, EIF2B2, RAD51, and RXFP2. The expression levels of these genes were analyzed to determine their differential expression between the groups. CONCLUSION This study was designed as a computational effort to identify diagnostic biomarkers in basic biological system research. Our findings proposed a list of discriminative biomarkers capable of distinguishing between different groups of chemically injured individuals. The identification of key genes highlights the potential for biomarkers to serve as indicators of chemical injury severity, warranting further investigation to validate their clinical relevance and utility in diagnosis and treatment.
Collapse
Affiliation(s)
- Masoud Arabfard
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Esmaeil Behmard
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000 Sichuan, China
| | - Emad Dadgar
- Students' Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shahram Parvin
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hasan Bagheri
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Vo DK, Trinh KTL. Polymerase Chain Reaction Chips for Biomarker Discovery and Validation in Drug Development. MICROMACHINES 2025; 16:243. [PMID: 40141854 PMCID: PMC11944077 DOI: 10.3390/mi16030243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025]
Abstract
Polymerase chain reaction (PCR) chips are advanced, microfluidic platforms that have revolutionized biomarker discovery and validation because of their high sensitivity, specificity, and throughput levels. These chips miniaturize traditional PCR processes for the speed and precision of nucleic acid biomarker detection relevant to advancing drug development. Biomarkers, which are useful in helping to explain disease mechanisms, patient stratification, and therapeutic monitoring, are hard to identify and validate due to the complexity of biological systems and the limitations of traditional techniques. The challenges to which PCR chips respond include high-throughput capabilities coupled with real-time quantitative analysis, enabling researchers to identify novel biomarkers with greater accuracy and reproducibility. More recent design improvements of PCR chips have further expanded their functionality to also include digital and multiplex PCR technologies. Digital PCR chips are ideal for quantifying rare biomarkers, which is essential in oncology and infectious disease research. In contrast, multiplex PCR chips enable simultaneous analysis of multiple targets, therefore simplifying biomarker validation. Furthermore, single-cell PCR chips have made it possible to detect biomarkers at unprecedented resolution, hence revealing heterogeneity within cell populations. PCR chips are transforming drug development, enabling target identification, patient stratification, and therapeutic efficacy assessment. They play a major role in the development of companion diagnostics and, therefore, pave the way for personalized medicine, ensuring that the right patient receives the right treatment. While this tremendously promising technology has exhibited many challenges regarding its scalability, integration with other omics technologies, and conformity with regulatory requirements, many still prevail. Future breakthroughs in chip manufacturing, the integration of artificial intelligence, and multi-omics applications will further expand PCR chip capabilities. PCR chips will not only be important for the acceleration of drug discovery and development but also in raising the bar in improving patient outcomes and, hence, global health care as these technologies continue to mature.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea;
| | - Kieu The Loan Trinh
- Bionano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
24
|
Mojgani N, Ashique S, Moradi M, Bagheri M, Garg A, Kaushik M, Hussain MS, Yasmin S, Ansari MY. Gut Microbiota and Postbiotic Metabolites: Biotic Intervention for Enhancing Vaccine Responses and Personalized Medicine for Disease Prevention. Probiotics Antimicrob Proteins 2025. [DOI: 10.1007/s12602-025-10477-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 05/04/2025]
|
25
|
Cebrián V, Pini V, Thon A, Marina-García N, Salvador-Mátar A, Rodriguez C, Ahumada Ó. Introducing AVAC as an ultra-sensitive platform with broad dynamical range for high-throughput multiplexed biomarker detection using digital counting of plasmonic nanoparticles. Sci Rep 2025; 15:5390. [PMID: 39948115 PMCID: PMC11825739 DOI: 10.1038/s41598-025-88992-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Accurate detection and quantification of biomarkers at ultra-low levels is critical for disease diagnosis and effective treatment. Traditional detection technologies often lack the sensitivity, specificity, throughput, or multiplexing capacity required for comprehensive diagnostics, providing only a subset of these requirements. Here, we introduce AVAC, an automated optical technology for rapid and accurate biomarker detection with ultra-high sensitivity that significantly outperforms standard clinical assays. The core of this technology is the digital counting of plasmonic nanoparticles used as optical labels, enabling multiplexed, high-throughput detection of biomarkers. Validation studies demonstrate AVAC's high accuracy, with 98.2% specificity and detection limits as low as 26 fg/mL for HIV p24 protein and a quantification range of 160 fg/mL to 850 pg/mL for interleukin-6 (IL-6). The technology supports multiplexed assays without compromising sensitivity, as demonstrated by the simultaneous detection of three key biomarkers associated with cardiovascular disease. A counting range spanning more than four orders of magnitude ensures robust detection from ultra-low to high biomarker concentrations, and its ability to analyze up to 1,000 samples per hour provides high throughput suitable for large laboratories. With its unique combination of capabilities, this versatile platform has significant potential to advance biomarker-based diagnostics in clinical and research settings.
Collapse
Affiliation(s)
- Virginia Cebrián
- Mecwins S.A., Ronda de Poniente, 15, 2ºD, Tres Cantos, 28760, Madrid, Spain
| | - Valerio Pini
- Mecwins S.A., Ronda de Poniente, 15, 2ºD, Tres Cantos, 28760, Madrid, Spain
| | - Andreas Thon
- Mecwins S.A., Ronda de Poniente, 15, 2ºD, Tres Cantos, 28760, Madrid, Spain.
| | | | | | - Chloé Rodriguez
- Mecwins S.A., Ronda de Poniente, 15, 2ºD, Tres Cantos, 28760, Madrid, Spain
| | - Óscar Ahumada
- Mecwins S.A., Ronda de Poniente, 15, 2ºD, Tres Cantos, 28760, Madrid, Spain
| |
Collapse
|
26
|
Boottanun P, Fuseya S, Kuno A. Toward spatial glycomics and glycoproteomics: Innovations and applications. BBA ADVANCES 2025; 7:100146. [PMID: 40027887 PMCID: PMC11869499 DOI: 10.1016/j.bbadva.2025.100146] [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: 10/31/2024] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
In this mini review, we provide an overview of the challenging field of spatial glycomics/glycoproteomics. Owing to their complexity, sophisticated analytical methods and innovative technologies are needed to advance this field. An agile development approach enables unraveling aberrant glycosylations, glycobiomarkers, and glycotargets for spatial imaging, diagnosis, and therapeutic purposes. We discuss glycopathology and tissue glycomic profiling using highly sensitive lectin-based analyses and introduce deep visual proteomics for glycomic/glycoproteomic sample preparation. Additionally, we highlight the importance of leveraging laser microdissection and artificial intelligence-driven visual software for cell-type assignment and automation techniques, which are crucial for advancing glycomics/glycoproteomics.
Collapse
Affiliation(s)
- Patcharaporn Boottanun
- Molecular and Cellular Glycoproteomics Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8565, Japan
- Glycan and Life Systems Integration Center (GaLSIC), Soka University, Hachioji, Tokyo 192-8577, Japan
| | - Sayaka Fuseya
- Molecular and Cellular Glycoproteomics Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8565, Japan
| | - Atsushi Kuno
- Molecular and Cellular Glycoproteomics Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8565, Japan
| |
Collapse
|
27
|
Zhang S, Xu Z, Chen Y, Jiang L, Wang A, Shen G, Ding X. Lanthanide Metal-Organic Framework Flowers for Proteome Profiling and Biomarker Identification in Ultratrace Biofluid Samples. ACS NANO 2025; 19:4377-4390. [PMID: 39841883 DOI: 10.1021/acsnano.4c12280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Identifying effective biomarkers has long been a persistent need for early diagnosis and targeted therapy of disease. While mass spectrometry-based label-free proteomics with trace cell has been demonstrated, deep proteomics with ultratrace human biofluid remains challenging due to low protein concentration, extremely limited patient sample volume, and substantial protein contact losses during preprocessing. Herein, we proposed and validated lanthanide metal-organic framework flowers (MOF-flowers), as effective materials, to trap and enrich protein in biofluid jointly through cation-π interaction and O-Ln coordination. We further developed a MOF-flower assisted simplified and single-pot Sample Preparation (Mass-SP) workflow that incorporates protein capture, digest, and peptide elute into one single PCR tube to maximally avoid adsorptive sample loss. We adopted Mass-SP to decipher aqueous humor (AH) proteome signatures from cataract and retinal vein occlusion (RVO) patients and quantified ∼3900 proteins in merely 1 μL of AH. Combined with machine learning, we further identified PFKL as a prioritization biomarker for RVO disease with the areas under the curves of 0.95 ± 0.04. Mass-SP presents a strategy to identify de novo biomarkers and explore potential therapeutic targets with extremely limited clinical human body fluid resources.
Collapse
Affiliation(s)
- Shuang Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Zhixiao Xu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Youming Chen
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Aiting Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Guangxia Shen
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| |
Collapse
|
28
|
Shin D, Kim Y, Park J, Kim Y. High-throughput proteomics-guided biomarker discovery of hepatocellular carcinoma. Biomed J 2025; 48:100752. [PMID: 38901798 PMCID: PMC11743302 DOI: 10.1016/j.bj.2024.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Liver cancer stands as the fifth leading cause of cancer-related deaths globally. Hepatocellular carcinoma (HCC) comprises approximately 85%-90% of all primary liver malignancies. However, only 20-30% of HCC patients qualify for curative therapy, primarily due to the absence of reliable tools for early detection and prognosis of HCC. This underscores the critical need for molecular biomarkers for HCC management. Since proteins reflect disease status directly, proteomics has been utilized in biomarker developments for HCC. In particular, proteomics coupled with liquid chromatography-mass spectrometer (LC-MS) methods facilitate the process of discovering biomarker candidates for diagnosis, prognosis, and therapeutic strategies. In this work, we investigated LC-MS-based proteomics methods through recent reference reviews, with a particular focus on sample preparation and LC-MS methods appropriate for the discovery of HCC biomarkers and their clinical applications. We classified proteomics studies of HCC according to sample types, and we examined the coverage of protein biomarker candidates based on LC-MS methods in relation to study scales and goals. Comprehensively, we proposed protein biomarker candidates categorized by sample types and biomarker types for appropriate clinical use. In this review, we summarized recent LC-MS-based proteomics studies on HCC and proposed potential protein biomarkers. Our findings are expected to expand the understanding of HCC pathogenesis and enhance the efficiency of HCC diagnosis and prognosis, thereby contributing to improved patient outcomes.
Collapse
Affiliation(s)
- Dongyoon Shin
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, South Korea
| | - Yeongshin Kim
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, South Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, South Korea
| | - Junho Park
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, South Korea; Department of Pharmacology, School of Medicine, CHA University, Seongnam, South Korea.
| | - Youngsoo Kim
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, South Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, South Korea.
| |
Collapse
|
29
|
Teng P, Gao Z, Quan Q, He G, Song Q, Zhang X, Xiao W, Zhao J, Cao D, Liang J, Tang Y. SERS-based CRISPR/Cas12a assays for protein biomarker prostate-specific antigen detection. Anal Bioanal Chem 2025; 417:573-582. [PMID: 39576313 DOI: 10.1007/s00216-024-05663-5] [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: 08/23/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 01/06/2025]
Abstract
Sensitive and accurate detection of protein biomarkers is crucial for disease diagnosis, especially for early diagnosis. Here, we describe surface-enhanced Raman scattering (SERS)-based CRISPR/Cas12a assays (S-CRISPR) for protein biomarker detection. Firstly, an S-CRISPR-driven enzyme-linked immunosorbent assay (S-CasLISA) was developed utilizing a capture antibody coated on a microplate to recognize the target and a detection antibody labeled with active DNA to trigger the activity of CRISPR/Cas12a. With this assay, we achieved detection of prostate-specific antigen (PSA) as models at the picogram level. The limit of detection (LoD) of S-CasLISA was 0.17 pg mL-1 and in the range of 0.1 pg mL-1 to 10 ng mL-1. Further, we applied aptamer to S-CRISPR (S-Apt-CRISPR), combining the high sensitivity of SERS with the high selectivity of aptamers, while simplifying the operation process of CRISPR detection of protein biomarkers. The proposed S-Apt-CRISPR also could detect picogram-level PSA and without repeated washing steps. The LoD of S-Apt-CRISPR was 0.35 pg mL-1 and in the range of 0.1 pg mL-1 to 10 ng mL-1. Both SERS-based CRISPR/Cas12a assays were validated with clinical samples and demonstrated accuracy consistent with the chemiluminescence immunoassay. The introduction of the CRISPR/Cas12a system with SERS has the effect of improving the analytical capabilities of the system, thereby broadening and facilitating its application in the analysis of sensitive and accurate protein biomarkers.
Collapse
Affiliation(s)
- Peijun Teng
- GuangDong Engineering Technology Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zhixing Gao
- GuangDong Engineering Technology Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qiang Quan
- Research Center of Cancer Diagnosis and Therapy, Department of Oncology, the First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Guangbo He
- Guangdong Zhongxin Biotech Limited, Guangzhou, 510000, China
| | - Qifang Song
- GuangDong Engineering Technology Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xiaoli Zhang
- GuangDong Engineering Technology Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Wei Xiao
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
| | - Jianfu Zhao
- Research Center of Cancer Diagnosis and Therapy, Department of Oncology, the First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
| | - Donglin Cao
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Jiajie Liang
- Research Center of Cancer Diagnosis and Therapy, Department of Oncology, the First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
- GuangDong Engineering Technology Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
- Guangdong Zhongxin Biotech Limited, Guangzhou, 510000, China.
| | - Yong Tang
- GuangDong Engineering Technology Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
- Guangdong Zhongxin Biotech Limited, Guangzhou, 510000, China.
| |
Collapse
|
30
|
Waury K. Decision Tree for Protein Biomarker Selection for Clinical Applications. Methods Mol Biol 2025; 2884:355-368. [PMID: 39716013 DOI: 10.1007/978-1-0716-4298-6_21] [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: 12/25/2024]
Abstract
Discovery of novel protein biomarkers for clinical applications is an active research field across a manifold of diseases. Despite some successes and progress, the biomarker development pipeline still frequently ends in failure. Biomarker candidates that are discovered by appropriate technologies such as unbiased mass spectrometry cannot be validated or translated to immunoassays in many cases. Selection of strong disease biomarker candidates that further constitute suitable targets for antibody binding in immunoassays is thus important to allow routine clinical use. This essential selection step can be supported and rationalized using bioinformatics tools such as protein databases. Here, we present a workflow in the form of decision trees to computationally investigate biomarker candidates and their available affinity reagents in depth. This analysis can identify the most promising biomarker candidates for assay development, while minimal time and effort are required.
Collapse
Affiliation(s)
- Katharina Waury
- Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Lan Z, Chen R, Zou D, Zhao C. Microfluidic Nanoparticle Separation for Precision Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411278. [PMID: 39632600 PMCID: PMC11775552 DOI: 10.1002/advs.202411278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/11/2024] [Indexed: 12/07/2024]
Abstract
A deeper understanding of disease heterogeneity highlights the urgent need for precision medicine. Microfluidics, with its unique advantages, such as high adjustability, diverse material selection, low cost, high processing efficiency, and minimal sample requirements, presents an ideal platform for precision medicine applications. As nanoparticles, both of biological origin and for therapeutic purposes, become increasingly important in precision medicine, microfluidic nanoparticle separation proves particularly advantageous for handling valuable samples in personalized medicine. This technology not only enhances detection, diagnosis, monitoring, and treatment accuracy, but also reduces invasiveness in medical procedures. This review summarizes the fundamentals of microfluidic nanoparticle separation techniques for precision medicine, starting with an examination of nanoparticle properties essential for separation and the core principles that guide various microfluidic methods. It then explores passive, active, and hybrid separation techniques, detailing their principles, structures, and applications. Furthermore, the review highlights their contributions to advancements in liquid biopsy and nanomedicine. Finally, it addresses existing challenges and envisions future development spurred by emerging technologies such as advanced materials science, 3D printing, and artificial intelligence. These interdisciplinary collaborations are anticipated to propel the platformization of microfluidic separation techniques, significantly expanding their potential in precision medicine.
Collapse
Affiliation(s)
- Zhenwei Lan
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| | - Rui Chen
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| | - Da Zou
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| | - Chun‐Xia Zhao
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| |
Collapse
|
32
|
Riaz M, Rasool G, Yousaf R, Fatima H, Munir N, Ejaz H. Anti-Rheumatic potential of biological DMARDS and protagonistic role of bio-markers in early detection and management of rheumatoid arthritis. Innate Immun 2025; 31:17534259251324820. [PMID: 40091354 PMCID: PMC11912179 DOI: 10.1177/17534259251324820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that primarily affects the synovial joint linings, resulting in progressive disability, increased mortality, and considerable economic costs. Early treatment with disease-modifying antirheumatic medications (DMARDs) can significantly improve the overall outlook for people with RA. Contemporary pharmaceutical interventions, encompassing standard, biological, and emerging small molecule disease- modifying anti-rheumatic medications continue to be the cornerstone of RA management, with substantial advancements made in the pursuit of achieving remission from the disease and preventing joint deformities. Nevertheless, a substantial segment of individuals with RA do not experience a satisfactory response to existing treatments, underscoring the pressing need for novel therapeutic options. Biologic DMARDs are among the therapy choices. Non-tumor necrosis factor inhibitors (Non-TNFi) such as abatacept, rituximab, tocilizumab, and sarilumab are examples, as are anti-tumor necrosis factor (TNF) medications such as infliximab, adalimumab, etanercept, golimumab, and certolizumab pegol. More recent biomarkers have emerged and showed usefulness in the early detection of RA. These biomarkers, often referred to simply as "biomarkers", are quantifiable indicators of normal or pathologic processes, and they can also gauge treatment response. The assessment of RA treatment response typically combines patient-reported outcomes, physical evaluations, and laboratory findings, as there isn't a single biomarker that has proven sufficient for measuring disease activity. This review explores the usage of biologic DMARDs as a therapeutic approach for RA, as well as the biomarkers typically used for RA early diagnosis, prognosis prediction, and disease activity evaluation.
Collapse
Affiliation(s)
- Muhammad Riaz
- Department of Allied Health Sciences, University of Sargodha, Sargodha, Pakistan
| | - Ghulam Rasool
- Department of Allied Health Sciences, University of Sargodha, Sargodha, Pakistan
| | - Ruhamah Yousaf
- Department of Health Professional Technologies, The University of Lahore, Lahore, Pakistan
| | - Hina Fatima
- Department of Biochemistry, Government College Women University, Faisalabad, Pakistan
| | - Naveed Munir
- Department of Biomedical Lab Sciences, School of Health Sciences, University of Management and Technology, Lahore, Pakistan
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
33
|
Cao Y, Xia J, Li L, Zeng Y, Zhao J, Li G. Electrochemical Biosensors for Cancer Diagnosis: Multitarget Analysis to Present Molecular Characteristics of Tumor Heterogeneity. JACS AU 2024; 4:4655-4672. [PMID: 39735934 PMCID: PMC11672140 DOI: 10.1021/jacsau.4c00989] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/31/2024]
Abstract
Electrochemical biosensors are gaining attention as powerful tools in cancer diagnosis, particularly in liquid biopsy, due to their high efficiency, rapid response, exceptional sensitivity, and specificity. However, the complexity of intra- and intertumor heterogeneity, with variations in genetic and protein expression profiles and epigenetic modifications, makes electrochemical biosensors susceptible to false-positive or false-negative diagnostic outcomes. To address this challenge, there is growing interest in simultaneously analyzing multiple biomarkers to reveal molecular characteristics of tumor heterogeneity for precise cancer diagnosis. In this Perspective, we highlight recent advancements in utilizing electrochemical biosensors for cancer diagnosis, with a specific emphasis on the multitarget analysis of cancer biomarkers including tumor-associated nucleic acids, tumor protein markers, extracellular vesicles, and tumor cells. These biosensors hold significant promise for improving precision in early cancer diagnosis and monitoring, as well as potentially offering new insights into personalized cancer management.
Collapse
Affiliation(s)
- Ya Cao
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jianan Xia
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Lijuan Li
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yujing Zeng
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jing Zhao
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Genxi Li
- Center
for Molecular Recognition and Biosensing, Shanghai Engineering Research
Center of Organ Repair, Joint International Research Laboratory of
Biomaterials and Biotechnology in Organ Repair (Ministry of Education),
School of Life Sciences, Shanghai University, Shanghai 200444, China
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
34
|
Miyano T, Hirouchi M, Yoshimura N, Hattori K, Mikkaichi T, Kiyosawa N. Plasma microRNAs Associate Positive, Negative, and Cognitive Symptoms with Inflammation in Schizophrenia. Int J Mol Sci 2024; 25:13522. [PMID: 39769285 PMCID: PMC11676741 DOI: 10.3390/ijms252413522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Schizophrenia is a complex and heterogenous psychiatric disorder characterized by positive, negative, and cognitive symptoms. Our previous study identified three subgroups of schizophrenia patients based on plasma microRNA (miRNA) profiles. The present study aims to (1) verify the reproducibility of the miRNA-based patient stratification and (2) explore the pathophysiological pathways linked to the symptoms using plasma miRNAs. We measured levels of 376 miRNAs in plasma samples of schizophrenia patients and obtained their Positive and Negative Syndrome Scale (PANSS) scores and the Brief Assessment of Cognition in Schizophrenia (BACS) scores. The plasma miRNA profiles identified similar subgroups of patients as in the previous study, suggesting miRNA-based patient stratification is potentially reproducible. Our multivariate analysis identified optimal combinations of miRNAs to estimate the PANSS positive and negative subscales and BACS composite scores. Those miRNAs consistently enriched 'inflammation' and 'NFκB1' according to miRNA set enrichment analysis. Our literature-based text mining and survey confirmed that those miRNAs were associated with IL-1β, IL-6, and TNFα, suggesting that exacerbated positive, negative, and cognitive symptoms are associated with high inflammation. In conclusion, miRNAs are a potential biomarker to identify patient subgroups reflecting pathophysiological conditions and to investigate symptom-related molecular mechanisms in schizophrenia.
Collapse
Affiliation(s)
- Takuya Miyano
- Translational Science Department II, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa, Tokyo 140-8710, Japan; (M.H.); (T.M.); (N.K.)
| | - Masakazu Hirouchi
- Translational Science Department II, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa, Tokyo 140-8710, Japan; (M.H.); (T.M.); (N.K.)
| | - Naoki Yoshimura
- Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan;
| | - Kotaro Hattori
- Department of Bioresources, Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan;
| | - Tsuyoshi Mikkaichi
- Translational Science Department II, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa, Tokyo 140-8710, Japan; (M.H.); (T.M.); (N.K.)
| | - Naoki Kiyosawa
- Translational Science Department II, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa, Tokyo 140-8710, Japan; (M.H.); (T.M.); (N.K.)
| |
Collapse
|
35
|
Vo DK, Trinh KTL. Emerging Biomarkers in Metabolomics: Advancements in Precision Health and Disease Diagnosis. Int J Mol Sci 2024; 25:13190. [PMID: 39684900 DOI: 10.3390/ijms252313190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Metabolomics has come to the fore as an efficient tool in the search for biomarkers that are critical for precision health approaches and improved diagnostics. This review will outline recent advances in biomarker discovery based on metabolomics, focusing on metabolomics biomarkers reported in cancer, neurodegenerative disorders, cardiovascular diseases, and metabolic health. In cancer, metabolomics provides evidence for unique oncometabolites that are important for early disease detection and monitoring of treatment responses. Metabolite profiling for conditions such as neurodegenerative and mental health disorders can offer early diagnosis and mechanisms into the disease especially in Alzheimer's and Parkinson's diseases. In addition to these, lipid biomarkers and other metabolites relating to cardiovascular and metabolic disorders are promising for patient stratification and personalized treatment. The gut microbiome and environmental exposure also feature among the influential factors in biomarker discovery because they sculpt individual metabolic profiles, impacting overall health. Further, we discuss technological advances in metabolomics, current clinical applications, and the challenges faced by metabolomics biomarker validation toward precision medicine. Finally, this review discusses future opportunities regarding the integration of metabolomics into routine healthcare to enable preventive and personalized approaches.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Kieu The Loan Trinh
- BioNano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
36
|
Remes PM, Jacob CC, Heil LR, Shulman N, MacLean BX, MacCoss MJ. Hybrid Quadrupole Mass Filter-Radial Ejection Linear Ion Trap and Intelligent Data Acquisition Enable Highly Multiplex Targeted Proteomics. J Proteome Res 2024; 23:5476-5486. [PMID: 39475161 PMCID: PMC11956834 DOI: 10.1021/acs.jproteome.4c00599] [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] [Indexed: 11/06/2024]
Abstract
Targeted mass spectrometry (MS) methods are powerful tools for the selective and sensitive analysis of peptides identified in global discovery experiments. Selected reaction monitoring (SRM) is the most widely accepted clinical MS method due to its reliability and performance. However, SRM and parallel reaction monitoring (PRM) are limited in throughput and are typically used for assays with around 100 targets or fewer. Here we introduce a new MS platform featuring a quadrupole mass filter, collision cell, and linear ion trap architecture, capable of targeting 5000-8000 peptides per hour. This high multiplexing capability is facilitated by acquisition rates of 70-100 Hz and real-time chromatogram alignment. We present a Skyline external software tool for building targeted methods based on data-independent acquisition chromatogram libraries or unscheduled analysis of heavy labeled standards. Our platform demonstrates ∼10× lower limits of quantitation (LOQs) than traditional SRM on a triple quadrupole instrument for highly multiplexed assays, due to parallel product ion accumulation. Finally, we explore how analytical figures of merit vary with method duration and the number of analytes, providing insights into optimizing assay performance.
Collapse
Affiliation(s)
- Philip M Remes
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134, United States
| | - Cristina C Jacob
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134, United States
| | - Lilian R Heil
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134, United States
| | - Nicholas Shulman
- Department of Genome Sciences, University of Washington, 3720 15th St. NE, Seattle, Washington 98195, United States
| | - Brendan X MacLean
- Department of Genome Sciences, University of Washington, 3720 15th St. NE, Seattle, Washington 98195, United States
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, 3720 15th St. NE, Seattle, Washington 98195, United States
| |
Collapse
|
37
|
Talab MJ, Valizadeh A, Tahershamsi Z, Housaindokht MR, Ranjbar B. Personalized biocorona as disease biomarker: The challenges and opportunities. Biochim Biophys Acta Gen Subj 2024; 1868:130724. [PMID: 39426758 DOI: 10.1016/j.bbagen.2024.130724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/22/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
It is well known that when nanoparticles interact with biological fluids, a layer of proteins and biological components forms on them. This layer may alter the biological fate and efficiency of the nanomaterial. Recent studies have shown that illness states have a major impact on the structure of the biocorona, sometimes referred to as the "personalized protein corona." Physiological factors like illness, which impact the proteome and metabolome pattern and result in conformational changes in proteins, give rise to this structure of discrimination in biocorona decoration. Improving the efficiency of precise platforms for developing new molecular biomarkers for accurate illness diagnosis is vitally necessary. The biocorona pattern's discrimination may be a diagnostic tool for designing biosensors. As a result, in this review, we summarize the most current studies on the relationship between physiological conditions and the variety of biocorona patterns that influence the biological responses of nanosystems. The biocorona pattern's flexibility may provide new research directions and be utilized to create nanoparticle-based therapeutic and diagnostic products suited to certain physiological situations.
Collapse
Affiliation(s)
- Mahtab Jahanshah Talab
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Valizadeh
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Housaindokht
- Biophysical Chemistry Laboratory, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Bijan Ranjbar
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
38
|
Keum C, Yeom H, Noh TI, Yi SY, Jin S, Kim C, Shim JS, Yoon SG, Kim H, Lee KH, Kang SH, Jeong Y. Diagnosis of early-stage bladder cancer via unprocessed urine samples at the point of care. Nat Biomed Eng 2024:10.1038/s41551-024-01298-0. [PMID: 39609560 DOI: 10.1038/s41551-024-01298-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/30/2024] [Indexed: 11/30/2024]
Abstract
Diagnostic kits for the optical detection of bladder cancer in urine can facilitate effective screening and surveillance. However, the heterogeneity of urine samples, owing to patients with bladder cancer often presenting with haematuria, interfere with the transduction of the optical signal. Here we describe the development and point-of-care performance of a device for the detection of bladder cancer that obviates the need for sample processing. The device leverages the enzymatic release of organogel particles carrying solvatochromic fluorophores in the presence of urinary hyaluronidases-a bladder cancer biomarker. Owing to buoyancy, the particles transfer from the urine sample into the organic phase, where the change in fluorescence can be measured via a smartphone without interference from blood proteins. In a double-blind study with 80 unprocessed urine samples from patients with bladder cancer (including samples with haematuria) or other genitourinary diseases and with 25 samples from healthy participants, our system distinguished the cancerous samples, including those with early-stage bladder cancer, with accuracies of about 90%. Obviating the need for sample pretreatment may facilitate the at-home detection of bladder cancer.
Collapse
Affiliation(s)
- Changjoon Keum
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Haejin Yeom
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Republic of Korea
| | - Tae Il Noh
- Department of Urology, Korea University School of Medicine, Seoul, Republic of Korea
| | - Seung Yong Yi
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Republic of Korea
| | - Soyeong Jin
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Chemistry, Hanyang University, Seoul, Republic of Korea
| | - Chaekyu Kim
- Fusion Biotechnology, Inc., Ulsan, Republic of Korea
| | - Ji Sung Shim
- Department of Urology, Korea University School of Medicine, Seoul, Republic of Korea
| | - Sung Goo Yoon
- Department of Urology, Korea University School of Medicine, Seoul, Republic of Korea
| | - Hojun Kim
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Kwan Hyi Lee
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.
| | - Seok Ho Kang
- Department of Urology, Korea University School of Medicine, Seoul, Republic of Korea.
| | - Youngdo Jeong
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Kędzierska M, Bańkosz M. Role of Proteins in Oncology: Advances in Cancer Diagnosis, Prognosis, and Targeted Therapy-A Narrative Review. J Clin Med 2024; 13:7131. [PMID: 39685591 DOI: 10.3390/jcm13237131] [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: 10/27/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Modern oncology increasingly relies on the role of proteins as key components in cancer diagnosis, prognosis, and targeted therapy. This review examines advancements in protein biomarkers across several cancer types, including breast cancer, lung cancer, ovarian cancer, and hepatocellular carcinoma. These biomarkers have proven critical for early detection, treatment response monitoring, and tailoring personalized therapeutic strategies. The article highlights the utility of targeted therapies, such as tyrosine kinase inhibitors and monoclonal antibodies, in improving treatment efficacy while minimizing systemic toxicity. Despite these advancements, challenges like tumor resistance, variability in protein expression, and diagnostic heterogeneity persist, complicating universal application. The review underscores future directions, including the integration of artificial intelligence, advanced protein analysis technologies, and the development of combination therapies to overcome these barriers and refine personalized cancer treatment.
Collapse
Affiliation(s)
- Magdalena Kędzierska
- Department of Chemotherapy, Medical University of Lodz, Copernicus Memorial Hospital of Lodz, 90-549 Lodz, Poland
| | - Magdalena Bańkosz
- CUT Doctoral School, Faculty of Materials Engineering and Physics, Department of Material Engineering, Cracow University of Technology, 37 Jana Pawla II Av., 31-864 Krakow, Poland
| |
Collapse
|
40
|
Luo Q, Zeng Q, Wang C, Zhang C, Yu H, Yang Y, Guan X. Ultrasensitive Single-Molecule Biosensor by Periodic Modulation of Magnetic Particle Motion. NANO LETTERS 2024; 24:13998-14003. [PMID: 39441689 DOI: 10.1021/acs.nanolett.4c03443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Ultrasensitive detection of low-abundance biomarkers by modern single-molecule technologies is critical for better diagnosis of severe diseases, but inevitable nonspecific bindings often cause fluctuations in the single-molecule counting results. Here we present an approach to improve the specificity in a single-molecule immunoassay by translating molecular binding signals into periodic nanomotion of magnetic particles. The sandwiched immunoassay is modified by using a long linker to tether one antibody onto a gold-covered substrate and a magnetic particle with another antibody coated as the reporter. By actively oscillating the particles with alternating magnetic fields, we could reliably identify specific binding through intensity fluctuation in plasmonic images of single particles. As a proof of concept, we demonstrate the detection of IFN-γ at the femtomolar level by the digital counting of specifically bound molecules. This active strategy outperforms existing passive motion-based approaches in sensitivity and speed, paving the way for disease diagnosis with low-abundance biomarkers.
Collapse
Affiliation(s)
- Qingqing Luo
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qiang Zeng
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chen Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Cheng Zhang
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hui Yu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yuting Yang
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xinping Guan
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
41
|
Di Luca A, Bennato F, Ianni A, Martino C, Henry M, Meleady P, Martino G. Label-free liquid chromatography mass spectrometry analysis of changes in broiler liver proteins under transport stress. PLoS One 2024; 19:e0311539. [PMID: 39466737 PMCID: PMC11515959 DOI: 10.1371/journal.pone.0311539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 09/19/2024] [Indexed: 10/30/2024] Open
Abstract
Transportation duration and distance are significant concerns for animal welfare, particularly in the poultry industry. However, limited proteomic studies have investigated the impact of transport duration on poultry welfare. In this study, mass spectrometry based bottom up proteomics was employed to sensitively and impartially profile the liver tissue proteome of chickens, addressing the issue of animal stress and welfare in response to transportation before slaughter. The liver exudates obtained from Ross 508 chickens exposed to either short or long road transportation underwent quantitative label-free LC-MS proteomic profiling. This method identified a total of 1,368 proteins, among which 35 were found to be significantly different (p < 0.05) and capable of distinguishing between short and long road transportation conditions. Specifically, 23 proteins exhibited up-regulation in the non stressed group, while 12 proteins showed up-regulation in the stressed group. The proteins identified in this pilot study encompassed those linked to homeostasis and cellular energetic balance, including heat shock proteins and the 5'-nucleotidase domain-containing family. These results contribute to a deeper understanding of the proteome in broiler liver tissues, shedding light on poultry adaptability to transport stress. Furthermore, the identified proteins present potential as biomarkers, suggesting promising approaches to enhance poultry care and management within the industry.
Collapse
Affiliation(s)
- Alessio Di Luca
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Francesca Bennato
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Andrea Ianni
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Camillo Martino
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Giuseppe Martino
- Department of Bioscience and Technology for Food Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| |
Collapse
|
42
|
Apatzidou DA, Violesti A, Konstantinidis A, Bao K, Silbereisen A, Bostanci N. Protein profile at newly restored implants compared to contralateral teeth over 12-months: a pilot study. Clin Oral Investig 2024; 28:590. [PMID: 39390228 DOI: 10.1007/s00784-024-05984-w] [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: 07/01/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
OBJECTIVES To determine crevicular fluid alterations in protein expression of newly restored implants during their first year of function and associate them with those of contralateral teeth. MATERIALS AND METHODS In ten non-smokers, successfully treated for periodontitis, one newly restored implant (baseline-T0) and one corresponding tooth were followed for 12-months (T1). Oral hygiene was monitored during the study. Periodontal clinical indices and crevicular fluid were collected from an implant-site (PICF) and a tooth-site (GCF). Total proteomic profiles of PICF and GCF were investigated using label-free quantitative proteomics. RESULTS Clinical recordings remained stable at 12-months on the tooth-/implant-site basis. The comparative analysis of protein enrichment between teeth and implants at T0 revealed 664 human proteins, with 93 found only in teeth and 217 exclusively in implants. Among the 354 overlapping proteins, 46 were upregulated (log2FC > 1) in teeth, while 61 in implants. At T1, 569 human proteins were exclusively identified, with 67 found only in teeth and 193 exclusively in implants. Of the 309 overlapping proteins, 22 were upregulated in teeth, while 48 were in implants. The over-representation enrichment analysis identified "interferon-alpha response" and "allograft rejection" pathways, as significantly regulated categories at T0, with the latter being over-represented at T1. CONCLUSIONS Peri-implant tissue maturation was evident during the study. Proteins expressed in crevicular fluid reflected unique patterns between implants and teeth that are worth studying. CLINICAL RELEVANCE Different proteomic patterns were observed at the implant-site compared to the contralateral tooth-site towards inflammatory processes that prevail within otherwise clinically healthy peri-implant tissues. CLINICAL TRIAL REGISTRATION NUMBER ClinicalTrials.gov ID: NCT06379022.
Collapse
Affiliation(s)
- Danae A Apatzidou
- Department of Preventive Dentistry, Periodontology and Implant Biology, Faculty of Dentistry, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Anastasia Violesti
- Department of Preventive Dentistry, Periodontology and Implant Biology, Faculty of Dentistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Antonis Konstantinidis
- Department of Preventive Dentistry, Periodontology and Implant Biology, Faculty of Dentistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kai Bao
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Angelika Silbereisen
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nagihan Bostanci
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
43
|
Mattoo TK, Spencer JD. Biomarkers for urinary tract infection: present and future perspectives. Pediatr Nephrol 2024; 39:2833-2844. [PMID: 38483594 DOI: 10.1007/s00467-024-06321-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 08/28/2024]
Abstract
A prompt diagnosis of urinary tract infection (UTI) is necessary to minimize its symptoms and limit sequelae. The current UTI screening by urine test strip analysis and microscopic examination has suboptimal diagnostic accuracy. A definitive diagnosis of UTI by urine culture takes two to three days for the results. These limitations necessitate a need for better biomarkers for the diagnosis and subsequent management of UTI in children. Here, we review the value of currently available UTI biomarkers and highlight the potential of emerging biomarkers that can facilitate a more rapid and accurate UTI diagnosis. Of the newer UTI biomarkers, the most promising are blood procalcitonin (PCT) and urinary neutrophil gelatinase-associated lipocalin (NGAL). PCT can provide diagnostic benefits and should be considered in patients who have a blood test for other reasons. NGAL, which is on the threshold of clinical care, needs more research to address its scope and utilization, including point-of-care application. Employment of these and other biomarkers may ultimately improve UTI diagnosis, guide UTI therapy, reduce antibiotic use, and mitigate UTI complications.
Collapse
Affiliation(s)
- Tej K Mattoo
- Pediatric Nephrologist, Wayne Pediatrics, Detroit, MI, USA.
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA.
- Department of Urology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - John David Spencer
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
44
|
Chandran V, Liao W, de Vlam K. Biomarkers in Psoriasis and Psoriatic Arthritis: Where Are We Now? J Rheumatol 2024; 51:74-76. [PMID: 39009392 DOI: 10.3899/jrheum.2024-0260] [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] [Accepted: 03/10/2024] [Indexed: 07/17/2024]
Abstract
At the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) 2023 annual conference and trainee symposium, the status of psoriatic disease (PsD) biomarkers was discussed in a workshop. The significant heterogeneity of PsD causes disease management to be very challenging, but biomarkers can prove helpful in disease diagnosis, stratification, and precision medicine. Although a few potential biomarkers have been discovered, none have been fully validated. Recent studies have used omic technologies that show promise but need further verification and validation. Many challenges remain, but the anticipated results of studies being conducted by recently established large consortia may lead to the identification of clinically actionable biomarkers.
Collapse
Affiliation(s)
- Vinod Chandran
- V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, Departments of Medicine and Laboratory Medicine and Pathobiology, University of Toronto, and Krembil Research Institute, University Health Network, Toronto, Ontario, Canada;
| | - Wilson Liao
- W. Liao, MD, Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Kurt de Vlam
- K. de Vlam, MD, PhD, Department of Rheumatology, University Hospitals Leuven, and Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
45
|
Dewey HM, Lamb A, Budhathoki-Uprety J. Recent advances on applications of single-walled carbon nanotubes as cutting-edge optical nanosensors for biosensing technologies. NANOSCALE 2024; 16:16344-16375. [PMID: 39157856 DOI: 10.1039/d4nr01892c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Single-walled carbon nanotubes (SWCNTs) possess outstanding photophysical properties which has garnered interest towards utilizing these materials for biosensing and imaging applications. The near-infrared (NIR) fluorescence within the tissue transparent region along with their photostability and sizes in the nanoscale make SWCNTs valued candidates for the development of optical sensors. In this review, we discuss recent advances in the development and the applications of SWCNT-based nano-biosensors. An overview of SWCNT's structural and photophysical properties, sensor development, and sensing mechanisms are described. Examples of SWCNT-based optical nanosensors for detection of disease biomarkers, pathogens (bacteria and viruses), plant stressors, and environmental contaminants including heavy metals and disinfectants are provided. Molecular detection in biofluids, in vitro, and in vivo (small animal models and plants) are highlighted, and sensor integration into portable substrates for implantable and wearable sensing devices has been discussed. Recent advancements, which include high throughput assays and the use of machine learning models to predict more sensitive and robust sensing outcomes are discussed. Current limitations and future perspectives on translation of SWCNT optical probes into clinical practices have been provided.
Collapse
Affiliation(s)
- Hannah M Dewey
- Department of Textile Engineering, Chemistry and Science, Wilson College of Textiles, North Carolina State University, Raleigh, NC, 27695, USA.
| | - Ashley Lamb
- Department of Textile Engineering, Chemistry and Science, Wilson College of Textiles, North Carolina State University, Raleigh, NC, 27695, USA.
| | - Januka Budhathoki-Uprety
- Department of Textile Engineering, Chemistry and Science, Wilson College of Textiles, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
46
|
Layek S, Sengupta N. Response of Foldable Protein Conformations to Non-Physiological Perturbations: Interplay of Thermal Factors and Confinement. Chemphyschem 2024:e202400618. [PMID: 39104119 DOI: 10.1002/cphc.202400618] [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: 06/06/2024] [Revised: 07/14/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024]
Abstract
Technological advances frequently interface biomolecules with nanomaterials at non-physiological conditions, necessitating response characterization of key processes. Similar encounters are expected in cellular contexts. We report in silico investigations of the response of diverse protein conformational states to lowering of temperature and imposition of spatial constraints. Conformational states are represented by folded form of the Albumin binding domain (ABD) protein, its compact denatured form, and structurally disordered nascent folding elements. Data from extensive simulations are evaluated to elicit structural, thermodynamic and dynamic responses of the states and their associated environment. Analyses reveal alterations to folding propensity with reduced thermal energy and confinement, with signatures of trend reversal in highly disordered states. Across temperatures, confinement has restrictive effects on volume and energetic fluctuations, leading to narrowing of differences in isothermal compressibility (κ) and heat capacities (Cp). While excess (over ideal gas) entropy of the hydration layer marks dependence on the conformational state at bulk, confinement triggers erasure of differences. These observations are largely consistent with timescales of protein-water hydrogen bonding dynamics. The results implicate multi-factorial associations within a simple bio-nano complex. We expect the current study to motivate investigations of more biologically relevant interfaces towards mechanistic understanding and potential applications.
Collapse
Affiliation(s)
- Sarbajit Layek
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Kolkata Mohanpur, West Bengal, 741246, India
| | - Neelanjana Sengupta
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Kolkata Mohanpur, West Bengal, 741246, India
| |
Collapse
|
47
|
George H, Sun Y, Wu J, Yan Y, Wang R, Pesavento RP, Mathew MT. Intelligent salivary biosensors for periodontitis: in vitro simulation of oral oxidative stress conditions. Med Biol Eng Comput 2024; 62:2409-2434. [PMID: 38609577 DOI: 10.1007/s11517-024-03077-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 03/16/2024] [Indexed: 04/14/2024]
Abstract
ASTRACT One of the most common oral diseases affecting millions of people worldwide is periodontitis. Usually, proteins in body fluids are used as biomarkers of diseases. This study focused on hydrogen peroxide, lipopolysaccharide (LPS), and lactic acid as salivary non-protein biomarkers for oxidative stress conditions of periodontitis. Electrochemical analysis of artificial saliva was done using Gamry with increasing hydrogen peroxide, bLPS, and lactic acid concentrations. Electrochemical impedance spectroscopy (EIS) and cyclic voltammetry (CV) were conducted. From EIS data, change in capacitance and CV plot area were calculated for each test condition. Hydrogen peroxide groups had a decrease in CV area and an increase in percentage change in capacitance, lipopolysaccharide groups had a decrease in CV area and a decrease in percentage change in capacitance, and lactic acid groups had an increase of CV area and an increase in percentage change in capacitance with increasing concentrations. These data showed a unique combination of electrochemical properties for the three biomarkers. Scanning electron microscopy (SEM) with energy dispersive spectroscopy (EDS) employed to observe the change in the electrode surface and elemental composition data present on the sensor surface also showed a unique trend of elemental weight percentages. Machine learning models using hydrogen peroxide, LPS, and lactic acid electrochemical data were applied for the prediction of risk levels of periodontitis. The detection of hydrogen peroxide, LPS, and lactic acid by electrochemical biosensors indicates the potential to use these molecules as electrochemical biomarkers and use the data for ML-driven prediction tool for the periodontitis risk levels.
Collapse
Affiliation(s)
- Haritha George
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Yani Sun
- Department of Material Science, University of Illinois at Chicago, Chicago, IL, USA
| | - Junyi Wu
- Department of Computer Science, Illinois Institute of Technology, Chicago, IL, USA
| | - Yan Yan
- Department of Computer Science, Illinois Institute of Technology, Chicago, IL, USA
| | - Rong Wang
- Department of Biological and Chemical Sciences, Illinois Institute of Technology, Chicago, IL, USA
| | - Russell P Pesavento
- Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Mathew T Mathew
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Material Science, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
48
|
Savva KV, MacKenzie A, Coombes RC, Zhifang NM, Hanna BG, Peters CJ. An original study assessing biomarker success rate in breast cancer recurrence biomarker research. BMC Med 2024; 22:307. [PMID: 39075505 PMCID: PMC11288100 DOI: 10.1186/s12916-024-03460-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/30/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Breast cancer is the second most common cause of cancer mortality worldwide. Biomarker discovery has led to advances in understanding molecular phenotyping and thus has a great potential for precision management of this diverse disease. Despite increased interest in the biomarker field, only a small number of breast cancer biomarkers are known to be clinically useful. Therefore, it is very important to characterise the success rate of biomarkers in this field and study potential reasons for the deficit. We therefore aim to achieve quantitative characterisation of the biomarker translation gap by tracking the progress of prognostic biomarkers associated with breast cancer recurrence. METHODS An electronic systematic search was conducted in Medline and Embase databases using keywords and mesh headings associated with breast cancer recurrence biomarkers (1940-2023). Abstracts were screened, and primary clinical studies involving breast cancer recurrence biomarkers were selected. Upon identification of relevant literature, we extracted the biomarker name, date of publication and journal name. All analyses were performed using IBM SPSS Statistics and GraphPad prism (La Jolla, California, USA). RESULTS A total of 19,195 articles were identified, from which 4597 articles reported breast cancer biomarkers associated with recurrence. Upon data extraction, 2437 individual biomarkers were identified. Out of these, 23 are currently recommended for clinical use, which corresponds to only 0.94% of all discovered biomarkers. CONCLUSIONS This study characterised for the first time the translational gap in the field of recurrence-related breast cancer biomarkers, indicating that only 0.94% of identified biomarkers were recommended for clinical use. This denotes an evident barrier in the biomarker research field and emphasises the need for a clearer route from biomarker discovery through to implementation.
Collapse
Affiliation(s)
- K-V Savva
- Department of Surgery and Cancer, Imperial College London, 10th Floor QEQM Wing, St Mary's Hospital, London, W2 1NY, UK.
| | - A MacKenzie
- Division of Surgery, Department of Surgery and Cancer, Imperial College London, London, UK
| | - R C Coombes
- Department of Surgery and Cancer, Imperial College London, 10th Floor QEQM Wing, St Mary's Hospital, London, W2 1NY, UK
| | - N M Zhifang
- Division of Surgery, Department of Surgery and Cancer, Imperial College London, London, UK
| | - B G Hanna
- Department of Surgery and Cancer, Imperial College London, 10th Floor QEQM Wing, St Mary's Hospital, London, W2 1NY, UK
| | - C J Peters
- Department of Surgery and Cancer, Imperial College London, 10th Floor QEQM Wing, St Mary's Hospital, London, W2 1NY, UK
| |
Collapse
|
49
|
Pazoki A, Dadfar S, Shadab A, Haghmorad D, Oksenych V. Soluble CD40 Ligand as a Promising Biomarker in Cancer Diagnosis. Cells 2024; 13:1267. [PMID: 39120299 PMCID: PMC11311304 DOI: 10.3390/cells13151267] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer remains a significant challenge in medicine due to its complexity and heterogeneity. Biomarkers have emerged as vital tools for cancer research and clinical practice, facilitating early detection, prognosis assessment, and treatment monitoring. Among these, CD40 ligand (CD40L) has gained attention for its role in immune response modulation. Soluble CD40 ligand (sCD40L) has shown promise as a potential biomarker in cancer diagnosis and progression, reflecting interactions between immune cells and the tumor microenvironment. This review explores the intricate relationship between sCD40L and cancer, highlighting its diagnostic and prognostic potential. It discusses biomarker discovery, emphasizing the need for reliable markers in oncology, and elucidates the roles of CD40L in inflammatory responses and interactions with tumor cells. Additionally, it examines sCD40L as a biomarker, detailing its significance across various cancer types and clinical applications. Moreover, the review focuses on therapeutic interventions targeting CD40L in malignancies, providing insights into cellular and gene therapy approaches and recombinant protein-based strategies. The clinical effectiveness of CD40L-targeted therapy is evaluated, underscoring the need for further research to unlock the full potential of this signaling pathway in cancer management.
Collapse
Affiliation(s)
- Alireza Pazoki
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Sepehr Dadfar
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Alireza Shadab
- Department of Health Science, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| |
Collapse
|
50
|
Harkness BM, Chen S, Kim K, Reddy AP, McFarland TJ, Hegarty DM, Everist SJ, Saugstad JA, Lapidus J, Galor A, Aicher SA. Tear Proteins Altered in Patients with Persistent Eye Pain after Refractive Surgery: Biomarker Candidate Discovery. J Proteome Res 2024; 23:2629-2640. [PMID: 38885176 DOI: 10.1021/acs.jproteome.4c00339] [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] [Indexed: 06/20/2024]
Abstract
Some patients develop persistent eye pain after refractive surgery, but factors that cause or sustain pain are unknown. We tested whether tear proteins of patients with pain 3 months after surgery differ from those of patients without pain. Patients undergoing refractive surgery (laser in situ keratomileusis or photorefractive keratectomy ) were recruited from 2 clinics, and tears were collected 3 months after surgery. Participants rated their eye pain using a numerical rating scale (NRS, 0-10; no pain-worst pain) at baseline, 1 day, and 3 months after surgery. Using tandem mass tag proteomic analysis, we examined tears from patients with pain [NRS ≥ 3 at 3 months (n = 16)] and patients with no pain [NRS ≤ 1 at 3 months (n = 32)] after surgery. A subset of proteins (83 of 2748 detected, 3.0%) were associated with pain 3 months after surgery. High-dimensional statistical models showed that the magnitude of differential expression was not the only important factor in classifying tear samples from pain patients. Models utilizing 3 or 4 proteins had better classification performance than single proteins and represented differences in both directions (higher or lower in pain). Thus, patterns of protein differences may serve as biomarkers of postsurgical eye pain as well as potential therapeutic targets.
Collapse
Affiliation(s)
- Brooke M Harkness
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| | - Siting Chen
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, Oregon 97239-4197, United States
- Biostatistics & Design Program, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| | - Kilsun Kim
- Proteomics Shared Resource, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| | - Ashok P Reddy
- Proteomics Shared Resource, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| | - Trevor J McFarland
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| | - Deborah M Hegarty
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| | - Steven J Everist
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| | - Julie A Saugstad
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| | - Jodi Lapidus
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, Oregon 97239-4197, United States
- Biostatistics & Design Program, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| | - Anat Galor
- Bascom Palmer Eye Institute, University of Miami Health System, Miami, Florida 33146, United States
- Miami Veterans Affairs Hospital, Miami, Florida 33125-1624, United States
| | - Sue A Aicher
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon 97239-4197, United States
| |
Collapse
|