1
|
He W, Li Y, Fan J, Liu Y, Yuan M, Cheng S, Huang X, Yan B, Zhang Z, Xiu Y, Zhu H, Lan T, Chang Z, Jiang Y, Li H, Meng X, Wang Y, Van Kaer L, Verkhratsky A, Wang Y, Shi FD, Jin WN. Gain-of-function PPM1D mutations attenuate ischemic stroke. Cell Death Differ 2025:10.1038/s41418-025-01523-6. [PMID: 40399534 DOI: 10.1038/s41418-025-01523-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 04/10/2025] [Accepted: 04/30/2025] [Indexed: 05/23/2025] Open
Abstract
Identification of genetic aberrations in stroke, the second leading cause of death worldwide, is of paramount importance for understanding the disease pathogenesis and generating new therapies. Whole-genome sequencing from 10,241 ischemic stroke patients identified eight patients carrying gain-of-function mutations on coding variants in the protein phosphatase magnesium-dependent 1 δ (PPM1D) gene. Patients carrying PPM1D mutations exhibit better stroke-related clinical phenotypes, including improvements in peripheral inflammation, fibrinogen, low-density lipoprotein, cholesterol and plateletcrit level. Experimental brain ischemia in Ppm1d-deficient (Ppm1d-/-) mice resulted in enlarged lesions and pronounced neurological impairments. Spatial transcriptomics revealed a distinct Ppm1d-associated gene expression pattern, indicating disrupted endothelial homeostasis during ischemic brain injury. Proteomic analysis demonstrated that differentially expressed proteins in primary brain endothelial cells from Ppm1d-/- mice were significantly enriched in the peroxisome proliferator-activated receptors (PPARs)-mediated metabolic signaling. Mechanistically, Ppm1d deficiency promoted aberrant fatty acid β-oxidation and increased oxidative stress, which impaired endothelial cell function through the PPARα pathway. A small molecule, T2755, was identified to engage Trp427 and stabilize PPM1D, thereby mitigating ischemic brain injury in mice. Collectively, we find that PPM1D protects against ischemic brain injury and validates its pharmacological stabilizer T2755 as a promising therapy for ischemic stroke. Gain-of-function PPM1D mutations attenuate ischemic cerebral injury. Whole-genome sequencing data of 10,241 ischemic stroke patients from the Third Chinese National Stroke Registry (CNSR-III) identified eight patients with gain-of-function mutations in the protein phosphatase magnesium-dependent 1 δ (PPM1D) gene (17q23.2). These mutation carriers displayed improved peripheral inflammation, decreased fibrinogen, low-density lipoprotein, cholesterol and plateletcrit level. Ppm1d-deficient (Ppm1d-/-) mice exhibited exacerbated stroke outcomes, characterized by enlarged infarct volumes, disrupted cerebrovascular architecture, and enhanced neuro-inflammation. Mechanistically, Ppm1d deficiency induced the disturbance of endothelial fatty acid metabolism involving the PPARα pathway. Through integrated computational modeling, virtual screening, and in vitro validation, T2755 was identified as a small molecule PPM1D stabilizer. Pharmacological PPM1D stabilization with T2755 significantly attenuated ischemic brain injury in murine models.
Collapse
Affiliation(s)
- Wenyan He
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Yan Li
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Junwan Fan
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Yang Liu
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Meng Yuan
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Si Cheng
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Xinying Huang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Bo Yan
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhuoran Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuwen Xiu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huimin Zhu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tian Lan
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Zhilin Chang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Yong Jiang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Hao Li
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Xia Meng
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Yilong Wang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Yongjun Wang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China.
| | - Fu-Dong Shi
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China.
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China.
| | - Wei-Na Jin
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of innovative Drug and Device Research & Development for Cerebrovascular Diseases, Beijing, China.
| |
Collapse
|
2
|
Angelis J, Schröder EA, Xiao Z, Gabriel W, Wilhelm M. Peptide Property Prediction for Mass Spectrometry Using AI: An Introduction to State of the Art Models. Proteomics 2025; 25:e202400398. [PMID: 40211610 PMCID: PMC12076536 DOI: 10.1002/pmic.202400398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 05/15/2025]
Abstract
This review explores state of the art machine learning and deep learning models for peptide property prediction in mass spectrometry-based proteomics, including, but not limited to, models for predicting digestibility, retention time, charge state distribution, collisional cross section, fragmentation ion intensities, and detectability. The combination of these models enables not only the in silico generation of spectral libraries but also finds many additional use cases in the design of targeted assays or data-driven rescoring. This review serves as both an introduction for newcomers and an update for experienced researchers aiming to develop accessible and reproducible models for peptide property predictions. Key limitations of the current models, including difficulties in handling diverse post-translational modifications and instrument variability, highlight the need for large-scale, harmonized datasets, and standardized evaluation metrics for benchmarking.
Collapse
Affiliation(s)
- Jesse Angelis
- Computational Mass SpectrometryTechnical University of MunichFreisingGermany
| | - Eva Ayla Schröder
- Computational Mass SpectrometryTechnical University of MunichFreisingGermany
| | - Zixuan Xiao
- Computational Mass SpectrometryTechnical University of MunichFreisingGermany
| | - Wassim Gabriel
- Computational Mass SpectrometryTechnical University of MunichFreisingGermany
| | - Mathias Wilhelm
- Computational Mass SpectrometryTechnical University of MunichFreisingGermany
- Munich Data Science Institute (MDSI)Technical University of MunichGarchingGermany
| |
Collapse
|
3
|
Tian Q, Cao H, Chu L, Gao H, Gao Q. Proteomic and Metabolomic Analysis of the Neuroprotective Effects of Lycium Ruthenicum Polyphenols in Diabetic Peripheral Neuropathy Mice. Food Sci Nutr 2025; 13:e70209. [PMID: 40321612 PMCID: PMC12045932 DOI: 10.1002/fsn3.70209] [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: 08/15/2024] [Revised: 03/15/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Lycium ruthenicum polyphenols (LRP) have been proven to be anti-inflammatory, antioxidant, and neuroprotective phytochemicals. This study applies proteomics and metabolomics to LRP-treated db/db mice to explore its potential effects mechanism. The experiments were divided into three groups: normal control db/m group, diabetic peripheral neuropathy (DPN) db/db group, and LRP-treated db/db group. We examined physiological and biochemical indicators, behavioral indicators, and histopathology. As for the mechanism, we used TMT-based quantification proteomics and LC-MS/MS-based metabolomics for sciatic nerve and serum. After 8 weeks of treatment, the fasting blood glucose level, mechanical withdrawal threshold, and thermal hyperalgesia were significantly improved. Pathological examination showed a significant alleviation in sciatic nerve histomorphology in the LRP group. Proteomics and metabolomics showed that the interventional effects of LRP were enriched mainly in oxidative phosphorylation, cardiac muscle contraction, and serum metabolites were enriched mainly in amino acid metabolism. LRP improves neurological function by improving mitochondrial functions, promoting neuronal development, and ameliorating dysregulation of amino acid metabolism. These results provide theoretical evidence for LRP as a potential functional food ingredient for the prevention and treatment of DPN.
Collapse
Affiliation(s)
- Qi Tian
- School of Public HealthNingxia Medical UniversityYinchuanNingxiaChina
- Key Laboratory of Environmental Factors and Chronic Disease ControlNingxia Medical UniversityYinchuanNingxiaChina
| | - Hongdou Cao
- School of Public HealthNingxia Medical UniversityYinchuanNingxiaChina
- Key Laboratory of Environmental Factors and Chronic Disease ControlNingxia Medical UniversityYinchuanNingxiaChina
| | - Liwen Chu
- School of Public HealthNingxia Medical UniversityYinchuanNingxiaChina
- Key Laboratory of Environmental Factors and Chronic Disease ControlNingxia Medical UniversityYinchuanNingxiaChina
| | - Hua Gao
- Department of PharmacyGeneral Hospital of Ningxia Medical UniversityYinchuanNingxiaChina
| | - Qinghan Gao
- School of Public HealthNingxia Medical UniversityYinchuanNingxiaChina
- Key Laboratory of Environmental Factors and Chronic Disease ControlNingxia Medical UniversityYinchuanNingxiaChina
| |
Collapse
|
4
|
Heo Y, Kim WJ, Cho YJ, Jung JW, Kim NS, Choi IY. Advances in cancer genomics and precision oncology. Genes Genomics 2025; 47:399-416. [PMID: 39849190 DOI: 10.1007/s13258-024-01614-7] [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: 11/07/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Next-generation sequencing has revolutionized genome science over the last two decades. Indeed, the wealth of sequence information on our genome has deepened our understanding on cancer. Cancer is a genetic disease caused by genetic or epigenetic alternations that affect the expression of genes that control cell functions, particularly cell growth and division. Utilization of next-generation sequencing in cancer gene panels has enabled the identification of actionable gene alterations in cancer patients to guide personalized precision medicine. OBJECTIVE The aim is to provide information that can identify actionable gene alterations, enabling personalized precision medicine for cancer patients. RESULTS & DISCUSSION Equipped with next-generation sequencing techniques, international collaboration programs on cancer genomics have identified numerous mutations, gene fusions, microsatellite variations, copy number variations, and epigenetics changes that promote the transformation of normal cells into tumors. Cancer classification has traditionally been based on cell type or tissue-of-origin and the morphological characteristics of the cancer. However, interactive genomic analyses have currently reclassified cancers based on systemic molecular-based taxonomy. Although all cancer-causing genes and mechanisms have yet to be completely understood or identified, personalized or precision medicine is now currently possible for some forms of cancer. Unlike the "one-size-fits-all" approach of traditional medicine, precision medicine allows for customized or personalized treatment based on genomic information. CONCLUSION Despite the availability of numerous cancer gene panels, technological innovation in genomics and expansion of knowledge on the cancer genome will allow precision oncology to manage even more types of cancers.
Collapse
Affiliation(s)
- Yonjong Heo
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Gangwon, Republic of Korea
| | - Woo-Jin Kim
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Gangwon, Republic of Korea
| | - Yong-Joon Cho
- Department of Molecular Bioscience, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jae-Won Jung
- Genetic Sciences Group, Thermo Fisher Scientific Solutions Korea Co., Ltd., Seoul, 06349, Republic of Korea
| | - Nam-Soo Kim
- Department of Molecular Bioscience, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- NBIT Co., Ltd., Chuncheon, 24341, Republic of Korea.
| | - Ik-Young Choi
- Department of Smart Farm and Agricultural Industry, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
5
|
Minayo Martín S, Villar M, Sánchez-Cano A, Fontoura-Gonçalves C, Hernández JM, Williams RAJ, Quevedo MÁ, Höfle U. Impact of urbanization on the house sparrow (Passer domesticus): Serum proteome and pathogen prevalence. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 968:178920. [PMID: 39987830 DOI: 10.1016/j.scitotenv.2025.178920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
The house sparrow (Passer domesticus) is a globally distributed species found in rural, urban and other humanised environments. In Europe, sparrow populations have significantly declined in recent decades, especially in urbanised areas. In the present study, we analysed the impact of urbanization on sparrow body condition, pathogen prevalence, and serum proteome changes. Sparrows were captured in four locations with two different urbanization status (rural/urban). Biometric data, blood samples and oral and cloacal swabs were collected. Rural sparrows exhibited significantly better body condition compared to urban sparrows, with no notable differences between sexes. Haemoparasite prevalence was higher in rural sparrows 70.16 % (87/124) than in urban sparrows 50 % (27/54). No avian influenza virus (AIV) or West Nile virus (WNV) genetic material was found, although one urban sparrow (0.58 %) had antibodies to AIV. Serum proteomics revealed that rural sparrows showed an up-regulation of proteins involved in the metabolism, in contrast to proteins of the immune system and the coagulation system, which were found to be over-represented in urban sparrows. Thus, we documented a worse body condition and immune system activation in urban sparrows in contrast to a more active metabolism and a higher prevalence of avian malaria in rural sparrows, and at least occasional exposure to AIV in urban habitats. This information suggests exposure to urban environments may alter the host-pathogen relationship. Urbanization in combination with exposure to AIV, could modulate their role in viral spread and transmission.
Collapse
Affiliation(s)
- Sara Minayo Martín
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain.
| | - Margarita Villar
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain; Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain
| | - Alberto Sánchez-Cano
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Catarina Fontoura-Gonçalves
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain; CIBIO- Centro de Investigação em Biodiversidade e Recursos Genéticos. Rua Padre Armando Quintas, N° 7, Vairão 4485-661, Portugal; BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, 4485-661 Vairão, Portugal
| | - José Manuel Hernández
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Richard A J Williams
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University of Madrid (UCM), Calle Antonio Nováis 12, Ciudad Universitaria, 28040 Madrid, Spain
| | - Miguel Ángel Quevedo
- Centro de Conservación de la Biodiversidad Zoobotánico Jerez. Calle Madreselva, s/n, 11408, Jerez de la Frontera, Spain
| | - Ursula Höfle
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain.
| |
Collapse
|
6
|
Rasul HO, Ghafour DD, Aziz BK, Hassan BA, Rashid TA, Kivrak A. Decoding Drug Discovery: Exploring A-to-Z In Silico Methods for Beginners. Appl Biochem Biotechnol 2025; 197:1453-1503. [PMID: 39630336 DOI: 10.1007/s12010-024-05110-2] [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: 11/19/2024] [Indexed: 03/29/2025]
Abstract
The drug development process is a critical challenge in the pharmaceutical industry due to its time-consuming nature and the need to discover new drug potentials to address various ailments. The initial step in drug development, drug target identification, often consumes considerable time. While valid, traditional methods such as in vivo and in vitro approaches are limited in their ability to analyze vast amounts of data efficiently, leading to wasteful outcomes. To expedite and streamline drug development, an increasing reliance on computer-aided drug design (CADD) approaches has merged. These sophisticated in silico methods offer a promising avenue for efficiently identifying viable drug candidates, thus providing pharmaceutical firms with significant opportunities to uncover new prospective drug targets. The main goal of this work is to review in silico methods used in the drug development process with a focus on identifying therapeutic targets linked to specific diseases at the genetic or protein level. This article thoroughly discusses A-to-Z in silico techniques, which are essential for identifying the targets of bioactive compounds and their potential therapeutic effects. This review intends to improve drug discovery processes by illuminating the state of these cutting-edge approaches, thereby maximizing the effectiveness and duration of clinical trials for novel drug target investigation.
Collapse
Affiliation(s)
- Hezha O Rasul
- Department of Pharmaceutical Chemistry, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq.
| | - Dlzar D Ghafour
- Department of Medical Laboratory Science, College of Science, Komar University of Science and Technology, 46001, Sulaimani, Iraq
- Department of Chemistry, College of Science, University of Sulaimani, 46001, Sulaimani, Iraq
| | - Bakhtyar K Aziz
- Department of Nanoscience and Applied Chemistry, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq
| | - Bryar A Hassan
- Computer Science and Engineering Department, School of Science and Engineering, University of Kurdistan Hewler, KRI, Iraq
- Department of Computer Science, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq
| | - Tarik A Rashid
- Computer Science and Engineering Department, School of Science and Engineering, University of Kurdistan Hewler, KRI, Iraq
| | - Arif Kivrak
- Department of Chemistry, Faculty of Sciences and Arts, Eskisehir Osmangazi University, Eskişehir, 26040, Turkey
| |
Collapse
|
7
|
Liu Y, Yang Z, Pu JJ, Zhong J, Khoo U, Su Y, Zhang G. Proteogenomic characterisation of primary oral cancer unveils extracellular matrix remodelling and immunosuppressive microenvironment linked to lymph node metastasis. Clin Transl Med 2025; 15:e70261. [PMID: 40038875 PMCID: PMC11879901 DOI: 10.1002/ctm2.70261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is an increasingly prevalent malignancy worldwide. This study aims to understand molecular alterations associated with lymph node metastasis of OSCC in order to improve treatment strategies. We analysed a cohort of 46 patients with primary OSCC, including 10 with lymph node metastasis and 36 without. Using a comprehensive multi-omics approach - encompassing genomic, transcriptomic, proteomic, epigenetic, single-cell, and spatial analyses - we integrated data to delineate the molecular landscape of OSCC in the context of lymph node metastasis. Our genomic analysis identified significant mutations in key genes within the MAPK, TGF-β and WNT signalling pathways, which are essential for tumour development. The proteogenomic analysis highlighted pathways critical for lymph node dissemination and factors contributing to an immunosuppressive tumour microenvironment. Elevated levels of POSTN were found to reorganise the extracellular matrix (ECM), interact with TGF-β, disrupt cell cycle regulation and suppress the immune response by reducing VCAM1 activity. Integrated analyses of single-cell and spatial transcriptome data revealed that cancer-associated fibroblasts (CAFs) secrete TGF-β1/2, promoting cancer cell metastasis through epithelial-mesenchymal transition (EMT). Our integrated multi-omics analysis provides a detailed understanding of molecular mechanisms driving lymph node metastasis of OSCC. These insights could lead to more precise diagnostics and targeted treatments. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yu Liu
- Department of Thoracic Surgery/Institute of Thoracic OncologyWest China HospitalSichuan UniversityChengduChina
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| | - Zhenyu Yang
- Department of Thoracic Surgery/Institute of Thoracic OncologyWest China HospitalSichuan UniversityChengduChina
| | - Jingya Jane Pu
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| | - Jie Zhong
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| | - Ui‐Soon Khoo
- Department of PathologySchool of Clinical MedicineThe University of Hong KongHong KongHong Kong
| | - Yu‐Xiong Su
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| | - Gao Zhang
- Faculty of DentistryThe University of Hong KongHong KongHong Kong
| |
Collapse
|
8
|
Liu Y, Kong X, Zhang X, Chen Z, Wang J, Chen H, Jiang L. SERPINA3, FGA, AGP1, ITIH3 and SAA1 as novel biomarkers for eosinophilic granulomatosis with polyangiitis diagnosis and activity assessment. Rheumatology (Oxford) 2025; 64:1316-1325. [PMID: 38552326 DOI: 10.1093/rheumatology/keae187] [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: 12/07/2023] [Accepted: 03/16/2024] [Indexed: 03/06/2025] Open
Abstract
OBJECTIVE The objective of this study was to identify novel biomarkers for diagnosis and prediction of active eosinophilic granulomatosis with polyangiitis (EGPA) through data-independent acquisition (DIA) analysis. METHODS Plasma samples from 11 EGPA patients and 10 healthy controls (HCs) were analysed through DIA to identify potential biomarkers. The results were validated in 32 EGPA patients, 24 disease controls (DCs), and 20 HCs using ELISA. The receiver operating characteristic (ROC) curve was used to assess the diagnostic value of candidate biomarkers. RESULTS Thirty-five differentially expressed proteins (DEPs) (24 upregulated and 11 downregulated) were screened between the EGPA and HC groups. Five proteins, including serine proteinase inhibitor A3 (SERPINA3), alpha-fibrinogen (FGA), alpha-1 acid glycoprotein 1(AGP1), inter-alpha-trypsin inhibitor heavy chain H3 (ITIH3), and serum amyloid A1 (SAA1), were significantly upregulated in EGPA compared with HCs. Apart from SAA1, all proteins were also higher in EGPA patients compared with DCs. Furthermore, a panel of SERPINA3 and SAA1 exhibited potential diagnostic value for EGPA, with an area under the curve (AUC) of 0.953, while a panel of SERPINA3, FGA, AGP1 and ITIH3 showed good discriminative power for differentiating EGPA from DCs, with an AUC of 0.926. Moreover, SERPINA3, FGA and AGP levels were significantly higher in active EGPA and correlated well with disease activity. A combination of SERPINA3 and AGP1 exhibited an excellent AUC of 0.918 for disease activity assessment. CONCLUSION SERPINA3, FGA, AGP1, ITIH3 and SAA1 were identified as potential biomarkers for EGPA diagnosis and disease activity assessment. Among them, as a single biomarker, SERPINA3 had the best diagnostic performance.
Collapse
Affiliation(s)
- Yun Liu
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiufang Kong
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao Zhang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihong Chen
- Department of Pulmonary and Critical Care Medicine, Shanghai Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinghua Wang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huiyong Chen
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lindi Jiang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Center of Clinical Epidemiology and Evidence-based Medicine, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Yuan X, Zhan Z, Lin W, Zhang C, Wang B. The membrane may be a key factor influencing browning: a mini review on browning mechanisms of fresh-cut fruit and vegetables from a multi-omics perspective. Front Nutr 2025; 12:1534594. [PMID: 40070475 PMCID: PMC11893370 DOI: 10.3389/fnut.2025.1534594] [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: 11/26/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Fresh-cut fruit and vegetables are susceptible to browning during storage and subsequent consumption. The cell membrane acts as a vital structural barrier, compartmentalizing various substances within living organisms. The fresh-cutting process induces mechanical injuries, disrupting these membranes and resulting in the leakage of cellular contents. This facilitates direct contact between substances and enzymes that mediate browning reactions. This mini review explores the potential roles of cell membranes in the browning of fresh-cut fruit and vegetables from a multi-omics perspective, aiming to provide novel insights into the underlying mechanisms of browning in fresh-cut fruit and vegetables. Considering potential roles of cell membranes in blocking the browning of fresh-cut fruit and vegetables, future studies should focus on elucidating the precise mechanisms by which membranes regulate browning reactions, aiming to provide directions for the development of more effective intervention strategies.
Collapse
Affiliation(s)
- Xiao Yuan
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region/College of Biology and Agriculture, Shaoguan University, Shaoguan, China
- Guangdong Provincial Engineering and Technology Research Center of Special Fruit and Vegetables in Northern Region, Shaoguan University, Shaoguan, China
| | - Zhaoxia Zhan
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region/College of Biology and Agriculture, Shaoguan University, Shaoguan, China
| | - Wei Lin
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region/College of Biology and Agriculture, Shaoguan University, Shaoguan, China
| | - Can Zhang
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region/College of Biology and Agriculture, Shaoguan University, Shaoguan, China
| | - Bin Wang
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region/College of Biology and Agriculture, Shaoguan University, Shaoguan, China
- Guangdong Provincial Engineering and Technology Research Center of Special Fruit and Vegetables in Northern Region, Shaoguan University, Shaoguan, China
| |
Collapse
|
10
|
Hao Y, Yang Y, Zhao H, Chen Y, Zuo T, Zhang Y, Yu H, Cui L, Song X. Multi-omics in Allergic Rhinitis: Mechanism Dissection and Precision Medicine. Clin Rev Allergy Immunol 2025; 68:19. [PMID: 39964644 PMCID: PMC11836232 DOI: 10.1007/s12016-025-09028-3] [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: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Allergic rhinitis (AR) is a common chronic inflammatory airway disease caused by inhaled allergens, and its prevalence has increased in recent decades. AR not only causes nasal leakage, itchy nose, nasal congestion, sneezing, and allergic conjunctivitis but also induces asthma, as well as sleep disorders, anxiety, depression, memory loss, and other phenomena that seriously affect the patient's ability to study and work, lower their quality of life, and burden society. The current methods used to diagnose and treat AR are still far from ideal. Multi-omics technology can be used to comprehensively and systematically analyze the differentially expressed DNA, RNA, proteins, and metabolites and their biological functions in patients with AR. These capabilities allow for an in-depth understanding of the intrinsic pathogenic mechanism of AR, the ability to explore key cells and molecules that drive its progression, and to design personalized treatment for AR. This article summarizes the progress made in studying AR by use of genomics, epigenomics, transcriptomics, proteomics, metabolomics, and microbiomics in order to illustrate the important role of multi-omics technologies in facilitating the precise diagnosis and treatment of AR.
Collapse
Affiliation(s)
- Yan Hao
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Yujuan Yang
- Qingdao Medical College, Qingdao University, Qingdao, 266000, Shandong, China
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Hongfei Zhao
- Qingdao Medical College, Qingdao University, Qingdao, 266000, Shandong, China
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Ying Chen
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- The 2Nd Medical College of Binzhou Medical University, Yantai, 264000, Shandong, China
| | - Ting Zuo
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- The 2Nd Medical College of Binzhou Medical University, Yantai, 264000, Shandong, China
| | - Yu Zhang
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Hang Yu
- Qingdao Medical College, Qingdao University, Qingdao, 266000, Shandong, China
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Limei Cui
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China.
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China.
| | - Xicheng Song
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China.
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China.
| |
Collapse
|
11
|
Kumar N, Du Z, Li Y. pLM4CPPs: Protein Language Model-Based Predictor for Cell Penetrating Peptides. J Chem Inf Model 2025; 65:1128-1139. [PMID: 39878455 DOI: 10.1021/acs.jcim.4c01338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Cell-penetrating peptides (CPPs) are short peptides capable of penetrating cell membranes, making them valuable for drug delivery and intracellular targeting. Accurate prediction of CPPs can streamline experimental validation in the lab. This study aims to assess pretrained protein language models (pLMs) for their effectiveness in representing CPPs and develop a reliable model for CPP classification. We evaluated peptide embeddings generated from BEPLER, CPCProt, SeqVec, various ESM variants (ESM, ESM-2 with expanded feature set, ESM-1b, and ESM-1v), ProtT5-XL UniRef50, ProtT5-XL BFD, and ProtBERT. We developed pLM4CCPs, a novel deep learning architecture using convolutional neural networks (CNNs) as the classifier for binary classification of CPPs. pLM4CCPs demonstrated superior performance over existing state-of-the-art CPP prediction models, achieving improvements in accuracy (ACC) by 4.9-5.5%, Matthews correlation coefficient (MCC) by 9.3-10.2%, and sensitivity (Sn) by 14.1-19.6%. Among all the tested models, ESM-1280 and ProtT5-XL BFD demonstrated the highest overall performance on the kelm data set. ESM-1280 achieved an ACC of 0.896, an MCC of 0.796, a Sn of 0.844, and a specificity (Sp) of 0.978. ProtT5-XL BFD exhibited superior performance with an ACC of 0.901, an MCC of 0.802, an Sn of 0.885, and an Sp of 0.917. pLM4CCPs combine predictions from multiple models to provide a consensus on whether a given peptide sequence is classified as a CPP or non-CPP. This approach will enhance prediction reliability by leveraging the strengths of each individual model. A user-friendly web server for bioactivity predictions, along with data sets, is available at https://ry2acnp6ep.us-east-1.awsapprunner.com. The source code and protocol for adapting pLM4CPPs can be accessed on GitHub at https://github.com/drkumarnandan/pLM4CPPs. This platform aims to advance CPP prediction and peptide functionality modeling, aiding researchers in exploring peptide functionality effectively.
Collapse
Affiliation(s)
- Nandan Kumar
- Department of Grain Science and Industry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Zhenjiao Du
- Department of Grain Science and Industry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Yonghui Li
- Department of Grain Science and Industry, Kansas State University, Manhattan, Kansas 66506, United States
| |
Collapse
|
12
|
Hashimoto-Roth E, Forget D, Gaspar VP, Bennett SAL, Gauthier MS, Coulombe B, Lavallée-Adam M. MAGPIE: A Machine Learning Approach to Decipher Protein-Protein Interactions in Human Plasma. J Proteome Res 2025; 24:383-396. [PMID: 39772751 DOI: 10.1021/acs.jproteome.4c00160] [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: 01/11/2025]
Abstract
Immunoprecipitation coupled to tandem mass spectrometry (IP-MS/MS) methods are often used to identify protein-protein interactions (PPIs). While these approaches are prone to false positive identifications through contamination and antibody nonspecific binding, their results can be filtered using negative controls and computational modeling. However, such filtering does not effectively detect false-positive interactions when IP-MS/MS is performed on human plasma samples. Therein, proteins cannot be overexpressed or inhibited, and existing modeling algorithms are not adapted for execution without such controls. Hence, we introduce MAGPIE, a novel machine learning-based approach for identifying PPIs in human plasma using IP-MS/MS, which leverages negative controls that include antibodies targeting proteins not expected to be present in human plasma. A set of negative controls used for false positive interaction modeling is first constructed. MAGPIE then assesses the reliability of PPIs detected in IP-MS/MS experiments using antibodies that target known plasma proteins. When applied to five IP-MS/MS experiments as a proof of concept, our algorithm identified 68 PPIs with an FDR of 20.77%. MAGPIE significantly outperformed a state-of-the-art PPI discovery tool and identified known and predicted PPIs. Our approach provides an unprecedented ability to detect human plasma PPIs, which enables a better understanding of biological processes in plasma.
Collapse
Affiliation(s)
- Emily Hashimoto-Roth
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Diane Forget
- Translational Proteomics Laboratory, Institut de recherches cliniques de Montréal, 110, avenue des Pins West, Montréal, Québec H2W 1R7, Canada
| | - Vanessa P Gaspar
- Translational Proteomics Laboratory, Institut de recherches cliniques de Montréal, 110, avenue des Pins West, Montréal, Québec H2W 1R7, Canada
| | - Steffany A L Bennett
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Department of Chemistry and Biomolecular Sciences, Centre for Catalysis and Research Innovation, University of Ottawa, 150 Louis-Pasteur Pvt, Ottawa, Ontario K1N 6N5, Canada
| | - Marie-Soleil Gauthier
- Translational Proteomics Laboratory, Institut de recherches cliniques de Montréal, 110, avenue des Pins West, Montréal, Québec H2W 1R7, Canada
| | - Benoit Coulombe
- Translational Proteomics Laboratory, Institut de recherches cliniques de Montréal, 110, avenue des Pins West, Montréal, Québec H2W 1R7, Canada
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, Pavillon Roger-Gaudry C.P. 6128, Succursale Centre-ville Montréal, Québec H3C 3J7, Canada
| | - Mathieu Lavallée-Adam
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
13
|
Peris-Díaz MD, Krężel A, Barran P. Deciphering the safeguarding role of cysteine residues in p53 against H 2O 2-induced oxidation using high-resolution native mass spectrometry. Commun Chem 2025; 8:13. [PMID: 39814824 PMCID: PMC11736120 DOI: 10.1038/s42004-024-01395-w] [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: 09/26/2024] [Accepted: 12/12/2024] [Indexed: 01/18/2025] Open
Abstract
The transcription factor p53 is exquisitely sensitive and selective to a broad variety of cellular environments. Several studies have reported that oxidative stress weakens the p53-DNA binding affinity for certain promoters depending on the oxidation mechanism. Despite this body of work, the precise mechanisms by which the physiologically relevant DNA-p53 tetramer complex senses cellular stresses caused by H2O2 are still unknown. Here, we employed native mass spectrometry (MS) and ion mobility (IM)-MS coupled to chemical labelling and H2O2-induced oxidation to examine the mechanism of redox regulation of the p53-p21 complex. Our approach has found that two reactive cysteines in p53 protect against H2O2-induced oxidation by forming reversible sulfenates.
Collapse
Affiliation(s)
- Manuel David Peris-Díaz
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, Manchester, UK.
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie 14a, Wrocław, Poland.
| | - Artur Krężel
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie 14a, Wrocław, Poland
| | - Perdita Barran
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, Manchester, UK.
| |
Collapse
|
14
|
Li Y, Li W, Zheng Y, Wang T, Pu R, Zhang Z. Desalting strategies for native mass spectrometry. Talanta 2025; 281:126824. [PMID: 39250868 DOI: 10.1016/j.talanta.2024.126824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
In native mass spectrometry (MS) salts are indispensable for preserving the native structures of biomolecules, but detrimental to mass sensitivity, resolution, and accuracy. Such a conflict makes desalting in native MS more challenging, distinctive, and sample-dependent than in peptide-centric MS. This review first briefly introduces the charged residue mechanism whereby native-like gaseous protein ions are released from electrospray droplets, revealing a higher degree of salt adduction than denatured proteins. Subsequently, this review summarizes and explores the existing strategies, underlying mechanisms and future perspectives of desalting in native MS. These strategies mainly focus on buffer exchange into volatile salts (offline and online approaches), addition of solution additives (e.g., anion, supercharging reagent, solution phase chelator and amino acid), use of submicron electrospray emitters (down to 60 nm), and other potential approaches (e.g., induced and electrophoretic nanoelectrospray ionization). The strategies of online buffer exchange and using nanoscale electrospray emitters are highlighted. This review would not only be a valuable addition to the field of sample preparation in MS, but would also serve as a beginner's guide to desalting in native MS.
Collapse
Affiliation(s)
- Yun Li
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Weijie Li
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Yajun Zheng
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China.
| | - Tong Wang
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Ruijin Pu
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Zhiping Zhang
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China.
| |
Collapse
|
15
|
Padilla-Garfias F, Araiza-Villanueva M, Calahorra M, Sánchez NS, Peña A. Advances in the Degradation of Polycyclic Aromatic Hydrocarbons by Yeasts: A Review. Microorganisms 2024; 12:2484. [PMID: 39770687 PMCID: PMC11728250 DOI: 10.3390/microorganisms12122484] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/20/2024] [Accepted: 11/30/2024] [Indexed: 01/16/2025] Open
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are toxic organic compounds produced during the incomplete combustion of organic materials and are commonly found in the environment due to anthropogenic activities such as industrial and vehicular emissions as well as natural sources, mainly volcanic eruptions and forest fires. PAHs are well known for their bioaccumulative capacity and environmental persistence, raising concerns due to their adverse effects on human health, including their carcinogenic potential. In recent years, bioremediation has emerged as a promising, effective, and sustainable solution for the degradation of PAHs in contaminated environments. In this context, yeasts have proven to be key microorganisms in the degradation of these compounds, owing to their ability to metabolize them through a series of enzymatic pathways. This review explores the advancements in yeast-mediated degradation of PAHs, with a particular focus on the role of enzymes such as cytochrome P450 (CYPs), epoxide hydrolases (EHs), and glutathione S-transferases (GSTs), which facilitate the breakdown of these compounds. The review also discusses the applications of genetic engineering to enhance the efficiency of yeasts in PAH degradation and the use of omics technologies to predict the catabolic potential of these organisms. Additionally, it examines studies addressing the degradation of benzo[a]pyrene (BaP) by yeasts such as Debaryomyces hansenii, and the potential future implications of omics sciences for developing new bioremediation.
Collapse
Affiliation(s)
- Francisco Padilla-Garfias
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Mexico City 04510, Mexico; (M.A.-V.); (M.C.); (N.S.S.)
| | | | | | | | - Antonio Peña
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Mexico City 04510, Mexico; (M.A.-V.); (M.C.); (N.S.S.)
| |
Collapse
|
16
|
Zhang X, Zhou X, Tu Z, Qiang L, Lu Z, Xie Y, Liu CH, Zhang L, Fu Y. Proteomic and ubiquitinome analysis reveal that microgravity affects glucose metabolism of mouse hearts by remodeling non-degradative ubiquitination. PLoS One 2024; 19:e0313519. [PMID: 39541295 PMCID: PMC11563481 DOI: 10.1371/journal.pone.0313519] [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: 07/13/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Long-term exposure to a microgravity environment leads to structural and functional changes in hearts of astronauts. Although several studies have reported mechanisms of cardiac damage under microgravity conditions, comprehensive research on changes at the protein level in these hearts is still lacking. In this study, proteomic analysis of microgravity-exposed hearts identified 156 differentially expressed proteins, and ubiquitinomic analysis of these hearts identified 169 proteins with differential ubiquitination modifications. Integrated ubiquitinomic and proteomic analysis revealed that differential proteomic changes caused by transcription affect the immune response in microgravity-exposed hearts. Additionally, changes in ubiquitination modifications under microgravity conditions excessively activated certain kinases, such as hexokinase and phosphofructokinase, leading to cardiac metabolic disorders. These findings provide new insights into the mechanisms of cardiac damage under microgravity conditions.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xuemei Zhou
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Zhiwei Tu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Lihua Qiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Zhe Lu
- Institute of Microbiology (Chinese Academy of Sciences), CAS Key Laboratory of Pathogenic Microbiology and Immunology, Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuping Xie
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Cui Hua Liu
- Institute of Microbiology (Chinese Academy of Sciences), CAS Key Laboratory of Pathogenic Microbiology and Immunology, Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yesheng Fu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
17
|
Tachino J, Togami Y, Matsumoto H, Matsubara T, Seno S, Ogura H, Oda J. Plasma proteomics profile-based comparison of torso versus brain injury: A prospective cohort study. J Trauma Acute Care Surg 2024; 97:557-565. [PMID: 38595266 PMCID: PMC11446512 DOI: 10.1097/ta.0000000000004356] [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: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Trauma-related deaths and posttraumatic sequelae are a global health concern, necessitating a deeper understanding of the pathophysiology to advance trauma therapy. Proteomics offers insights into identifying and analyzing plasma proteins associated with trauma and inflammatory conditions; however, current proteomic methods have limitations in accurately measuring low-abundance plasma proteins. This study compared plasma proteomics profiles of patients from different acute trauma subgroups to identify new therapeutic targets and devise better strategies for personalized medicine. METHODS This prospective observational single-center cohort study was conducted between August 2020 and September 2021 in the intensive care unit of Osaka University Hospital in Japan. Enrolling 59 consecutive patients with blunt trauma, we meticulously analyzed plasma proteomics profiles in participants with torso or head trauma, comparing them with those of controls (mild trauma). Using the Olink Explore 3072 instrument (Olink Proteomics AB, Uppsala, Sweden), we identified five endotypes (α-ε) via unsupervised hierarchical clustering. RESULTS The median time from injury to blood collection was 47 minutes [interquartile range, 36-64 minutes]. The torso trauma subgroup exhibited 26 unique proteins with significantly altered expression, while the head trauma subgroup showed 68 unique proteins with no overlap between the two. The identified endotypes included α (torso trauma, n = 8), β (young patients with brain injury, n = 5), γ (severe brain injury postsurgery, n = 8), δ (torso or brain trauma with mild hyperfibrinolysis, n = 18), and ε (minor trauma, n = 20). Patients with torso trauma showed changes in blood pressure, smooth muscle adaptation, hypermetabolism, and hypoxemia. Patients with traumatic brain injury had dysregulated blood coagulation and altered nerves regeneration and differentiation. CONCLUSION This study identified unique plasma protein expression patterns in patients with torso trauma and traumatic brain injury, helping categorize five distinct endotypes. Our findings may offer new insights for clinicians, highlighting potential strategies for personalized medicine and improved trauma-related care. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level III.
Collapse
|
18
|
Mondelo-Macía P, García-González J, León-Mateos L, Abalo A, Bravo S, Chantada Vazquez MDP, Muinelo-Romay L, López-López R, Díaz-Peña R, Dávila-Ibáñez AB. Identification of a Proteomic Signature for Predicting Immunotherapy Response in Patients With Metastatic Non-Small Cell Lung Cancer. Mol Cell Proteomics 2024; 23:100834. [PMID: 39216661 PMCID: PMC11474190 DOI: 10.1016/j.mcpro.2024.100834] [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: 05/26/2023] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Immunotherapy has improved survival rates in patients with cancer, but identifying those who will respond to treatment remains a challenge. Advances in proteomic technologies have enabled the identification and quantification of nearly all expressed proteins in a single experiment. Integrating mass spectrometry with high-throughput technologies has facilitated comprehensive analysis of the plasma proteome in cancer, facilitating early diagnosis and personalized treatment. In this context, our study aimed to investigate the predictive and prognostic value of plasma proteome analysis using the SWATH-MS (Sequential Window Acquisition of All Theoretical Mass Spectra) strategy in newly diagnosed patients with non-small cell lung cancer (NSCLC) receiving pembrolizumab therapy. We enrolled 64 newly diagnosed patients with advanced NSCLC treated with pembrolizumab. Blood samples were collected from all patients before and during therapy. A total of 171 blood samples were analyzed using the SWATH-MS strategy. Plasma protein expression in metastatic NSCLC patients prior to receiving pembrolizumab was analyzed. A first cohort (discovery cohort) was employed to identify a proteomic signature predicting immunotherapy response. Thus, 324 differentially expressed proteins between responder and non-responder patients were identified. In addition, we developed a predictive model and found a combination of seven proteins, including ATG9A, DCDC2, HPS5, FIL1L, LZTL1, PGTA, and SPTN2, with stronger predictive value than PD-L1 expression alone. Additionally, survival analyses showed an association between the levels of ATG9A, DCDC2, SPTN2 and HPS5 with progression-free survival (PFS) and/or overall survival (OS). Our findings highlight the potential of proteomic technologies to detect predictive biomarkers in blood samples from NSCLC patients, emphasizing the correlation between immunotherapy response and the idenfied protein set.
Collapse
Affiliation(s)
- Patricia Mondelo-Macía
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Jorge García-González
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain; Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Luis León-Mateos
- Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain; Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Alicia Abalo
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain
| | - Susana Bravo
- Proteomic Unit, Instituto de Investigaciones Sanitarias-IDIS, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - María Del Pilar Chantada Vazquez
- Proteomic Unit, Instituto de Investigaciones Sanitarias-IDIS, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - Rafael López-López
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain; Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain; Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain; Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Roberto Díaz-Peña
- Fundación Pública Galega de Medicina Xenómica, SERGAS; Grupo de Medicina Xenomica-USC, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; Faculty of Health Sciences, Universidad Autónoma de Chile, Talca, Chile
| | - Ana B Dávila-Ibáñez
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain; CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain; Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
| |
Collapse
|
19
|
Tan P, Wei X, Huang H, Wang F, Wang Z, Xie J, Wang L, Liu D, Hu Z. Application of omics technologies in studies on antitumor effects of Traditional Chinese Medicine. Chin Med 2024; 19:123. [PMID: 39252074 PMCID: PMC11385818 DOI: 10.1186/s13020-024-00995-x] [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: 06/28/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Traditional Chinese medicine (TCM) is considered to be one of the most comprehensive and influential form of traditional medicine. It plays an important role in clinical treatment and adjuvant therapy for cancer. However, the complex composition of TCM presents challenges to the comprehensive and systematic understanding of its antitumor mechanisms, which hinders further development of TCM with antitumor effects. Omics technologies can immensely help in elucidating the mechanism of action of drugs. They utilize high-throughput sequencing and detection techniques to provide deeper insights into biological systems, revealing the intricate mechanisms through which TCM combats tumors. Multi-omics approaches can be used to elucidate the interrelationships among different omics layers by integrating data from various omics disciplines. By analyzing a large amount of data, these approaches further unravel the complex network of mechanisms underlying the antitumor effects of TCM and explain the mutual regulations across different molecular levels. In this study, we presented a comprehensive overview of the recent progress in single-omics and multi-omics research focused on elucidating the mechanisms underlying the antitumor effects of TCM. We discussed the significance of omics technologies in advancing research on the antitumor properties of TCM and also provided novel research perspectives and methodologies for further advancing this research field.
Collapse
Affiliation(s)
- Peng Tan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xuejiao Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Huiming Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fei Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhuguo Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jinxin Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Longyan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dongxiao Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
20
|
Vitorino R. Transforming Clinical Research: The Power of High-Throughput Omics Integration. Proteomes 2024; 12:25. [PMID: 39311198 PMCID: PMC11417901 DOI: 10.3390/proteomes12030025] [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: 06/17/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024] Open
Abstract
High-throughput omics technologies have dramatically changed biological research, providing unprecedented insights into the complexity of living systems. This review presents a comprehensive examination of the current landscape of high-throughput omics pipelines, covering key technologies, data integration techniques and their diverse applications. It looks at advances in next-generation sequencing, mass spectrometry and microarray platforms and highlights their contribution to data volume and precision. In addition, this review looks at the critical role of bioinformatics tools and statistical methods in managing the large datasets generated by these technologies. By integrating multi-omics data, researchers can gain a holistic understanding of biological systems, leading to the identification of new biomarkers and therapeutic targets, particularly in complex diseases such as cancer. The review also looks at the integration of omics data into electronic health records (EHRs) and the potential for cloud computing and big data analytics to improve data storage, analysis and sharing. Despite significant advances, there are still challenges such as data complexity, technical limitations and ethical issues. Future directions include the development of more sophisticated computational tools and the application of advanced machine learning techniques, which are critical for addressing the complexity and heterogeneity of omics datasets. This review aims to serve as a valuable resource for researchers and practitioners, highlighting the transformative potential of high-throughput omics technologies in advancing personalized medicine and improving clinical outcomes.
Collapse
Affiliation(s)
- Rui Vitorino
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Surgery and Physiology, Cardiovascular R&D Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
21
|
Yang S, Hu J, Chen Y, Zhang Z, Wang J, Zhu G. DCC, a potential target for controlling fear memory extinction and hippocampal LTP in male mice receiving single prolonged stress. Neurobiol Stress 2024; 32:100666. [PMID: 39224830 PMCID: PMC11366904 DOI: 10.1016/j.ynstr.2024.100666] [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: 04/13/2024] [Revised: 06/29/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a severe stress-dependent psychiatric disorder characterized by impairment of fear memory extinction; however, biological markers to determine impaired fear memory extinction in PTSD remain unclear. In male mice with PTSD-like behaviors elicited by single prolonged stress (SPS), 19 differentially expressed proteins in the hippocampus were identified compared with controls. Among them, a biological macromolecular protein named deleted in colorectal cancer (DCC) was highly upregulated. Specific overexpression of DCC in the hippocampus induced similar impairment of long-term potentiation (LTP) and fear memory extinction as observed in SPS mice. The impairment of fear memory extinction in SPS mice was improved by inhibiting the function of hippocampal DCC using a neutralizing antibody. Mechanistic studies have shown that knocking down or inhibiting μ-calpain in hippocampal neurons increased DCC expression and induced impairment of fear memory extinction. Additionally, SPS-triggered impairment of hippocampal LTP and fear memory extinction could be rescued through activation of the Rac1-Pak1 signaling pathway. Our study provides evidence that calpain-mediated regulation of DCC controls hippocampal LTP and fear memory extinction in SPS mice, which likely through activation of the Rac1-Pak1 signaling pathway.
Collapse
Affiliation(s)
- Shaojie Yang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Shouchun Road 300, Hefei, 230061, China
| | - Jiamin Hu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Longzhihu Road 350, Hefei, 230012, China
| | - Yuzhuang Chen
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Longzhihu Road 350, Hefei, 230012, China
| | - Zhengrong Zhang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Longzhihu Road 350, Hefei, 230012, China
| | - Jingji Wang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Shouchun Road 300, Hefei, 230061, China
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Longzhihu Road 350, Hefei, 230012, China
| |
Collapse
|
22
|
Jiang Y, Rex DA, Schuster D, Neely BA, Rosano GL, Volkmar N, Momenzadeh A, Peters-Clarke TM, Egbert SB, Kreimer S, Doud EH, Crook OM, Yadav AK, Vanuopadath M, Hegeman AD, Mayta M, Duboff AG, Riley NM, Moritz RL, Meyer JG. Comprehensive Overview of Bottom-Up Proteomics Using Mass Spectrometry. ACS MEASUREMENT SCIENCE AU 2024; 4:338-417. [PMID: 39193565 PMCID: PMC11348894 DOI: 10.1021/acsmeasuresciau.3c00068] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 08/29/2024]
Abstract
Proteomics is the large scale study of protein structure and function from biological systems through protein identification and quantification. "Shotgun proteomics" or "bottom-up proteomics" is the prevailing strategy, in which proteins are hydrolyzed into peptides that are analyzed by mass spectrometry. Proteomics studies can be applied to diverse studies ranging from simple protein identification to studies of proteoforms, protein-protein interactions, protein structural alterations, absolute and relative protein quantification, post-translational modifications, and protein stability. To enable this range of different experiments, there are diverse strategies for proteome analysis. The nuances of how proteomic workflows differ may be challenging to understand for new practitioners. Here, we provide a comprehensive overview of different proteomics methods. We cover from biochemistry basics and protein extraction to biological interpretation and orthogonal validation. We expect this Review will serve as a handbook for researchers who are new to the field of bottom-up proteomics.
Collapse
Affiliation(s)
- Yuming Jiang
- Department
of Computational Biomedicine, Cedars Sinai
Medical Center, Los Angeles, California 90048, United States
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Devasahayam Arokia
Balaya Rex
- Center for
Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Dina Schuster
- Department
of Biology, Institute of Molecular Systems
Biology, ETH Zurich, Zurich 8093, Switzerland
- Department
of Biology, Institute of Molecular Biology
and Biophysics, ETH Zurich, Zurich 8093, Switzerland
- Laboratory
of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, Villigen 5232, Switzerland
| | - Benjamin A. Neely
- Chemical
Sciences Division, National Institute of
Standards and Technology, NIST, Charleston, South Carolina 29412, United States
| | - Germán L. Rosano
- Mass
Spectrometry
Unit, Institute of Molecular and Cellular
Biology of Rosario, Rosario, 2000 Argentina
| | - Norbert Volkmar
- Department
of Biology, Institute of Molecular Systems
Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Amanda Momenzadeh
- Department
of Computational Biomedicine, Cedars Sinai
Medical Center, Los Angeles, California 90048, United States
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Trenton M. Peters-Clarke
- Department
of Pharmaceutical Chemistry, University
of California—San Francisco, San Francisco, California, 94158, United States
| | - Susan B. Egbert
- Department
of Chemistry, University of Manitoba, Winnipeg, Manitoba, R3T 2N2 Canada
| | - Simion Kreimer
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Emma H. Doud
- Center
for Proteome Analysis, Indiana University
School of Medicine, Indianapolis, Indiana, 46202-3082, United States
| | - Oliver M. Crook
- Oxford
Protein Informatics Group, Department of Statistics, University of Oxford, Oxford OX1 3LB, United
Kingdom
| | - Amit Kumar Yadav
- Translational
Health Science and Technology Institute, NCR Biotech Science Cluster 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad, Haryana 121001, India
| | | | - Adrian D. Hegeman
- Departments
of Horticultural Science and Plant and Microbial Biology, University of Minnesota, Twin Cities, Minnesota 55108, United States
| | - Martín
L. Mayta
- School
of Medicine and Health Sciences, Center for Health Sciences Research, Universidad Adventista del Plata, Libertador San Martin 3103, Argentina
- Molecular
Biology Department, School of Pharmacy and Biochemistry, Universidad Nacional de Rosario, Rosario 2000, Argentina
| | - Anna G. Duboff
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Nicholas M. Riley
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Robert L. Moritz
- Institute
for Systems biology, Seattle, Washington 98109, United States
| | - Jesse G. Meyer
- Department
of Computational Biomedicine, Cedars Sinai
Medical Center, Los Angeles, California 90048, United States
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| |
Collapse
|
23
|
van Bergen W, Žuna K, Fiala J, Pohl EE, Heck AJ, Baggelaar MP. Dual-Probe Activity-Based Protein Profiling Reveals Site-Specific Differences in Protein Binding of EGFR-Directed Drugs. ACS Chem Biol 2024; 19:1705-1718. [PMID: 39052621 PMCID: PMC11334109 DOI: 10.1021/acschembio.3c00637] [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: 10/19/2023] [Revised: 06/24/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Comparative, dose-dependent analysis of interactions between small molecule drugs and their targets, as well as off-target interactions, in complex proteomes is crucial for selecting optimal drug candidates. The affinity of small molecules for targeted proteins is largely dictated by interactions between amino acid side chains and these drugs. Thus, studying drug-protein interactions at an amino acid resolution provides a comprehensive understanding of the drug selectivity and efficacy. In this study, we further refined the site-specific activity-based protein profiling strategy (ABPP), PhosID-ABPP, on a timsTOF HT mass spectrometer. This refinement enables dual dose-dependent competition of inhibitors within a single cellular proteome. Here, a comparative analysis of two activity-based probes (ABPs), developed to selectively target the epidermal growth factor receptor (EGFR), namely, PF-06672131 (PF131) and PF-6422899 (PF899), facilitated the simultaneous identification of ABP-specific binding sites at a proteome-wide scale within a cellular proteome. Dose-dependent probe-binding preferences for proteinaceous cysteines, even at low nanomolar ABP concentrations, could be revealed. Notably, in addition to the intrinsic affinity of the electrophilic probes for specific sites in targeted proteins, the observed labeling intensity is influenced by several other factors. These include the efficiency of cellular uptake, the stability of the probes, and their intracellular distribution. While both ABPs showed comparable labeling efficiency for EGFR, PF131 had a broader off-target reactivity profile. In contrast, PF899 exhibited a higher labeling efficiency for the ERBB2 receptor and bound to catalytic cysteines in several other enzymes, which is likely to disrupt their catalytic activity. Notably, PF131 effectively labeled ADP/ATP translocase proteins at a concentration of just 1 nm, and we found this affected ATP transport. Analysis of the effect of PF131 and its parent inhibitor Afatinib on murine translocase SLC25A4 (ANT1)-mediated ATP transport strongly indicated that PF131 (10 μM) partially blocked ATP transport. Afatinib was less efficient at inhibiting ATP transport by SLC25A4 than PF131, and the reduction of ATP transport by Afatinib was not significant. Follow-up analysis is required to evaluate the affinity of these inhibitors for ADP/ATP translocase SLC25A4 in more detail. Additionally, the analysis of different binding sites within the EGF receptor and the voltage-dependent anion channel 2 revealed secondary binding sites of both probes and provided insights into the binding poses of inhibitors on these proteins. Insights from the PhosID-ABPP analysis of these two ABPs serve as a valuable resource for understanding drug on- and off-target engagement in a dose- and site-specific manner.
Collapse
Affiliation(s)
- Wouter van Bergen
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, Utrecht 3584 CH, The Netherlands
| | - Kristina Žuna
- Physiology
and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Wien, Vienna 1210, Austria
| | - Jan Fiala
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, Utrecht 3584 CH, The Netherlands
| | - Elena E. Pohl
- Physiology
and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine, Wien, Vienna 1210, Austria
| | - Albert J.R. Heck
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, Utrecht 3584 CH, The Netherlands
| | - Marc P. Baggelaar
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, Utrecht 3584 CH, The Netherlands
| |
Collapse
|
24
|
Guo Y, Wen H, Chen Z, Jiao M, Zhang Y, Ge D, Liu R, Gu J. Conjoint analysis of succinylome and phosphorylome reveals imbalanced HDAC phosphorylation-driven succinylayion dynamic contibutes to lung cancer. Brief Bioinform 2024; 25:bbae415. [PMID: 39179249 PMCID: PMC11343571 DOI: 10.1093/bib/bbae415] [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: 04/16/2024] [Revised: 07/17/2024] [Indexed: 08/26/2024] Open
Abstract
Cancerous genetic mutations result in a complex and comprehensive post-translational modification (PTM) dynamics, in which protein succinylation is well known for its ability to reprogram cell metabolism and is involved in the malignant evolution. Little is known about the regulatory interactions between succinylation and other PTMs in the PTM network. Here, we developed a conjoint analysis and systematic clustering method to explore the intermodification communications between succinylome and phosphorylome from eight lung cancer patients. We found that the intermodification coorperation in both parallel and series. Besides directly participating in metabolism pathways, some phosphosites out of mitochondria were identified as an upstream regulatory modification directing succinylome dynamics in cancer metabolism reprogramming. Phosphorylated activation of histone deacetylase (HDAC) in lung cancer resulted in the removal of acetylation and favored the occurrence of succinylation modification of mitochondrial proteins. These results suggest a tandem regulation between succinylation and phosphorylation in the PTM network and provide HDAC-related targets for intervening mitochondrial succinylation and cancer metabolism reprogramming.
Collapse
Affiliation(s)
- Yifan Guo
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Haoyu Wen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Zongwei Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Mengxia Jiao
- Shanghai Fifth People's Hospital and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 131 Dongan Road, Shanghai 200032, China
| | - Yuchen Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Ronghua Liu
- Shanghai Fifth People's Hospital and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 131 Dongan Road, Shanghai 200032, China
| | - Jie Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
25
|
Jiang Y, Ren X, Zhao J, Liu G, Liu F, Guo X, Hao M, Liu H, Liu K, Huang H. Exploring the Molecular Therapeutic Mechanisms of Gemcitabine through Quantitative Proteomics. J Proteome Res 2024; 23:2343-2354. [PMID: 38831540 DOI: 10.1021/acs.jproteome.3c00890] [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: 06/05/2024]
Abstract
Gemcitabine (GEM) is widely employed in the treatment of various cancers, including pancreatic cancer. Despite their clinical success, challenges related to GEM resistance and toxicity persist. Therefore, a deeper understanding of its intracellular mechanisms and potential targets is urgently needed. In this study, through mass spectrometry analysis in data-dependent acquisition mode, we carried out quantitative proteomics (three independent replications) and thermal proteome profiling (TPP, two independent replications) on MIA PaCa-2 cells to explore the effects of GEM. Our proteomic analysis revealed that GEM led to the upregulation of the cell cycle and DNA replication proteins. Notably, we observed the upregulation of S-phase kinase-associated protein 2 (SKP2), a cell cycle and chemoresistance regulator. Combining SKP2 inhibition with GEM showed synergistic effects, suggesting SKP2 as a potential target for enhancing the GEM sensitivity. Through TPP, we pinpointed four potential GEM binding targets implicated in tumor development, including in breast and liver cancers, underscoring GEM's broad-spectrum antitumor capabilities. These findings provide valuable insights into GEM's molecular mechanisms and offer potential targets for improving treatment efficacy.
Collapse
Affiliation(s)
- Yue Jiang
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xuelian Ren
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Jing Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guobin Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fangfang Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinlong Guo
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Ming Hao
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kun Liu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang 110819, China
| | - He Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| |
Collapse
|
26
|
Karpov OA, Stotland A, Raedschelders K, Chazarin B, Ai L, Murray CI, Van Eyk JE. Proteomics of the heart. Physiol Rev 2024; 104:931-982. [PMID: 38300522 PMCID: PMC11381016 DOI: 10.1152/physrev.00026.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/25/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Mass spectrometry-based proteomics is a sophisticated identification tool specializing in portraying protein dynamics at a molecular level. Proteomics provides biologists with a snapshot of context-dependent protein and proteoform expression, structural conformations, dynamic turnover, and protein-protein interactions. Cardiac proteomics can offer a broader and deeper understanding of the molecular mechanisms that underscore cardiovascular disease, and it is foundational to the development of future therapeutic interventions. This review encapsulates the evolution, current technologies, and future perspectives of proteomic-based mass spectrometry as it applies to the study of the heart. Key technological advancements have allowed researchers to study proteomes at a single-cell level and employ robot-assisted automation systems for enhanced sample preparation techniques, and the increase in fidelity of the mass spectrometers has allowed for the unambiguous identification of numerous dynamic posttranslational modifications. Animal models of cardiovascular disease, ranging from early animal experiments to current sophisticated models of heart failure with preserved ejection fraction, have provided the tools to study a challenging organ in the laboratory. Further technological development will pave the way for the implementation of proteomics even closer within the clinical setting, allowing not only scientists but also patients to benefit from an understanding of protein interplay as it relates to cardiac disease physiology.
Collapse
Affiliation(s)
- Oleg A Karpov
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Aleksandr Stotland
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Koen Raedschelders
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Blandine Chazarin
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Lizhuo Ai
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Christopher I Murray
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| |
Collapse
|
27
|
Wang J, Tan H, Fu Y, Mishra A, Sun H, Wang Z, Wu Z, Wang X, Serrano GE, Beach TG, Peng J, High AA. Evaluation of Protein Identification and Quantification by the diaPASEF Method on timsTOF SCP. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1253-1260. [PMID: 38754071 DOI: 10.1021/jasms.4c00067] [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: 05/18/2024]
Abstract
Accurate and precise quantification is crucial in modern proteomics, particularly in the context of exploring low-amount samples. While the innovative 4D-data-independent acquisition (DIA) quantitative proteomics facilitated by timsTOF mass spectrometers gives enhanced sensitivity and selectivity for protein identification, the diaPASEF (parallel accumulation-serial fragmentation combined with data-independent acquisition) parameters have not been systematically optimized, and a comprehensive evaluation of the quantification is currently lacking. In this study, we conducted a thorough optimization of key parameters on a timsTOF SCP instrument, including sample loading amount (50 ng), ramp/accumulation time (140 ms), isolation window width (20 m/z), and gradient time (60 min). To further improve the identification of proteins in low-amount samples, we utilized different column settings and introduced 0.02% n-dodecyl-β-d-maltoside (DDM) in the sample reconstitution solution, resulting in a remarkable 19-fold increase in protein identification at the single-cell-equivalent level. Moreover, a comprehensive comparison of protein quantification using a tandem mass tag reporter (TMT-reporter), complement TMT ions (TMTc), and diaPASEF revealed a strong correlation between these methods. Both diaPASEF and TMTc have effectively addressed the issue of ratio compression, highlighting the diaPASEF method's effectiveness in achieving accurate quantification data compared to TMT reporter quantification. Additionally, an in-depth analysis of in-group variation positioned diaPASEF between the TMT-reporter and TMTc methods. Therefore, diaPASEF quantification on the timsTOF SCP instrument emerges as a precise and accurate methodology for quantitative proteomics, especially for samples with small amounts.
Collapse
Affiliation(s)
- Ju Wang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Haiyan Tan
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Yingxue Fu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Ashutosh Mishra
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Huan Sun
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Zhen Wang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Zhiping Wu
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Geidy E Serrano
- Banner Sun Health Research Institute, Sun City, Arizona 85351, United States
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, Arizona 85351, United States
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Anthony A High
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| |
Collapse
|
28
|
Jiang Y, Ren X, Liu G, Chen S, Hao M, Deng X, Huang H, Liu K. Exploring the mechanism of contact-dependent cell-cell communication on chemosensitivity based on single-cell high-throughput drug screening platform. Talanta 2024; 273:125869. [PMID: 38490027 DOI: 10.1016/j.talanta.2024.125869] [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: 11/30/2023] [Revised: 02/23/2024] [Accepted: 03/01/2024] [Indexed: 03/17/2024]
Abstract
High-throughput drug screening (HTDS) has significantly reduced the time and cost of new drug development. Nonetheless, contact-dependent cell-cell communication (CDCCC) may impact the chemosensitivity of tumour cells. There is a pressing need for low-cost single-cell HTDS platforms, alongside a deep comprehension of the mechanisms by which CDCCC affects drug efficacy, to fully unveil the efficacy of anticancer drugs. In this study, we develop a microfluidic chip for single-cell HTDS and evaluate the molecular mechanisms impacted by CDCCC using quantitative mass spectrometry-based proteomics. The chip achieves high-quality drug mixing and single-cell capture, with single-cell drug screening results on the chip showing consistency with those on the 96-well plates under varying concentration gradients. Through quantitative proteomic analysis, we deduce that the absence of CDCCC in single tumour cells can enhance their chemoresistance potential, but simultaneously subject them to stronger proliferation inhibition. Additionally, pathway enrichment analysis suggests that CDCCC could impact several signalling pathways in tumour single cells that regulate vital biological processes such as tumour proliferation, adhesion, and invasion. These results offer valuable insights into the potential connection between CDCCC and the chemosensitivity of tumour cells. This research paves the way for the development of single-cell HTDC platforms and holds the promise of advancing tumour personalized treatment strategies.
Collapse
Affiliation(s)
- Yue Jiang
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xuelian Ren
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Guobin Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shulei Chen
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Ming Hao
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Xinran Deng
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - He Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Kun Liu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China; National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China; Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, China.
| |
Collapse
|
29
|
Sun T, Chen J, Xu Y, Li Y, Liu X, Li H, Fu R, Liu W, Xue F, Ju M, Dong H, Wang W, Chi Y, Yang R, Chen Y, Zhang L. Proteomics landscape and machine learning prediction of long-term response to splenectomy in primary immune thrombocytopenia. Br J Haematol 2024; 204:2418-2428. [PMID: 38513635 DOI: 10.1111/bjh.19420] [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: 12/28/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
This study aimed to identify key proteomic analytes correlated with response to splenectomy in primary immune thrombocytopenia (ITP). Thirty-four patients were retrospectively collected in the training cohort and 26 were prospectively enrolled as validation cohort. Bone marrow biopsy samples of all participants were collected prior to the splenectomy. A total of 12 modules of proteins were identified by weighted gene co-expression network analysis (WGCNA) method in the developed cohort. The tan module positively correlated with megakaryocyte counts before splenectomy (r = 0.38, p = 0.027), and time to peak platelet level after splenectomy (r = 0.47, p = 0.005). The blue module significantly correlated with response to splenectomy (r = 0.37, p = 0.0031). KEGG pathways analysis found that the PI3K-Akt signalling pathway was predominantly enriched in the tan module, while ribosomal and spliceosome pathways were enriched in the blue module. Machine learning algorithm identified the optimal combination of biomarkers from the blue module in the training cohort, and importantly, cofilin-1 (CFL1) was independently confirmed in the validation cohort. The C-index of CFL1 was >0.7 in both cohorts. Our results highlight the use of bone marrow proteomics analysis for deriving key analytes that predict the response to splenectomy, warranting further exploration of plasma proteomics in this patient population.
Collapse
Affiliation(s)
- Ting Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jia Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yang Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaofan Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huiyuan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Rongfeng Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Feng Xue
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mankai Ju
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huan Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wentian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ying Chi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Renchi Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yunfei Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Kong B, Owens C, Bottje W, Shakeri M, Choi J, Zhuang H, Bowker B. Proteomic analyses on chicken breast meat with white striping myopathy. Poult Sci 2024; 103:103682. [PMID: 38593545 PMCID: PMC11016796 DOI: 10.1016/j.psj.2024.103682] [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: 12/14/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024] Open
Abstract
White striping (WS) is an emerging myopathy that results in significant economic losses as high as $1 billion (combined with losses derived from other breast myopathies including woody breast and spaghetti meat) to the global poultry industry. White striping is detected as the occurrence of white lines on raw poultry meat. The exact etiologies for WS are still unclear. Proteomic analyses of co-expressed WS and woody breast phenotypes previously demonstrated dysfunctions in carbohydrate metabolism, protein synthesis, and calcium buffering capabilities in muscle cells. In this study, we conducted shotgun proteomics on chicken breast fillets exhibiting only WS that were collected at approximately 6 h postmortem. After determining WS severity, protein extractions were conducted from severe WS meat with no woody breast (WB) condition (n = 5) and normal non-affected (no WS) control meat (n = 5). Shotgun proteomics was conducted by Orbitrap Lumos, tandem mass tag (TMT) analysis. As results, 148 differentially abundant proteins (|fold change|>1.4; p-value < 0.05) were identified in the WS meats compared with controls. The significant canonical pathways included BAG2 signaling pathway, glycogen degradation II, isoleucine degradation I, aldosterone signaling in epithelial cells, and valine degradation I. The potential upstream regulators include LIPE, UCP1, ATP5IF1, and DMD. The results of this study provide additional insights into the cellular mechanisms on the WS myopathy and meat quality.
Collapse
Affiliation(s)
- Byungwhi Kong
- USDA, Agricultural Research Service, U.S. National Poultry Research Center, Quality & Safety Assessment Research Unit, Athens, GA, USA.
| | - Casey Owens
- Department of Poultry Science, Division of Agriculture, University of Arkansas System, Fayetteville, AR, USA
| | - Walter Bottje
- Department of Poultry Science, Division of Agriculture, University of Arkansas System, Fayetteville, AR, USA
| | - Majid Shakeri
- USDA, Agricultural Research Service, U.S. National Poultry Research Center, Quality & Safety Assessment Research Unit, Athens, GA, USA
| | - Janghan Choi
- USDA, Agricultural Research Service, U.S. National Poultry Research Center, Quality & Safety Assessment Research Unit, Athens, GA, USA
| | - Hong Zhuang
- USDA, Agricultural Research Service, U.S. National Poultry Research Center, Quality & Safety Assessment Research Unit, Athens, GA, USA
| | - Brian Bowker
- USDA, Agricultural Research Service, U.S. National Poultry Research Center, Quality & Safety Assessment Research Unit, Athens, GA, USA
| |
Collapse
|
31
|
Pham T, Chen Y, Labaer J, Guo J. Ultrasensitive and Multiplexed Protein Imaging with Clickable and Cleavable Fluorophores. Anal Chem 2024; 96:7281-7288. [PMID: 38663032 DOI: 10.1021/acs.analchem.4c01273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Single-cell spatial proteomic analysis holds great promise to advance our understanding of the composition, organization, interaction, and function of the various cell types in complex biological systems. However, the current multiplexed protein imaging technologies suffer from low detection sensitivity, limited multiplexing capacity, or are technically demanding. To tackle these issues, here, we report the development of a highly sensitive and multiplexed in situ protein profiling method using off-the-shelf antibodies. In this approach, the protein targets are stained with horseradish peroxidase (HRP) conjugated antibodies and cleavable fluorophores via click chemistry. Through repeated cycles of target staining, fluorescence imaging, and fluorophore cleavage, many proteins can be profiled in single cells in situ. Applying this approach, we successfully quantified 28 different proteins in human formalin-fixed paraffin-embedded (FFPE) tonsil tissue, which represents the highest multiplexing capacity among the tyramide signal amplification (TSA) methods. Based on their unique protein expression patterns and their microenvironment, ∼820,000 cells in the tissue are classified into distinct cell clusters. We also explored the cell-cell interactions between these varied cell clusters and observed that different subregions of the tissue are composed of cells from specific clusters.
Collapse
Affiliation(s)
- Thai Pham
- Biodesign Institute & School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Yi Chen
- Biodesign Institute & School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Joshua Labaer
- Biodesign Institute & School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Jia Guo
- Biodesign Institute & School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
32
|
Weng Y, Chen W, Kong Q, Wang R, Zeng R, He A, Liu Y, Mao Y, Qin Y, Ngai WSC, Zhang H, Ke M, Wang J, Tian R, Chen PR. DeKinomics pulse-chases kinase functions in living cells. Nat Chem Biol 2024; 20:615-623. [PMID: 38167916 DOI: 10.1038/s41589-023-01497-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024]
Abstract
Cellular context is crucial for understanding the complex and dynamic kinase functions in health and disease. Systematic dissection of kinase-mediated cellular processes requires rapid and precise stimulation ('pulse') of a kinase of interest, as well as global and in-depth characterization ('chase') of the perturbed proteome under living conditions. Here we developed an optogenetic 'pulse-chase' strategy, termed decaging kinase coupled proteomics (DeKinomics), for proteome-wide profiling of kinase-driven phosphorylation at second-timescale in living cells. We took advantage of the 'gain-of-function' feature of DeKinomics to identify direct kinase substrates and further portrayed the global phosphorylation of understudied receptor tyrosine kinases under native cellular settings. DeKinomics offered a general activation-based strategy to study kinase functions with high specificity and temporal resolution under living conditions.
Collapse
Affiliation(s)
- Yicheng Weng
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, New Cornerstone Science Laboratory, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Wendong Chen
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China
- South China Institute of Biomedicine, Academy of Phronesis Medicine, Guangzhou, China
| | - Qian Kong
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China
| | - Ruixiang Wang
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, New Cornerstone Science Laboratory, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Ruxin Zeng
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, New Cornerstone Science Laboratory, College of Chemistry and Molecular Engineering, Peking University, Beijing, 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, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China
| | - Yanjun Liu
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, New Cornerstone Science Laboratory, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Yiheng Mao
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China
| | - Yunqiu Qin
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China
| | | | - Heng Zhang
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Mi Ke
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China
| | - Jie Wang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, School of Science, Southern University of Science and Technology, Shenzhen, 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, China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China.
| | - Peng R Chen
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, New Cornerstone Science Laboratory, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
33
|
Chu G, Li YM. Lighting up kinase contacts in situ. Nat Chem Biol 2024; 20:544-545. [PMID: 38302605 DOI: 10.1038/s41589-024-01543-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Affiliation(s)
- Guochao Chu
- School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, China
| | - Yi-Ming Li
- School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, China.
| |
Collapse
|
34
|
Makey DM, Gadkari VV, Kennedy RT, Ruotolo BT. Cyclic Ion Mobility-Mass Spectrometry and Tandem Collision Induced Unfolding for Quantification of Elusive Protein Biomarkers. Anal Chem 2024; 96:6021-6029. [PMID: 38557001 PMCID: PMC11081454 DOI: 10.1021/acs.analchem.4c00477] [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] [Indexed: 04/04/2024]
Abstract
Sensitive analytical techniques that are capable of detecting and quantifying disease-associated biomolecules are indispensable in our efforts to understand disease mechanisms and guide therapeutic intervention through early detection, accurate diagnosis, and effective monitoring of disease. Parkinson's Disease (PD), for example, is one of the most prominent neurodegenerative disorders in the world, but the diagnosis of PD has primarily been based on the observation of clinical symptoms. The protein α-synuclein (α-syn) has emerged as a promising biomarker candidate for PD, but a lack of analytical methods to measure complex disease-associated variants of α-syn has prevented its widespread use as a biomarker. Antibody-based methods such as immunoassays and mass spectrometry-based approaches have been used to measure a limited number of α-syn forms; however, these methods fail to differentiate variants of α-syn that display subtle differences in only the sequence and structure. In this work, we developed a cyclic ion mobility-mass spectrometry method that combines multiple stages of activation and timed ion selection to quantify α-syn variants using both mass- and structure-based measurements. This method can allow for the quantification of several α-syn variants present at physiological levels in biological fluid. Taken together, this approach can be used to galvanize future efforts aimed at understanding the underlying mechanisms of PD and serves as a starting point for the development of future protein-structure-based diagnostics and therapeutic interventions.
Collapse
Affiliation(s)
- Devin M. Makey
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Varun V. Gadkari
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Robert T. Kennedy
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brandon T. Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
35
|
Panda A, Falasca M, Ragunath K. Extracellular vesicles in pancreatic cancer: a new era in precision medicine. Transl Gastroenterol Hepatol 2024; 9:29. [PMID: 38716212 PMCID: PMC11074477 DOI: 10.21037/tgh-23-53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/31/2023] [Indexed: 01/04/2025] Open
Abstract
Pancreatic cancer (PC) is a lethal disease that presents a considerable challenge to healthcare providers and patients, given its low survival rate. However, recent advancements in precision medicine and innovative technologies have transformed the management of this disease. Among these advancements, extracellular vesicles (EVs) have emerged as crucial players in cancer progression. In PC, EVs play a pivotal role by facilitating cell-cell communication, impeding immune response, promoting cancer cell proliferation and survival, and supporting angiogenesis and chemoresistance. Cancer-derived EVs have a distinct oncogenic composition supporting tumour development and progression. Hence, they are critical biomarker candidates for various cancers, including PC. Notably, EVs can be isolated from diverse biological fluids such as blood, urine, and saliva, making them an ideal minimally invasive diagnostic and monitoring tool for PC patients. Despite the promising findings in the field of EVs, clinical validation as biomarkers is lacking. Furthermore, EVs being biocompatible, can act as drug carriers, delivering therapeutic molecules directly to cancer cells while minimizing toxicity to healthy cells. Therefore, understanding the role of EVs as biomarkers and their potential as drug cargo vehicles may revolutionise early detection, prognostication, and treatment in cancer. This mini-review summarises the latest understanding of their role in intercellular communication, involvement as potential biomarkers and drug carriers.
Collapse
Affiliation(s)
- Arunima Panda
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Marco Falasca
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Krish Ragunath
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
- Department of Gastroenterology and Hepatology, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
36
|
Nava AA, Arboleda VA. The omics era: a nexus of untapped potential for Mendelian chromatinopathies. Hum Genet 2024; 143:475-495. [PMID: 37115317 PMCID: PMC11078811 DOI: 10.1007/s00439-023-02560-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 04/10/2023] [Indexed: 04/29/2023]
Abstract
The OMICs cascade describes the hierarchical flow of information through biological systems. The epigenome sits at the apex of the cascade, thereby regulating the RNA and protein expression of the human genome and governs cellular identity and function. Genes that regulate the epigenome, termed epigenes, orchestrate complex biological signaling programs that drive human development. The broad expression patterns of epigenes during human development mean that pathogenic germline mutations in epigenes can lead to clinically significant multi-system malformations, developmental delay, intellectual disabilities, and stem cell dysfunction. In this review, we refer to germline developmental disorders caused by epigene mutation as "chromatinopathies". We curated the largest number of human chromatinopathies to date and our expanded approach more than doubled the number of established chromatinopathies to 179 disorders caused by 148 epigenes. Our study revealed that 20.6% (148/720) of epigenes cause at least one chromatinopathy. In this review, we highlight key examples in which OMICs approaches have been applied to chromatinopathy patient biospecimens to identify underlying disease pathogenesis. The rapidly evolving OMICs technologies that couple molecular biology with high-throughput sequencing or proteomics allow us to dissect out the causal mechanisms driving temporal-, cellular-, and tissue-specific expression. Using the full repertoire of data generated by the OMICs cascade to study chromatinopathies will provide invaluable insight into the developmental impact of these epigenes and point toward future precision targets for these rare disorders.
Collapse
Affiliation(s)
- Aileen A Nava
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Valerie A Arboleda
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
- Department of Computational Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Jurkovic CM, Boisvert FM. Evolution of techniques and tools for replication fork proteome and protein interaction studies. Biochem Cell Biol 2024; 102:135-144. [PMID: 38113480 DOI: 10.1139/bcb-2023-0215] [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: 12/21/2023] Open
Abstract
Understanding the complex network of protein-protein interactions (PPI) that govern cellular functions is essential for unraveling the molecular basis of biological processes and diseases. Mass spectrometry (MS) has emerged as a powerful tool for studying protein dynamics, enabling comprehensive analysis of protein function, structure, post-translational modifications, interactions, and localization. This article provides an overview of MS techniques and their applications in proteomics studies, with a focus on the replication fork proteome. The replication fork is a multi-protein assembly involved in DNA replication, and its proper functioning is crucial for maintaining genomic integrity. By combining quantitative MS labeling techniques with various data acquisition methods, researchers have made significant strides in elucidating the complex processes and molecular mechanisms at the replication fork. Overall, MS has revolutionized our understanding of protein dynamics, offering valuable insights into cellular processes and potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Carla-Marie Jurkovic
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - François-Michel Boisvert
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
38
|
Yuan L, Zhao J, Liu Y, Zhao J, Olnood CG, Xu YJ, Liu Y. Multiomics analysis revealed the mechanism of the anti-diabetic effect of Salecan. Carbohydr Polym 2024; 327:121694. [PMID: 38171651 DOI: 10.1016/j.carbpol.2023.121694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024]
Abstract
Salecan, a natural β-glucan compromising nine residues connected by β-(1 → 3)/α-(1 → 3) glycosidic bonds, is one of the newly approved food ingredients. Salecan has multiple health-improving effects, yet its mechanism against Type 2 diabetes mellitus (T2DM) remains poorly understood. In this study, the hypoglycemic effect and underlying mechanism of Salecan intervention on STZ-induced diabetic model mice were investigated. After 8 weeks of gavage, Salecan attenuated insulin resistance and repaired pancreatic β cells in a dose-dependent manner. In addition, Salecan supplement remodel the structure of the gut microbiota and altered the level of intestinal metabolites. Serum metabolites, especially unsaturated fatty acids, were also affected significantly. In addition, tight junction proteins in the colon and autophagy-related proteins in the pancreas were upregulated. Multiomics analysis indicated that Lactobacillus johnsonii, Muribaculaceae, and Lachnoclostridium were highly associated with fatty acid esters of hydroxy fatty acids (FAHFA) levels in the colon, accordingly enhancing arachidonic acid and linoleic acid in serum, and promoting GLP-1 release in the intestine and insulin secretion in the pancreas, thus relieving insulin resistance and exhibiting hypoglycemic effects. These findings provide a novel understanding of the anti-diabetic effect of Salecan in mice from a molecular perspective, paving the way for the wide use of Salecan.
Collapse
Affiliation(s)
- Liyang Yuan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Juan Zhao
- Sichuan Synlight Biotech Ltd, 88 Keyuan South Road, Chengdu 610000, Sichuan, China
| | - Yanjun Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Jialiang Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
| | - Chen Guang Olnood
- Sichuan Synlight Biotech Ltd, 88 Keyuan South Road, Chengdu 610000, Sichuan, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China.
| | - Yuanfa Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China.
| |
Collapse
|
39
|
Zubeldia-Varela E, Ibáñez-Sandín MD, Gomez-Casado C, Pérez-Gordo M. Allergy-associated biomarkers in early life identified by Omics techniques. FRONTIERS IN ALLERGY 2024; 5:1359142. [PMID: 38464396 PMCID: PMC10920277 DOI: 10.3389/falgy.2024.1359142] [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: 12/20/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024] Open
Abstract
The prevalence and severity of allergic diseases have increased over the last 30 years. Understanding the mechanisms responsible for these diseases is a major challenge in current allergology, as it is crucial for the transition towards precision medicine, which encompasses predictive, preventive, and personalized strategies. The urge to identify predictive biomarkers of allergy at early stages of life is crucial, especially in the context of major allergic diseases such as food allergy and atopic dermatitis. Identifying these biomarkers could enhance our understanding of the immature immune responses, improve allergy handling at early ages and pave the way for preventive and therapeutic approaches. This minireview aims to explore the relevance of three biomarker categories (proteome, microbiome, and metabolome) in early life. First, levels of some proteins emerge as potential indicators of mucosal health and metabolic status in certain allergic diseases. Second, bacterial taxonomy provides insight into the composition of the microbiota through high-throughput sequencing methods. Finally, metabolites, representing the end products of bacterial and host metabolic activity, serve as early indicators of changes in microbiota and host metabolism. This information could help to develop an extensive identification of biomarkers in AD and FA and their potential in translational personalized medicine in early life.
Collapse
Affiliation(s)
- Elisa Zubeldia-Varela
- Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Department of Basic Medical Sciences, Facultad de Medicina. Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - María Dolores Ibáñez-Sandín
- Department of Allergy, H. Infantil Universitario Niño Jesús, FibHNJ, ARADyAL- RETICs Instituto de Salud Carlos III, IIS-P, Madrid, Spain
| | - Cristina Gomez-Casado
- Department of Dermatology, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Marina Pérez-Gordo
- Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Department of Basic Medical Sciences, Facultad de Medicina. Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
40
|
Ren Y, Gao Y, Du W, Qiao W, Li W, Yang Q, Liang Y, Li G. Classifying breast cancer using multi-view graph neural network based on multi-omics data. Front Genet 2024; 15:1363896. [PMID: 38444760 PMCID: PMC10912483 DOI: 10.3389/fgene.2024.1363896] [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: 12/31/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Introduction: As the evaluation indices, cancer grading and subtyping have diverse clinical, pathological, and molecular characteristics with prognostic and therapeutic implications. Although researchers have begun to study cancer differentiation and subtype prediction, most of relevant methods are based on traditional machine learning and rely on single omics data. It is necessary to explore a deep learning algorithm that integrates multi-omics data to achieve classification prediction of cancer differentiation and subtypes. Methods: This paper proposes a multi-omics data fusion algorithm based on a multi-view graph neural network (MVGNN) for predicting cancer differentiation and subtype classification. The model framework consists of a graph convolutional network (GCN) module for learning features from different omics data and an attention module for integrating multi-omics data. Three different types of omics data are used. For each type of omics data, feature selection is performed using methods such as the chi-square test and minimum redundancy maximum relevance (mRMR). Weighted patient similarity networks are constructed based on the selected omics features, and GCN is trained using omics features and corresponding similarity networks. Finally, an attention module integrates different types of omics features and performs the final cancer classification prediction. Results: To validate the cancer classification predictive performance of the MVGNN model, we conducted experimental comparisons with traditional machine learning models and currently popular methods based on integrating multi-omics data using 5-fold cross-validation. Additionally, we performed comparative experiments on cancer differentiation and its subtypes based on single omics data, two omics data, and three omics data. Discussion: This paper proposed the MVGNN model and it performed well in cancer classification prediction based on multiple omics data.
Collapse
Affiliation(s)
- Yanjiao Ren
- College of Information Technology, Smart Agriculture Research Institute, Jilin Agricultural University, Changchun, Jilin, China
| | - Yimeng Gao
- College of Information Technology, Smart Agriculture Research Institute, Jilin Agricultural University, Changchun, Jilin, China
| | - Wei Du
- College of Computer Science and Technology, Jilin University, Changchun, China
| | - Weibo Qiao
- College of Computer Science and Technology, Jilin University, Changchun, China
| | - Wei Li
- College of Information Technology, Smart Agriculture Research Institute, Jilin Agricultural University, Changchun, Jilin, China
| | - Qianqian Yang
- College of Information Technology, Smart Agriculture Research Institute, Jilin Agricultural University, Changchun, Jilin, China
| | - Yanchun Liang
- College of Computer Science and Technology, Jilin University, Changchun, China
- School of Computer Science, Zhuhai College of Science and Technology, Zhuhai, China
| | - Gaoyang Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
41
|
Fu L, Guldiken N, Remih K, Karl AS, Preisinger C, Strnad P. Serum/Plasma Proteome in Non-Malignant Liver Disease. Int J Mol Sci 2024; 25:2008. [PMID: 38396688 PMCID: PMC10889128 DOI: 10.3390/ijms25042008] [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: 12/22/2023] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
The liver is the central metabolic organ and produces 85-90% of the proteins found in plasma. Accordingly, the plasma proteome is an attractive source of liver disease biomarkers that reflects the different cell types present in this organ, as well as the processes such as responses to acute and chronic injury or the formation of an extracellular matrix. In the first part, we summarize the biomarkers routinely used in clinical evaluations and their biological relevance in the different stages of non-malignant liver disease. Later, we describe the current proteomic approaches, including mass spectrometry and affinity-based techniques, that allow a more comprehensive assessment of the liver function but also require complex data processing. The many approaches of analysis and interpretation and their potential caveats are delineated. While these advances hold the promise to transform our understanding of liver diseases and support the development and validation of new liver-related drugs, an interdisciplinary collaboration is needed.
Collapse
Affiliation(s)
- Lei Fu
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (L.F.); (N.G.); (K.R.); (A.S.K.)
| | - Nurdan Guldiken
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (L.F.); (N.G.); (K.R.); (A.S.K.)
| | - Katharina Remih
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (L.F.); (N.G.); (K.R.); (A.S.K.)
| | - Anna Sophie Karl
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (L.F.); (N.G.); (K.R.); (A.S.K.)
| | - Christian Preisinger
- Proteomics Facility, Interdisciplinary Centre for Clinical Research (IZKF), Medical School, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany;
| | - Pavel Strnad
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (L.F.); (N.G.); (K.R.); (A.S.K.)
| |
Collapse
|
42
|
Karlo J, Dhillon AK, Siddhanta S, Singh SP. Reverse stable isotope labelling with Raman spectroscopy for microbial proteomics. JOURNAL OF BIOPHOTONICS 2024; 17:e202300341. [PMID: 38010366 DOI: 10.1002/jbio.202300341] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/29/2023]
Abstract
Global proteome changes in microbes affect the survival and overall production of commercially relevant metabolites through different bioprocesses. The existing methods to monitor proteome level changes are destructive in nature. Stable isotope probing (SIP) coupled with Raman spectroscopy is a relatively new approach for proteome analysis. However, applying this approach for monitoring changes in a large culture volume is not cost-effective. In this study, for the first time we are presenting a novel method of combining reverse SIP using 13 C-glucose and Deuterium to monitor the proteome changes through Raman spectroscopy. The findings of the study revealed visible changes (blue shifts) in proteome related peaks that can be used for monitoring proteome dynamics, that is, synthesis of nascent amino acids and its turnover with time in a non-destructive, cost-effective, and label-free manner.
Collapse
Affiliation(s)
- Jiro Karlo
- Department of Biosciences and Bioengineering, Indian Institute of Technology Dharwad, Dharwad, India
| | | | - Soumik Siddhanta
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi, India
| | - Surya Pratap Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Dharwad, Dharwad, India
| |
Collapse
|
43
|
Watkins JM, Montes C, Clark NM, Song G, Oliveira CC, Mishra B, Brachova L, Seifert CM, Mitchell MS, Yang J, Braga Dos Reis PA, Urano D, Muktar MS, Walley JW, Jones AM. Phosphorylation Dynamics in a flg22-Induced, G Protein-Dependent Network Reveals the AtRGS1 Phosphatase. Mol Cell Proteomics 2024; 23:100705. [PMID: 38135118 PMCID: PMC10837098 DOI: 10.1016/j.mcpro.2023.100705] [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: 04/03/2023] [Revised: 11/22/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023] Open
Abstract
The microbe-associated molecular pattern flg22 is recognized in a flagellin-sensitive 2-dependent manner in root tip cells. Here, we show a rapid and massive change in protein abundance and phosphorylation state of the Arabidopsis root cell proteome in WT and a mutant deficient in heterotrimeric G-protein-coupled signaling. flg22-induced changes fall on proteins comprising a subset of this proteome, the heterotrimeric G protein interactome, and on highly-populated hubs of the immunity network. Approximately 95% of the phosphorylation changes in the heterotrimeric G-protein interactome depend, at least partially, on a functional G protein complex. One member of this interactome is ATBα, a substrate-recognition subunit of a protein phosphatase 2A complex and an interactor to Arabidopsis thaliana Regulator of G Signaling 1 protein (AtRGS1), a flg22-phosphorylated, 7-transmembrane spanning modulator of the nucleotide-binding state of the core G-protein complex. A null mutation of ATBα strongly increases basal endocytosis of AtRGS1. AtRGS1 steady-state protein level is lower in the atbα mutant in a proteasome-dependent manner. We propose that phosphorylation-dependent endocytosis of AtRGS1 is part of the mechanism to degrade AtRGS1, thus sustaining activation of the heterotrimeric G protein complex required for the regulation of system dynamics in innate immunity. The PP2A(ATBα) complex is a critical regulator of this signaling pathway.
Collapse
Affiliation(s)
- Justin M Watkins
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Christian Montes
- Department of Plant Pathology and Microbiology, Iowa State University, Ames, Iowa, USA
| | - Natalie M Clark
- Department of Plant Pathology and Microbiology, Iowa State University, Ames, Iowa, USA
| | - Gaoyuan Song
- Department of Plant Pathology and Microbiology, Iowa State University, Ames, Iowa, USA
| | - Celio Cabral Oliveira
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Biochemistry and Molecular Biology/BIOAGRO, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Bharat Mishra
- Department of Biology, University of Alabama-Birmingham, Birmingham, Alabama, USA
| | - Libuse Brachova
- Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa, USA
| | - Clara M Seifert
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Malek S Mitchell
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jing Yang
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Daisuke Urano
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - M Shahid Muktar
- Department of Biology, University of Alabama-Birmingham, Birmingham, Alabama, USA
| | - Justin W Walley
- Department of Plant Pathology and Microbiology, Iowa State University, Ames, Iowa, USA.
| | - Alan M Jones
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| |
Collapse
|
44
|
Shi K, Xiong Y, Wang Y, Deng Y, Wang W, Jing B, Gao X. PractiCPP: a deep learning approach tailored for extremely imbalanced datasets in cell-penetrating peptide prediction. Bioinformatics 2024; 40:btae058. [PMID: 38305405 PMCID: PMC11212486 DOI: 10.1093/bioinformatics/btae058] [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: 11/29/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/03/2024] Open
Abstract
MOTIVATION Effective drug delivery systems are paramount in enhancing pharmaceutical outcomes, particularly through the use of cell-penetrating peptides (CPPs). These peptides are gaining prominence due to their ability to penetrate eukaryotic cells efficiently without inflicting significant damage to the cellular membrane, thereby ensuring optimal drug delivery. However, the identification and characterization of CPPs remain a challenge due to the laborious and time-consuming nature of conventional methods, despite advances in proteomics. Current computational models, however, are predominantly tailored for balanced datasets, an approach that falls short in real-world applications characterized by a scarcity of known positive CPP instances. RESULTS To navigate this shortfall, we introduce PractiCPP, a novel deep-learning framework tailored for CPP prediction in highly imbalanced data scenarios. Uniquely designed with the integration of hard negative sampling and a sophisticated feature extraction and prediction module, PractiCPP facilitates an intricate understanding and learning from imbalanced data. Our extensive computational validations highlight PractiCPP's exceptional ability to outperform existing state-of-the-art methods, demonstrating remarkable accuracy, even in datasets with an extreme positive-to-negative ratio of 1:1000. Furthermore, through methodical embedding visualizations, we have established that models trained on balanced datasets are not conducive to practical, large-scale CPP identification, as they do not accurately reflect real-world complexities. In summary, PractiCPP potentially offers new perspectives in CPP prediction methodologies. Its design and validation, informed by real-world dataset constraints, suggest its utility as a valuable tool in supporting the acceleration of drug delivery advancements. AVAILABILITY AND IMPLEMENTATION The source code of PractiCPP is available on Figshare at https://doi.org/10.6084/m9.figshare.25053878.v1.
Collapse
Affiliation(s)
- Kexin Shi
- Syneron Technology, Guangzhou 510000, China
- Individualized Interdisciplinary Program (Data Science and Analytics), The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | | | - Yu Wang
- Syneron Technology, Guangzhou 510000, China
| | - Yifan Deng
- Syneron Technology, Guangzhou 510000, China
| | - Wenjia Wang
- Data Science and Analytics Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511400, Guangdong, China
| | - Bingyi Jing
- Department of Statistics and Data Science, Southern University of Science and Technology, Shenzhen 518000, China
| | - Xin Gao
- Syneron Technology, Guangzhou 510000, China
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
- Computational Bioscience Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| |
Collapse
|
45
|
Geng H, Qian R, Zhong Y, Tang X, Zhang X, Zhang L, Yang C, Li T, Dong Z, Wang C, Zhang Z, Zhu C. Leveraging synthetic lethality to uncover potential therapeutic target in gastric cancer. Cancer Gene Ther 2024; 31:334-348. [PMID: 38040871 DOI: 10.1038/s41417-023-00706-y] [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: 05/19/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 12/03/2023]
Abstract
Since trastuzumab was approved in 2012 for the first-line treatment of gastric cancer (GC), no significant advancement in GC targeted therapies has occurred. Synthetic lethality refers to the concept that simultaneous dysfunction of a pair of genes results in a lethal effect on cells, while the loss of an individual gene does not cause this effect. Through exploiting synthetic lethality, novel targeted therapies can be developed for the individualized treatment of GC. In this study, we proposed a computational strategy named Gastric cancer Specific Synthetic Lethality inference (GSSL) to identify synthetic lethal interactions in GC. GSSL analysis was used to infer probable synthetic lethality in GC using four accessible clinical datasets. In addition, prediction results were confirmed by experiments. GSSL analysis identified a total of 34 candidate synthetic lethal pairs, which included 33 unique targets. Among the synthetic lethal gene pairs, TP53-CHEK1 was selected for further experimental validation. Both computational and experimental results indicated that inhibiting CHEK1 could be a potential therapeutic strategy for GC patients with TP53 mutation. Meanwhile, in vitro experimental validation of two novel synthetic lethal pairs TP53-AURKB and ARID1A-EP300 further proved the universality and reliability of GSSL. Collectively, GSSL has been shown to be a reliable and feasible method for comprehensive analysis of inferring synthetic lethal interactions of GC, which may offer novel insight into the precision medicine and individualized treatment of GC.
Collapse
Affiliation(s)
- Haigang Geng
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruolan Qian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqing Zhong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangyu Tang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojun Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linmeng Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Li
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhongyi Dong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cun Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zizhen Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Chunchao Zhu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
46
|
Pereira CA, Reis-de-Oliveira G, Pierone BC, Martins-de-Souza D, Kaster MP. Depicting the molecular features of suicidal behavior: a review from an "omics" perspective. Psychiatry Res 2024; 332:115682. [PMID: 38198856 DOI: 10.1016/j.psychres.2023.115682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 12/05/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024]
Abstract
Background Suicide is one of the leading global causes of death. Behavior patterns from suicide ideation to completion are complex, involving multiple risk factors. Advances in technologies and large-scale bioinformatic tools are changing how we approach biomedical problems. The "omics" field may provide new knowledge about suicidal behavior to improve identification of relevant biological pathways associated with suicidal behavior. Methods We reviewed transcriptomic, proteomic, and metabolomic studies conducted in blood and post-mortem brains from individuals who experienced suicide or suicidal behavior. Omics data were combined using systems biology in silico, aiming at identifying major biological mechanisms and key molecules associated with suicide. Results Post-mortem samples of suicide completers indicate major dysregulations in pathways associated with glial cells (astrocytes and microglia), neurotransmission (GABAergic and glutamatergic systems), neuroplasticity and cell survivor, immune responses and energy homeostasis. In the periphery, studies found alterations in molecules involved in immune responses, polyamines, lipid transport, energy homeostasis, and amino and nucleic acid metabolism. Limitations We included only exploratory, non-hypothesis-driven studies; most studies only included one brain region and whole tissue analysis, and focused on suicide completers who were white males with almost none confounding factors. Conclusions We can highlight the importance of synaptic function, especially the balance between the inhibitory and excitatory synapses, and mechanisms associated with neuroplasticity, common pathways associated with psychiatric disorders. However, some of the pathways highlighted in this review, such as transcriptional factors associated with RNA splicing, formation of cortical connections, and gliogenesis, point to mechanisms that still need to be explored.
Collapse
Affiliation(s)
- Caibe Alves Pereira
- Laboratory of Translational Neurosciences, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Guilherme Reis-de-Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Bruna Caroline Pierone
- Laboratory of Translational Neurosciences, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil; Instituto Nacional de Biomarcadores Em Neuropsiquiatria (INBION) Conselho Nacional de Desenvolvimento Científico E Tecnológico, São Paulo, Brazil; Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, Brazil; D'Or Institute for Research and Education (IDOR), São Paulo, Brazil; INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D), São Paulo, Brazil.
| | - Manuella Pinto Kaster
- Laboratory of Translational Neurosciences, Department of Biochemistry, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil.
| |
Collapse
|
47
|
Lin Y, Du C, Ying H, Zhou Y, Kong F, Zhao H, Lan M. Multiply-mesoporous hydrophilic titanium dioxide nanohybrid for the highly-performed enrichment of N-glycopeptides from human serum. Anal Chim Acta 2024; 1287:342058. [PMID: 38182336 DOI: 10.1016/j.aca.2023.342058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 01/07/2024]
Abstract
N-glycopeptide is considered as one of significant biomarkers which provide guidance for the diagnosis and drug design of diseases. However, the direct analysis of N-glycopeptides is nearly impracticable mainly owing to their extremely low abundance and grave signal suppression from other interfering substances in the bio-samples. In this research, a multiply-mesoporous hydrophilic TiO2 nanohybrid (mM-TiO2@Cys) was synthesized by immobilizing Cys on a TiO2 substrate with hierarchical mesopores to achieve the highly-performed enrichment of N-glycopeptides. With the advantages of superior hydrophilicity and multiply-mesoporous structure, the obtained material exhibited an excellent selectivity (IgG digests and BSA digests at the molar ratio of 1/500), a high sensitivity (1 fmol μL-1 for IgG digests) and a good size-exclusion ability (IgG digests, IgG and BSA at the molar ratio of 1/500/500) in the enrichment of N-glycopeptides from IgG digests. As a result, 281 N-glycopeptides corresponded with 109 glycoproteins were identified from 2 μL serum digests of the patients with nasopharyngeal carcinoma, and 181 N-glycopeptides corresponded with 78 glycoproteins were identified from 2 μL serum digests of the healthy volunteers, revealing the potential application value of mM-TiO2@Cys in glycoproteomics.
Collapse
Affiliation(s)
- Yunfan Lin
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Chengrun Du
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Hongmei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
| | - Yifan Zhou
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Fangfang Kong
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Hongli Zhao
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
48
|
Mondello A, Dal Bo M, Toffoli G, Polano M. Machine learning in onco-pharmacogenomics: a path to precision medicine with many challenges. Front Pharmacol 2024; 14:1260276. [PMID: 38264526 PMCID: PMC10803549 DOI: 10.3389/fphar.2023.1260276] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
Over the past two decades, Next-Generation Sequencing (NGS) has revolutionized the approach to cancer research. Applications of NGS include the identification of tumor specific alterations that can influence tumor pathobiology and also impact diagnosis, prognosis and therapeutic options. Pharmacogenomics (PGx) studies the role of inheritance of individual genetic patterns in drug response and has taken advantage of NGS technology as it provides access to high-throughput data that can, however, be difficult to manage. Machine learning (ML) has recently been used in the life sciences to discover hidden patterns from complex NGS data and to solve various PGx problems. In this review, we provide a comprehensive overview of the NGS approaches that can be employed and the different PGx studies implicating the use of NGS data. We also provide an excursus of the ML algorithms that can exert a role as fundamental strategies in the PGx field to improve personalized medicine in cancer.
Collapse
Affiliation(s)
| | | | | | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
49
|
Danishuddin, Khan S, Kim JJ. From cancer big data to treatment: Artificial intelligence in cancer research. J Gene Med 2024; 26:e3629. [PMID: 37940369 DOI: 10.1002/jgm.3629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/12/2023] [Accepted: 10/18/2023] [Indexed: 11/10/2023] Open
Abstract
In recent years, developing the idea of "cancer big data" has emerged as a result of the significant expansion of various fields such as clinical research, genomics, proteomics and public health records. Advances in omics technologies are making a significant contribution to cancer big data in biomedicine and disease diagnosis. The increasingly availability of extensive cancer big data has set the stage for the development of multimodal artificial intelligence (AI) frameworks. These frameworks aim to analyze high-dimensional multi-omics data, extracting meaningful information that is challenging to obtain manually. Although interpretability and data quality remain critical challenges, these methods hold great promise for advancing our understanding of cancer biology and improving patient care and clinical outcomes. Here, we provide an overview of cancer big data and explore the applications of both traditional machine learning and deep learning approaches in cancer genomic and proteomic studies. We briefly discuss the challenges and potential of AI techniques in the integrated analysis of omics data, as well as the future direction of personalized treatment options in cancer.
Collapse
Affiliation(s)
- Danishuddin
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | - Shawez Khan
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Jong Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| |
Collapse
|
50
|
Haq SAU, Bashir T, Roberts TH, Husaini AM. Ameliorating the effects of multiple stresses on agronomic traits in crops: modern biotechnological and omics approaches. Mol Biol Rep 2023; 51:41. [PMID: 38158512 DOI: 10.1007/s11033-023-09042-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 10/13/2023] [Indexed: 01/03/2024]
Abstract
While global climate change poses a significant environmental threat to agriculture, the increasing population is another big challenge to food security. To address this, developing crop varieties with increased productivity and tolerance to biotic and abiotic stresses is crucial. Breeders must identify traits to ensure higher and consistent yields under inconsistent environmental challenges, possess resilience against emerging biotic and abiotic stresses and satisfy customer demands for safer and more nutritious meals. With the advent of omics-based technologies, molecular tools are now integrated with breeding to understand the molecular genetics of genotype-based traits and develop better climate-smart crops. The rapid development of omics technologies offers an opportunity to generate novel datasets for crop species. Identifying genes and pathways responsible for significant agronomic traits has been made possible by integrating omics data with genetic and phenotypic information. This paper discusses the importance and use of omics-based strategies, including genomics, transcriptomics, proteomics and phenomics, for agricultural and horticultural crop improvement, which aligns with developing better adaptability in these crop species to the changing climate conditions.
Collapse
Affiliation(s)
- Syed Anam Ul Haq
- Genome Engineering and Societal Biotechnology Lab, Division of Plant Biotechnology, SKUAST-K, Shalimar, Srinagar, Jammu and Kashmir, 190025, India
| | - Tanzeel Bashir
- Genome Engineering and Societal Biotechnology Lab, Division of Plant Biotechnology, SKUAST-K, Shalimar, Srinagar, Jammu and Kashmir, 190025, India
| | - Thomas H Roberts
- Plant Breeding Institute, School of Life and Environmental Sciences, Faculty of Science, Sydney Institute of Agriculture, The University of Sydney, Eveleigh, Australia
| | - Amjad M Husaini
- Genome Engineering and Societal Biotechnology Lab, Division of Plant Biotechnology, SKUAST-K, Shalimar, Srinagar, Jammu and Kashmir, 190025, India.
| |
Collapse
|